Background: Cellular therapies leveraging genetically engineered immune effector cells have witnessed a remarkable surge in success, particularly evident in the notable high rates of remission induction and durable remissions observed in a substantial proportion of heavily pretreated patients with refractory B-lineage malignancies. A diverse array of effector cells and therapeutic strategies are now at our disposal, representing the culmination of advancements made over the past 3 decades. The swift pace of development in modern genetic diagnostics, the emergence of spatial proteomics, and the expanding capabilities and precision of computational sciences have profoundly enriched our comprehension of tumor biology and the intricate workings of our immune system. Empowered by advancements in synthetic biology and genome editing, we can expedite the development of next-generation immune effector cells tailored for clinical applications, balancing safety with efficacy. Summary: Universal adapter chimeric antigen receptor (CAR) technologies present the most straightforward solution to tackle antigen heterogeneity and antigen evasion mechanisms employed by tumors. Moreover, due to the decoupling of antigen recognition and signaling in adapter CAR technologies, additional effector functions can safely enhance anticancer activity and most importantly, synergistic combination of patient-specific cellular products with off-the-shelf manufactured antibodies promise increased cost-efficiency. The pivotal collaboration between clinical trial units and regulatory institutions holds the key to surmounting contemporary challenges in trial design, potentially paving the way for the exploration of patient-individualized therapies. Key Messages: In this review, we elaborate on the concept of antibody-dependent cellular cytotoxicity mediated by universal adapter CARs and delineate how recent strides in CAR engineering have the potential to furnish a versatile cellular platform, ushering in an era of cancer-adapted, multitargeted immunotherapies employing universal CAR effector cells.

Redirecting immune effector cells to treat cancer has been exploited with antibodies, such as rituximab and trastuzumab since the 1990s (1997 and 1998 US-FDA approval, respectively) [1] and the milestone of the 100th monoclonal antibody product being FDA approved was reached in 2021. During the same time, the use of antibody variable chains in artificial immune receptors was introduced in the 1980s [2, 3] and optimized for 30 years, before the first CD19-chimeric antigen receptor (CAR) T-cell product tisagenlecleucel was FDA approved in 2017 after achieving sustained remissions in a series of pediatric patients with B-cell precursor acute lymphoblastic leukemia [4‒6]. Shortly thereafter, the FDA approved three more CD19-targeted CAR T-cell products, along with two CAR T-cell products targeted to BCMA [5].

The concept of enhancing the effector function by combining CAR-expressing effector cells in T and NK cells with antibodies and their fragments, thereby creating a potentiated artificial version of antibody-dependent cellular cytotoxicity (ADCC), has matured significantly over the past decade. This evolution and synthesis of ideas have been thoroughly reviewed by Arndt et al. [7] in their article entitled “Adaptor CAR Platforms – Next Generation of T Cell-Based Cancer Immunotherapy.”

Artificial immune receptors derived from the T-cell receptor (TcR) are depicted in Figure 1. The prevailing CAR architecture illustrated in Figure 1b is widely acknowledged as a sturdy foundation for the present phase of CAR-based cellular product development. Despite the diversity in available intracellular signaling domains, the fundamental components, such as the costimulatory signaling units sourced from CD28 and 4-1BB, along with the primary signaling driven by the CD3ζ chain [8] – have consistently demonstrated reliable performances across various cancer types in both preclinical models and clinical trials [6, 9‒13].

Fig. 1.

Artificial immune receptors based on TcR signaling. a Native TcRs are comprised of eight protein chains. Signal transduction is initiated by phosphorylation of ten available immunoreceptor tyrosine-based activation motifs (ITAMs). b Conventional CAR receptors are based on an extracellular recognition domain, e.g., single chain fragment variable (scFv), a hinge region, a transmembrane domain and intracellular costimulatory and signaling domains. They are comprised of two identical protein monomers that become fully functional after homodimerization and can signal via 6 ITAMS (CD3ζ chains). c T-cell receptor fusion constructs (TRuCs) are based on the integration of a fusion protein, e.g., scFv-CD3ε chain into the native TcR configuration. d The T-cell antigen coupler (TAC) is another concept how to mimic the TcR signaling by utilizing a CD4 or CD8α cell membrane anchored bispecific T-cell engager (BiTE) that closely connects the CD3ε chain with the target of interest via the second scFv of the anchored BiTE. e In the HLA-independent T-cell receptor (HIT) the TcRα and TcRβ variable fragments are substituted by the variable light and heavy fragment of antibodies via site-specific integration into the TRAC locus which in effect leads to the knockout of the native TcR. TRuC, TAC, and HIT are integrated into the TcR and therefore can theoretically signal from 10 ITAMs like the native TcR.

Fig. 1.

Artificial immune receptors based on TcR signaling. a Native TcRs are comprised of eight protein chains. Signal transduction is initiated by phosphorylation of ten available immunoreceptor tyrosine-based activation motifs (ITAMs). b Conventional CAR receptors are based on an extracellular recognition domain, e.g., single chain fragment variable (scFv), a hinge region, a transmembrane domain and intracellular costimulatory and signaling domains. They are comprised of two identical protein monomers that become fully functional after homodimerization and can signal via 6 ITAMS (CD3ζ chains). c T-cell receptor fusion constructs (TRuCs) are based on the integration of a fusion protein, e.g., scFv-CD3ε chain into the native TcR configuration. d The T-cell antigen coupler (TAC) is another concept how to mimic the TcR signaling by utilizing a CD4 or CD8α cell membrane anchored bispecific T-cell engager (BiTE) that closely connects the CD3ε chain with the target of interest via the second scFv of the anchored BiTE. e In the HLA-independent T-cell receptor (HIT) the TcRα and TcRβ variable fragments are substituted by the variable light and heavy fragment of antibodies via site-specific integration into the TRAC locus which in effect leads to the knockout of the native TcR. TRuC, TAC, and HIT are integrated into the TcR and therefore can theoretically signal from 10 ITAMs like the native TcR.

Close modal

Alternative signaling domains, such as DAP10 and DAP12, are interestingly recognized for their unique advantages over CD3ζ signaling due to their distinct downstream signaling with reduced exhaustion (PD-1), reduced secretion of inhibitory cytokines (IL10) and enhanced function of CAR+ tumor-infiltrating lymphocytes [14, 15]. To date, they are viewed as complementary to CD3ζ, rather than replacements for CD3ζ, or they can be used in combination with CD3ζ [15]. Genetic modifications in the CD3ζ chain leading to the inactivation of immunoreceptor tyrosine-based activation motifs (ITAMs) counterbalance T-cell exhaustion but may also reduce the antigen-density dependent activation threshold of these distinct CARs [16]. However, the high sensitivity of CARs to recognize antigen low expressing target cells (>200 molecules per cell sufficient to induce cytolysis) [17] is a prerequisite for profound cancer cell eradication and sustained remission [6, 18].

Efforts to incorporate the artificial immune receptor into the native TcR structure and harness the TcR-signaling machinery for CAR function have emerged and been revived [2] as an intriguing and alternative strategy compared to the conventional CAR architecture [19‒21]. Native TcRs (Fig. 1a) are comprised of eight protein chains assembled in a defined configuration. The TcRα and TcRβ chains provide the TcR with its specificity through their variable alpha- and variable beta fragment. Additionally, the TcR is comprised of two CD3ε chains, one CD3γ and CD3δ as well as two intracellular CD3ζ chains. Signal transduction by TcRs is initiated by the phosphorylation of potentially ten available ITAMs of which 6 ITAMs are incorporated in the two CD3ζ chains [22]. Conventional CARs (Fig. 2b) are based on an extracellular recognition domain, e.g., single chain fragment variable (scFv), a hinge region, a transmembrane domain and intracellular costimulatory and signaling domains [8]. CAR products approved by the US-FDA are second-generation CAR T-cell therapies featuring a CD28 or 4-1BB costimulatory domain coupled with CD3ζ chain signaling. They are comprised of two identical protein monomers that become fully functional after homodimerization [5]. They contain two CD3ζ chains and thus potentially signal via 6 ITAMs which is 4 ITAMs short of native TcRs or TcR-like immune receptors.

Fig. 2.

Conventional CAR and universal adapter CAR elements and function. a The immune synapse is highly conserved in the range of 15 nm (cell to cell distance) to operate efficaciously. In conventional CAR design, the positioning of the targeted epitope and the spacer define the distance between the immune cell and the target cell. In adapter CARs, the stereometry is extended by the adapter molecule. b Adapter molecules are comprised of an antigen binding domain, a structural domain, and a connecting domain. c CAR effector functions are activation threshold dependent and can be categorized in activation stages. I. Activation, II. cytolysis, III. cytokine and chemokine production, and IV. proliferation.

Fig. 2.

Conventional CAR and universal adapter CAR elements and function. a The immune synapse is highly conserved in the range of 15 nm (cell to cell distance) to operate efficaciously. In conventional CAR design, the positioning of the targeted epitope and the spacer define the distance between the immune cell and the target cell. In adapter CARs, the stereometry is extended by the adapter molecule. b Adapter molecules are comprised of an antigen binding domain, a structural domain, and a connecting domain. c CAR effector functions are activation threshold dependent and can be categorized in activation stages. I. Activation, II. cytolysis, III. cytokine and chemokine production, and IV. proliferation.

Close modal

One elegant technology utilizing the natural TcR-signaling machinery is referred to as TcR fusion constructs (TRuCs) (Fig. 1c), in which the extracellular recognition domain (scFv) is fused to the TcRα, TcRβ, CD3γ, CD3δ, or CD3ε chain and thus integrated into the native TcR configuration and therefore can theoretically signal from 10 ITAMs like the native TcR. Since the CD3ε chain is represented twice in the TcR, three different TcRs can be paired in CD3ε-TRuC positive T cells. The TcR in TRuC+ cells may remain native (without a CD3ε-scFv), contain one or two CD3ε-scFv for MHC-independent target-antigen recognition. The CD3ε-based TRuC emerged as the most promising candidate, yielding robust activity and response in different in vivo cancer models including B-lineage ALL and lymphoma, multiple myeloma, and glioblastoma multiforme. Surprisingly, the tumor control of the TRuCs was superior to conventional 2nd generation CAR T with incorporated CD28 or 4-1BB costimulatory domain [19]. Another approach integrates an anchored (CD4 or CD8α) bispecific T-cell engager (BiTE) to the cell membrane (Fig. 1d), referred to as T-cell antigen coupler (TAC) connecting the CD3ε chain with the targeted antigen can also signal with 10 ITAMs [21]. Both, the TRuC and the TAC share a similar CD3ε-dependent downstream activation pattern like blinatumomab [23].

To avoid mispairing and enhance correct pairing of the synthetic TcR-like receptor chains, murinized TcR-like CAR T constructs have been shown feasible in preclinical proof-of-concept studies [24]. By far the most advanced idea and technically challenging is to substitute the TcRα and TcRβ variable fragment with the variable light and heavy fragment of antibodies, realized in the HLA-independent TcR (HIT). This concept has been studied in different variations with the hope that TcR-like receptors may be more sensitive than conventional CAR designs since they can employ 10 ITAMs like a native TcR for the downstream signaling. The site-specific integration of the TcR-like CAR construct combines different aspects of T-cell physiology and concepts (Fig. 1e). Thus, the HIT is used to kill two birds with one stone. First, the TRAC integration inactivates the TcR alpha chain and thus reduces the likelihood of mispairing, and second it leads to a more physiological synthetic receptor regulation, comparable to the native TcR and results in a tightly regulated and activation-dependent cell surface expression of the HIT which has been proven to be beneficial for the CAR function [20, 25].

Over the last decade conventional CAR-based therapeutics have created the notion that immunotherapy with genetically modified cells can make the difference in advanced hematological malignancies [26], have the potential to restructure hematology, and sparked high hopes and expectations also in immune oncology. Contemporary CAR architectures have proven to reliably exert anticancer activity in various blood cancers. These homodimeric receptors are comprised of an antigen recognition domain, a hinge region, a transmembrane domain and a combination of intracellular signaling domains [8].

FDA-approved products consist of murine antibody-derived single chain variable fragments (scFv) [6] or camelid heavy chain variable (VHH) [10] based recognition domains. Alternative strategies to overcome immunogenicity, such as fully human VHH (12 kDa) [27], fully human scFv (25–30 kDa) or natural human ligand-based CARs [28], but also artificial small dimerization domains with a molecular weight below 10 kDa [29] have been utilized with success to equip CARs with targeting specificity.

The hinge region connects the recognition domain with the CAR-expressing cell in a certain distance to allow the formation of the cytolytic synapse. The ideal proximity between the CAR-expressing cell and the target cell typically falls within the distance range of approximately 15 nm, akin to the TcR-pMHC complex. However, the optimal spacer length, and thus the ideal distance is influenced and defined by the location, accessibility, and configuration of the epitope on the targeted antigen [30, 31]. Despite the abundance of available proteins, the selection of suitable spacers is somewhat constrained, as it primarily relies on proteins capable of homodimerization, such as CD28, CD8α, or the IgG-derived hinge and constant regions used in contemporary CAR architecture [8]. This limitation also applies to the most frequently utilized transmembrane domains, namely CD28 and CD8α. The costimulatory signaling domains are derived from the immunoglobulin-like coreceptor CD28 family or the tumor necrosis factor receptor superfamily. The individual costimulatory domains are distinct in activation kinetics, induction of cytotoxicity, secretion of cytokines, induction of proliferation [32] but also determine the metabolic signature [33]. The CD3ζ signaling domain stands out as the most commonly and effectively utilized signaling component in CAR receptors, often considered the gold standard [5, 6, 10]. The intentional fusion of functional protein units is diverse and enables tailored and finely tuned functionality to meet specific requirements. Nonetheless, while numerous variations of CAR signaling have proven to be effective in preclinical models, the conventional CAR design was solidified in clinical trials and serves as the cornerstone for any CAR product seeking to leverage clinical data.

Universal CAR technologies or adapter CAR technologies can deliberately redirect CAR-expressing cells to any accessible surface-expressed antigen of interest [7]. The mode of action and its terminology of “redirecting” is key to its function. The primary specificity of the adapter CAR should be designed to be immunologically irrelevant because the targeted epitope is supposed to not be present in the human body under physiological circumstances. Only the administration of adapter molecules introduces the targeted moiety into the system (human body) since the highly defined structure recognized by the adapter CAR recognition domain is only carried by adapter molecules. Therefore, the adapter molecule redirects the adapter CAR highly specifically to the selected surface-expressed target of interest. Conclusively, the targeted epitope of adapter CAR recognition domains is exclusively accessible on the adapter molecules. Otherwise, the CAR may interact with its naturally present epitope and could cause significant toxicities. Therefore, the moiety (connecting domain) targeted by adapter CARs must be nonhuman or nonexistent and non-cross-reactive with structures present in the human body [34, 35], or inaccessible for the CAR on the cell surface of cells [36], and exclusively conjugated to or integrated into the adapter molecules [7].

At the same time, one major limiting factor of adapter CAR function is the technology’s dependency on the availability of the adapter molecule everywhere in the patient’s body. Therefore, the adapter molecule concentration needs to reach CAR-activating concentrations at the tumor site, and the primary antigen density on the target cells must be above the CAR-activating expression threshold [32] and optimally the targeted antigen should not be subject to shedding [37], rapid antigen turnover, and/or internalization of bound adapter molecules [38]. Strategic adapter molecule selection of the molecule format may contribute to realize optimized targeting. Adapter molecules need to possess the capability to permeate all pertinent body compartments and peripheral areas, including low-perfused tissues such as bone or tumor niches and metastatic sites [39]. The penetration of antibodies into the central nervous system for instance is limited, which makes it harder but not impossible to reach sufficient adapter molecule concentrations in all body compartments. Especially, the blood-brain barrier can be overcome by adapter molecule design with CNS penetration capabilities [40] or the direct administration of the adapter molecules into the cerebrospinal fluid, e.g., intrathecal injection or the application through artificial CNS access, such as the Ommaya reservoir an intraventricular catheter system that can be used for the aspiration of cerebrospinal fluid or drug administration for diagnostic and therapeutic procedures [41, 42].

Under ideal conditions, the target cells are recognized by surface-bound adapter molecules on the target cells and facilitate the formation of the cytolytic synapse via the engagement with the adapter CAR-expressing effector cells. In vivo, the pharmacokinetics and -dynamics of both the CAR-expressing cells and the adapter molecules determine the adapter CAR recruitment, engagement, and effector functions. The underlying mechanism of action and the induced basic effector functions of adapter CARs are illustrated in Figure 2.

In conventional CARs (Fig. 2a), the antigen recognition is dependent only on the CAR-expressing cell and the target cells that express the targeted antigen. They are targeted to one single antigen and cannot recognize antigen-negative targets which is one of the major reasons of CAR treatment failure – antigen or epitope loss and antigen downregulation [6]. To address antigen heterogeneity and antigen loss, bispecific (CD19/CD22 or CD19/CD20) [43, 44] and trispecific (CD19/CD20/CD22) [45] CAR T cells have been created to target B-lineage-specific antigens.

In adapter CARs, the adapter molecule interconnects the CAR-expressing cell and the target cells. Thus, the physiology of adapter CARs, in terms of pharmacokinetics and -dynamics, is more complex than that of conventional CAR technologies, sharing similarities with antibody-mediated immunity. The features of conventional CAR and universal CAR technologies are summarized in Table 1.

Table 1.

Features of conventional CAR- and universal CAR technologies

Conventional CARUniversal CAR
Antigen density threshold for activation +++++ ++++ 
Antigen specificity High High 
Switch-on mechanism Unsupported Yes 
Switch-off mechanism Unsupported Passive inactivation, PK dependent 
On-target-off-tumor toxicity Uncontrolled Regulatable 
Combinatorial targeting Dual/triple targeting Yes 
Sequential targeting Unsupported Yes 
Transient targeting Unsupported Yes 
Treatment in cycles Unsupported Yes 
Selective targeting Unsupported Partially supported 
Distribution of toxicity Unsupported Yes 
Bystander effect Limited Limited 
Conventional CARUniversal CAR
Antigen density threshold for activation +++++ ++++ 
Antigen specificity High High 
Switch-on mechanism Unsupported Yes 
Switch-off mechanism Unsupported Passive inactivation, PK dependent 
On-target-off-tumor toxicity Uncontrolled Regulatable 
Combinatorial targeting Dual/triple targeting Yes 
Sequential targeting Unsupported Yes 
Transient targeting Unsupported Yes 
Treatment in cycles Unsupported Yes 
Selective targeting Unsupported Partially supported 
Distribution of toxicity Unsupported Yes 
Bystander effect Limited Limited 

Conventional CAR design appears to be more responsive. Universal CAR technologies facilitate safety and versatile combinatorial targeting via the on-switch mechanism.

In contrast to direct CARs, the antigen recognition and the CAR signaling are decoupled from each other in adapter CARs (Fig. 2a), and the CAR-positive immune cells are inactive in the absence of adapter molecules [7]. The adapter molecule is comprised of an antigen-binding domain, a structural domain, and a connecting domain. The most relevant criterion for antibody therapies, bispecific antibody therapy, and CAR therapy is the stereometry of the receptor target interaction to initiate an immune response by interconnecting immune effector cells with target cells. Mechanistically, this entails drawing the cells closer together to enhance the interaction between the immune cells and the target cells, thereby facilitating activation and the formation of the immune synapse. The distance of the immune synapse (Fig. 2a) with close proximity between the immune cell membrane and the target cell membrane is highly conserved in the range of 15 nm (˜TcR-pMHC interaction distance) and a prerequisite to activate immune cells efficaciously and mediate effects via ADCC [46], BiTE, and CARs [47, 48], whereas antibody-dependent cellular phagocytosis (ADCP) favors slightly greater distances [46]. In conventional CAR design, the operational distance is tailored through 1st the careful selection of the targeted epitope on the antigen [49] and 2nd the size of the spacer defining the length of the extracellular CAR domain (distance) [50]. In adapter CARs, the distance between the CAR-expressing cell and the target cell is extended by the adapter molecule [34, 35]. The additional distance of the adapter CAR cell membrane to the target cell membrane corresponds to the distance between the connecting domain to the antigen binding domain (Fig. 2b). If the connecting domain is directly linked to the N-terminus of the adapter molecule, the extension of the distance can be minimal in the subnanometer range but can also exceed 7 nm if placed at the C-terminal end of a Fab fragment or even 10 nm if placed at the C-terminal end of the CH3 domain of an antibody [51].

Adapter CARs mediate the same effector functions as conventional CAR technologies but require the correct assembly and configuration of the CAR receptor, the adapter molecule, and the targeted antigen. The effector functions are activation threshold dependent and can be categorized in activation stages and as shown in Figure 2c. Minimal activation is required to modify the receptor expression profile and to induce cytolytic activity. In direct comparison, the cytolytic activity of CARs is induced at 10-fold lower antigen density than cytokine secretion [17]. Thus, cytokine and chemokine secretion require a significantly enhanced CAR engagement than cytolysis. Cytokines provide relevant signals to facilitate a robust and sustained immune response, the recruitment of accessory cells and CAR proliferation. As such, they have been incorporated into modern CAR design and are used to enhance both cytotoxicity and the proliferative capacity [52].

Since the specificity of targeting is defined by the adapter molecule, versatile polyimmunotargeting is the greatest feature of adapter CAR technologies to overcome antigen-based immune evasion [34, 53]. Hence, adapter CARs offer the distinct advantage of being activatable through the introduction of adapter molecules with differential specificity [35]. This capability enables transient targeting, synchronized combinatorial targeting, and sequential combinatorial targeting, allowing the primary targeting mechanism to adjust in accordance with the pharmacokinetics and -dynamics of the adapter molecule, increase the overall potency of targeting and spare toxicities [34, 35, 54‒56].

In universal CAR technologies, adapter molecules enable CAR transactivation in the sense of next-generation ADCC [34, 53] and ADCP. The specified terminology for adapter CARs as of adapter molecule-dependent cellular cytotoxicity (AMDCC) and adapter molecule-dependent cellular phagocytosis (AMDCP) cover both, antibody fragments, and alternative adapter molecule formats beyond wildtype IgG [35].

Conventional CARs are more sensitive than antibody-based therapies and universal adapter CAR technologies (Fig. 3a). Most clinical experience has been gained with second generation-based CAR T-cell products in B-lineage cancer targeted to CD19 [4, 6] and BCMA [57] that have illuminated the potential of CAR-based therapies. They have evidenced that artificially enhanced signaling utilizing CARs can outperform any other available therapeutic today including combination therapies with antibodies as well as bispecific antibodies [18, 58]. Adapter CARs are more sensitive and less toxic than BiTEs (Fig. 4a, b) and are more potent than antibody therapies in preclinical models (unpublished data). This is attributed to the target-independent activation of T cells by blinatumomab. Universal CARs require stronger forces to induce target-independent activation.

Fig. 3.

Features of universal CAR technologies and the comparison of AMDCC with ADCC and ADCP. a Conventional CARs are more sensitive and potent than antibody-based therapies and universal adapter CAR technologies. Nonetheless, adapter CARs are more sensitive and less toxic than bispecific T-cell engagers (BiTE) and are more potent than antibody therapies (unpublished data). b Adapter molecules are generated like antibodies and follow the same optimization steps, such as humanization and affinity maturation. c Fc-engineered antibodies mediate enhanced ADCC, ADCP, and CDC. In adapter CARs either Fab or immune-silenced antibodies are used to circumvent activation-induced cell death. d The placement of the targeted moiety on the adapter molecules is empirically optimized by functional AMDCC selection to identify the best-performing stereometry. e The main cell source for CAR therapies have been T cells due to their proliferative capacity, great functional diversity (TC1, TC22, TH1, and TH2 and others) and long-term persistence based due to self-renewal and memory function. Universal CAR technologies equip T cells with NK-like functions to mediate ADCC. NKT, NK, and macrophages feature unique properties and deserve further exploration as CAR-expressing cells.

Fig. 3.

Features of universal CAR technologies and the comparison of AMDCC with ADCC and ADCP. a Conventional CARs are more sensitive and potent than antibody-based therapies and universal adapter CAR technologies. Nonetheless, adapter CARs are more sensitive and less toxic than bispecific T-cell engagers (BiTE) and are more potent than antibody therapies (unpublished data). b Adapter molecules are generated like antibodies and follow the same optimization steps, such as humanization and affinity maturation. c Fc-engineered antibodies mediate enhanced ADCC, ADCP, and CDC. In adapter CARs either Fab or immune-silenced antibodies are used to circumvent activation-induced cell death. d The placement of the targeted moiety on the adapter molecules is empirically optimized by functional AMDCC selection to identify the best-performing stereometry. e The main cell source for CAR therapies have been T cells due to their proliferative capacity, great functional diversity (TC1, TC22, TH1, and TH2 and others) and long-term persistence based due to self-renewal and memory function. Universal CAR technologies equip T cells with NK-like functions to mediate ADCC. NKT, NK, and macrophages feature unique properties and deserve further exploration as CAR-expressing cells.

Close modal
Fig. 4.

Pharmacodynamic differences of bispecific T-cell engager and universal CAR therapy. Both technologies are sensitive and are operational at doses significantly lower (>1,000-fold) than in conventional antibody therapy. a BiTEs have a narrow therapeutic margin because they induce target-independent activation of T cells, leading to life-threatening hyperinflammatory complications. b Universal CAR technologies display a much broader therapeutic spectrum. The tendency of target-independent activation is much lower compared to BiTEs. Supraoptimal dosing leads to reduced activity and toxicity of universal CAR due to competitive inhibition rather than hyperinflammatory conditions.

Fig. 4.

Pharmacodynamic differences of bispecific T-cell engager and universal CAR therapy. Both technologies are sensitive and are operational at doses significantly lower (>1,000-fold) than in conventional antibody therapy. a BiTEs have a narrow therapeutic margin because they induce target-independent activation of T cells, leading to life-threatening hyperinflammatory complications. b Universal CAR technologies display a much broader therapeutic spectrum. The tendency of target-independent activation is much lower compared to BiTEs. Supraoptimal dosing leads to reduced activity and toxicity of universal CAR due to competitive inhibition rather than hyperinflammatory conditions.

Close modal

Blinatumomab is clinically used strictly at serum concentrations of 0.5–0.7 ng/mL [59] due to life-threatening hyperinflammatory complications at higher dosing. In contrast, clinical data concerning the use of adapter molecules at similar low concentrations, ranging from nanograms to micrograms per milliliter, is scarce [60, 61]. The broad therapeutic spectrum of adapter molecule dosing in universal CAR therapy will support reaching clinically relevant concentrations of adapter molecules in various pharmacokinetic compartments [60, 61] as well as sequential or simultaneous combinatorial targeting which is compromised in BiTE therapy due to dose-limiting toxicities [62].

The generation and optimization of adapter molecules are comparable to antibody development including immunization strategies predominantly in mice (Fig. 3b). However, adapter CAR technologies can also build on clinically proven and US-FDA/EMA-approved antibodies [34, 35]. Immunization with proteins, protein fragments, Fc-fusion proteins, peptides, DNA, or mRNA is used to generate new antibodies [63]. The target and epitope selection may vary between conventional antibody therapy and adapter CARs due to the unique requirements for each distinct application [49, 64].

Contemporary antibody optimization includes the humanization and affinity maturation of the antibody of interest to fit their purpose. In most instances, the goal is to increase the antibody affinity during this process [63] and optionally can be grafted into an Fc-modified antibody framework (Fig. 3c), to enhance ADCC, ADCP, and CDC [65, 66]. Contrarily, in adapter CARs either antibody fragments or strategically silenced antibodies [67] are used to circumvent activation-induced cell death which is mediated by the interaction of CAR+ cells with FcR-expressing immune cells [68] leading to unwanted in vivo depletion of CARs.

Natural ADCC and Universal Adapter CAR Mediated AMDCC

Normally, NK cells are the major immune effector cell population to exert ADCC, and M1 macrophages to mediate ADCP predominantly via IgG1 antibodies [58, 69]. The substantial contribution of ADCP in achieving tumor control with antibody therapies, such as rituximab, cetuximab, and trastuzumab underscores the importance in future efforts to exploit macrophages or induced pluripotent stem cell (iPSC)-derived macrophage-like cells for CAR-based immunotherapies [69‒71]. Although native IgG1 antibodies may exhibit limited recruitment and anticancer function with NK cells due to their generally low affinity to FcγRIIIA in the micromolar range [72], often leading to insufficient downstream signaling and cytotoxicity [58], they robustly mediate ADCP [69]. Advanced antibody technologies enhance the affinity of wildtype IgG1 to FcRγIIIA of KD 252 nm by greater two log-fold in IgG1S239d/I332E to KD 2 nm through the substitution of amino acids in the CH2 domain [66] or similarly by glycoengineering in the means of defocusylation [73‒76]. These modifications enhance the affinity of the modified IgG1 for FcRγIIIA, thereby boosting their anticancer efficacy [77]. Moreover, such changes can transform antibodies into highly potent biological agents with significant clinical effects [58]. Two prominent examples of antibody engineering are represented by the clinical success of the humanized IgG1S239d/I332E CD19 antibody tafasitamab in the treatment of patients with diffuse large B-cell lymphoma [78] as well as the humanized glycoengineered CD20 antibody obinutuzumab [75] for the administration in patients with chronic lymphocytic leukemia and small cell lymphocytic leukemia [79], despite the need to identify the best functional affinity of therapeutic antibodies which depends on their mode of action, e.g., immune activating antibodies [80] versus antigen-targeted antibodies [81]. Through the evolution of antibody engineering, it has become apparent that both affinities, 1st the KD of the antibody to the targeted antigen [82] and 2nd the KD of the immune receptor, e.g., FcRγIIIA to the antibody significantly impact on the overall potency of therapeutic antibody [66, 74]. Antibodies must reach a minimum affinity threshold to exert immune functions and in most instances high affinity antibodies are superior to antibodies with lower affinity, but there certainly is a controversy around the best affinity, e.g., claiming that moderate antibody affinities might increase the penetration of antibodies into the tumor in solid cancer [83]. Absolute affinities for therapeutic antibodies seem to be less important than affinity thresholds [81] if the affinities lie in the low nanomolar or sub nanomolar range, like the EGFR-targeted antibodies panitumumab (IgG2a, KD 0.05) and cetuximab (IgG1, KD 0.39) [84]. In general, the best affinity for therapeutic antibodies is to be identified empirically by their biological activity which is also impacted by various factors, such as the binding epitope, the internalization kinetics, the IgG subtype as well as functional modifications [63]. For the use in adapter CARs, the IgG subclass seems irrelevant since the recruitment of effector cells is independent of the primary antibody subclass and function [34, 53]. Therefore, the development of adapter molecules can leverage clinically proven and effective antibodies, such as the CD33-targeted IgG4 antibody gemtuzumab, which features a core-hinge mutation [85]. Additionally, the conversion of the IgG isotype to IgG1 remains an option during the empirical optimization process.

Besides the expression level, accessibility and configuration of the targeted antigen, the proximity of the binding epitope to the target cell surface [86], the stereometry including the presentation of the CAR-targeted moiety (connecting domain), and the antibody affinity are the most relevant functionally determining factors [35]. The optimization of the stereometry (Fig. 3d) by placement (integration or site-specific conjugation) of the connecting domain on adapter molecules is a meticulous and challenging process, yet it is crucial for determining the best system’s potency. Antibodies that are used to mediate natural ADCC, ADCP, and CDC do not allow to optimize the accessibility, configuration, or distance of the highly conserved binding structure for Fc-receptors and complement factors. This is a significant advantage of AMDCC and AMDCP over ADCC and ADCP.

NK, Invariant NK-T Cells, and Macrophage CAR Effector Cells

Furthermore, adapter CAR technologies have the potential to enhance the functionality of ADCC and ADCP across different populations of immune effector cells (Fig. 3e). Various CAR-expressing effector cells can theoretically be recruited by adapter molecules of any format with a functionally tailored downstream signaling. This immune modulatory function may be proinflammatory activating, or altered to mediate target-antigen dependent immunosuppression [87]. Deductively, introducing a CAR not only enhances the function of T cells [4, 6] but also extends its benefits to a broad spectrum of other immune cells, including NK cells [88], invariant NK-T cells (iNKT) [89], and macrophages [70]. The ADCC and ADCP conversion into AMDCC and AMDCP facilitates the technology to build on versatile adapter CAR concepts that have been fully optimized for this bespoke purpose including the integration of application- and cell type-specific signaling and allow the flexible optimization of the adapter molecules far beyond the possible modifications and optimizations of basic antibody functions, such as ADCC, ADCP, and CDC [65, 66, 74, 90].

Even though T cells have been the main cell population for CAR therapies, other immune subsets like NK cells, iNKT or macrophages display unique properties which are highly valuable for immunotherapy [70, 88, 89]. The major reason why T cells have been most efficacious in the context of CAR therapies is 1st their exponential proliferation capacity [4, 6], 2nd T cells comprise an autonomously managed immune compartment with various T cell subsets and functions, e.g., TC1, TC22, TH1, and TH2 [91, 92], and 3rd CAR T cells can be transferred into patients and retain their high capacity for self-renewal and can persist long-term [4, 6, 10], especially through naïve T cells, stem-cell memory, and central memory T cell subsets that are able to engraft permanently and become part of the patient’s integratively regulated immune system [6]. The value of alternative immune cell populations has been recognized and are increasingly being harnessed to enhance anticancer immune responses with NK cells [93], iNKT cells [94] as well as specific subsets of macrophages M1, and neutrophils N1, but on the other hand excluding immunosuppressive, cancer-promoting M2 macrophages and N2 neutrophils [95, 96].

The signaling in CAR-expressing immune cells is expanded and enhanced compared to native immune cells. To further enhance the potency of adapter CARs, additional effector functions can be incorporated into the basic CAR construct and are constitutively [89] or inducibly expressed in an antigen-dependent manner, e.g., under NFAT, AP-1, or NFKB and others [97, 98] referred to as T cells redirected for antigen-unrestricted cytokine-initiated killing or 4th generation CAR constructs [5, 97]. The transgene expression of additional effector functions in T cells redirected for antigen-unrestricted cytokine-initiated killings is tightly regulated and antigen-dependent via the CAR activation [99] and due to the decoupled signaling in adapter CARs, it is directly related to the presence or absence of the adapter molecule in a dose-dependent manner [34].

To overcome immunosuppressive signals in the tumor microenvironment [100], proinflammatory cytokines have proven to potentiate the CAR performance. Among these are the common γ chain cytokines IL7 [101], IL15 [102], and IL21 [103], as well as IL12 [104] and IL18 [105] triggering an increase of cytotoxicity in immune cells and interferon-γ production [106, 107]. IL12 represents one of the most potent but also toxic cytokines and thus requires reliable safety measures [108].

Other strategies overcome inhibitory signaling with functionally inert ligand or cytokine receptors also referred to as dominant-negative immune receptors. This strategy has increased the potency of CARs through competitive ligand/cytokine binding for PD-1 and TGF-β also in a clinical setting with acceptable toxicity profiles [109, 110] but also PD-1 and CTLA-4 gene disruption has been used to overcome immune checkpoint inhibition [111, 112]. Furthermore, immune checkpoint inhibition of PD-1 and switch receptor signaling via PD-1 to CD28 and TGF-β to IL7 signaling conversion has enhanced the potency and persistence of CAR T cells [113, 114].

According to the safety concerns, the pharmacokinetic features of the adapter molecule can be adjusted by selecting the most suitable adapter molecule format with the intended half-life, e.g., Fab fragment for rapid clearance within hours to days [115], Fc-fusion for medium-term half-life within days and full-size antibody within several days up to weeks [116]. It is worth mentioning that the on-switch mechanism and passive elimination is also dependent on the application route and obviously on the molecular weight of the adapter molecule [35].

Monotargeted CAR therapy is insufficient to control most cancers and antigen loss is a major cause of relapse in CD19- and BCMA-targeted CAR therapy [6, 10]. Besides multitargeting [44, 45], additional effector functions are necessary to overcome roadblocks, such as the hazardous immunosuppressive milieu in and around the cancer, leading to reluctant chemotaxis and CAR trafficking to the tumor site and lack of penetration into the tumor [117]. Moreover, tumors impair antigen-dependent proliferation, induce hypoxia and metabolic deficiency, immune cell exhaustion, and apoptosis [100] not only in solid cancers but also impact on the performance in CD19CAR T therapy [118].

Consequently, the success in CAR therapy relies on multitargeting and the integration of additional effector functions. IL2, IL7, and IL15 are key homeostatic T-cell cytokines [119]. The co-expression of IL15 and the inducible expression of IL7 under NFAT responsive elements have been shown to be safe in clinical trials (ClinicalTrials.gov identifier: NCT03056339, NCT03258047) and to improve the engraftment, proliferation, anticancer function, and persistence of CARs [88, 120]. Due to the existing clinical experience on safety, IL15 and IL7 co-expression should be prioritized in adapter CAR clinical trial efforts. The same accounts for 3rd generation adapter CAR architecture CD28-BB-CD3ζ which was superior to 2nd generation CARs with CD28-CD3ζ and BB-CD3ζ signaling in anti-LLE CAR T (Fig. 5a) and also demonstrated convincing preclinical activity in anti-FITC CARs in various cancer models [34, 121].

Fig. 5.

Universal CAR today and the future perspective. a Advanced CAR T design to boost the CAR performance of adapter CARs can be safely implemented in universal CAR technologies, for instance the inclusion of constitutive IL15 expression. The combination of adapter molecules for synchronic combinatorial targeting in clinical studies will allow to trial the most prominent feature of adapter CARs – multitargeting. b Lineage-specific toxicities can limit the toxicity profile significantly, e.g., the targeting of the B-lineage associated antigens CD19, CD20, and CD22 will increase the potency of targeting while retaining the toxicity profile to the B-lineage compartment. c The future idea of universal CAR therapy is built on cost-efficient manufacturing of allogeneic iPSC-derived CAR products with a multitude of different cell types and effector functions cryopreserved and at our disposal for treatment without delay. d The future scenario may offer patients off-the-shelf manufactured immune cells with tailored immune functions as well as patient-individualized combinations of adapter molecules (off-the-shelf products).

Fig. 5.

Universal CAR today and the future perspective. a Advanced CAR T design to boost the CAR performance of adapter CARs can be safely implemented in universal CAR technologies, for instance the inclusion of constitutive IL15 expression. The combination of adapter molecules for synchronic combinatorial targeting in clinical studies will allow to trial the most prominent feature of adapter CARs – multitargeting. b Lineage-specific toxicities can limit the toxicity profile significantly, e.g., the targeting of the B-lineage associated antigens CD19, CD20, and CD22 will increase the potency of targeting while retaining the toxicity profile to the B-lineage compartment. c The future idea of universal CAR therapy is built on cost-efficient manufacturing of allogeneic iPSC-derived CAR products with a multitude of different cell types and effector functions cryopreserved and at our disposal for treatment without delay. d The future scenario may offer patients off-the-shelf manufactured immune cells with tailored immune functions as well as patient-individualized combinations of adapter molecules (off-the-shelf products).

Close modal

New clinical trials with adapter CARs are recommended to enter the clinical stage with a multitargeted approach utilizing well-characterized target antigens, such as CD19, CD20, and CD22 to prove their capabilities for combinatorial targeting. This would represent a risk-averse strategy because adapter CARs are theoretically safer than conventional CARs. The objective would built on the facts that adapter CARs are currently being trialed in patients with relapsed/refractory CD19 positive malignancies (NCT04450069) and conventional CD19- [6], CD20- [122], and CD22-targeted [43] CAR T cells are used in the clinic as well as multitargeted CD19/CD20- and CD19/CD22-targeted CAR T products are used in clinical trials [44, 123]. The concept of “lineage-specific toxicities” in CAR T-cell therapy (Fig. 5b) reduces the risk for unexpected adverse events that can significantly compromise, slow down, or discontinue clinical trials [124]. The antigens CD19, CD20, and CD22 are “clean” B-lineage expressed proteins and have been studied carefully with various therapeutics including antibodies [78], BiTEs [125], antibody drug conjugates [126], and CAR T cells [6, 43, 122]. Once multitargeted adapter CAR therapy has been shown to be safe, the combination with immune checkpoint blockade may additionally improve the clinical outcome like it has in selected patients treated with conventional CD19CAR T [127]. The intermediate step in universal CAR therapy will be the use of patient-individualized adapter molecule combinations to increase the potency and spare unnecessary toxicities.

The future aspiration of transgenic immune cell therapy has no limits. Novel immune receptors will be developed that contain tailored immune functions, with complex bespoke signaling and logic gating, cytokine, and chemokine delivery. The immunomodulatory function of future transgenic immune cells will go beyond oncology and the plain depletion of B cells like it has rescued patients with systemic lupus erythematosus [128].

The exploitation transgenic immune cells in different naturally occurring cell populations and distinct subsets of T cells [6], NK cells [88], iNKT [89], and macrophages [70] will expand the arsenal and therapeutic options. Most likely this will transition into the phase of generating a versatile library of universal allogeneic iPSC-derived effector cells for various indications and applications [129]. There is a chance these artificial immune cells will grow in large tanks, will be cryopreserved (Fig. 5c) and at our disposal at any time in conjunction with off-the-shelf adapter molecule combinations (Fig. 5d). These artificial iPSC-derived effectors will have little in common with their natural counterparts but rather serve as their role model to create the most efficacious next-generation therapeutics. It is a question of scientific art and creativity to combine all possible modifications per individual cancer indication but technically it will be possible to generate these cells. Modifications will be comprised of TcR-like receptors [20], cytokines (e.g., IL12, IL18) [97, 105, 130], and chemokines (e.g., CCL19) [117] as well as their corresponding receptors [131], switch receptors [113], calibrated signaling [16], reverse signaling receptors, logic gating (e.g., SynNotch) [132], site-specific integration of transgenes [25], serial gene disruption of immune checkpoint receptors, and gene regulators [133].

Indirect CAR technologies may become the future of CAR therapies. These include universal adapter CARs [35, 53] and remote switch-on CARs [134‒136]. Indirect CARs are complementary to CD19 and BCMA CARs; however beyond B-lineage cancers, universal adapter CARs are the straightforward solution to the complex challenge of antigen heterogeneity in CAR therapy and the antigen question after all. There is a bright outlook for adapter CAR technologies today and in the future and thus the challenges of adapter molecule development and most importantly to create new flexible concepts for clinical trial design in close collaboration with regulatory institutions need to be taken on. In the era of patient-individualized therapies, this will be the most important achievement. Otherwise, time is against individualized immunotherapy and the dream of versatile futuristic therapies is unlikely to become reality.

During the preparation of this work the authors used ChatGPT 3.5 (OpenAI) to find suitable substitute words (thesaurus function) and overcome non-native language barriers. After using this tool, the authors reviewed and edited the content as needed and take full responsibility for the content of the publication.

P.S. is an inventor of patents in cancer immunology in the areas of antibody and CAR therapy. L.S. has nothing to disclose. The authors have no ethical conflicts to disclose.

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

P.S. conceptualized the manuscript. L.S. and P.S. wrote the manuscript. Both authors reviewed and approved the final version of the manuscript prior to submission.

1.
Lyu
X
,
Zhao
Q
,
Hui
J
,
Wang
T
,
Lin
M
,
Wang
K
, et al
.
The global landscape of approved antibody therapies
.
Antib Ther
.
2022
;
5
(
4
):
233
57
.
2.
Eshhar
Z
,
Waks
T
,
Gross
G
.
The emergence of T-bodies/CAR T cells
.
Cancer J
.
2014
;
20
(
2
):
123
6
.
3.
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 USA
.
1989
;
86
(
24
):
10024
8
.
4.
Grupp
SA
,
Kalos
M
,
Barrett
D
,
Aplenc
R
,
Porter
DL
,
Rheingold
SR
, et al
.
Chimeric antigen receptor-modified T cells for acute lymphoid leukemia
.
N Engl J Med
.
2013
;
368
(
16
):
1509
18
.
5.
Boettcher
M
,
Joechner
A
,
Li
Z
,
Yang
SF
,
Schlegel
P
.
Development of CAR T cell therapy in children-A comprehensive overview
.
J Clin Med
.
2022
;
11
(
8
):
2158
.
6.
Maude
SL
,
Laetsch
TW
,
Buechner
J
,
Rives
S
,
Boyer
M
,
Bittencourt
H
, et al
.
Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia
.
N Engl J Med
.
2018
;
378
(
5
):
439
48
.
7.
Arndt
C
,
Fasslrinner
F
,
Loureiro
LR
,
Koristka
S
,
Feldmann
A
,
Bachmann
M
.
Adaptor CAR platforms-next generation of T cell-based cancer immunotherapy
.
Cancers
.
2020
;
12
(
5
):
1302
.
8.
Jayaraman
J
,
Mellody
MP
,
Hou
AJ
,
Desai
RP
,
Fung
AW
,
Pham
AHT
, et al
.
CAR-T design: elements and their synergistic function
.
EBioMedicine
.
2020
;
58
:
102931
.
9.
Rodriguez-Otero
P
,
Ailawadhi
S
,
Arnulf
B
,
Patel
K
,
Cavo
M
,
Nooka
AK
, et al
.
Ide-cel or standard regimens in relapsed and refractory multiple myeloma
.
N Engl J Med
.
2023
;
388
(
11
):
1002
14
.
10.
Berdeja
JG
,
Madduri
D
,
Usmani
SZ
,
Jakubowiak
A
,
Agha
M
,
Cohen
AD
, et al
.
Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study
.
Lancet
.
2021
;
398
(
10297
):
314
24
.
11.
Neelapu
SS
,
Locke
FL
,
Bartlett
NL
,
Lekakis
LJ
,
Miklos
DB
,
Jacobson
CA
, et al
.
Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma
.
N Engl J Med
.
2017
;
377
(
26
):
2531
44
.
12.
Ye
H
,
Huang
RR
,
Shuch
BM
,
Chen
Z
,
Said
JW
,
Pantuck
AJ
, et al
.
CD70 is a promising CAR-T cell target in patients with advanced renal cell carcinoma
.
J Clin Oncol
.
2022
;
40
(
6_Suppl l
):
384
.
13.
Flieswasser
T
,
Van den Eynde
A
,
Van Audenaerde
J
,
De Waele
J
,
Lardon
F
,
Riether
C
, et al
.
The CD70-CD27 axis in oncology: the new kids on the block
.
J Exp Clin Cancer Res
.
2022
;
41
(
1
):
12
.
14.
Wang
E
,
Wang
LC
,
Tsai
CY
,
Bhoj
V
,
Gershenson
Z
,
Moon
E
, et al
.
Generation of potent T-cell immunotherapy for cancer using DAP12-based, multichain, chimeric immunoreceptors
.
Cancer Immunol Res
.
2015
;
3
(
7
):
815
26
.
15.
Kintz
H
,
Nylen
E
,
Barber
A
.
Inclusion of Dap10 or 4-1BB costimulation domains in the chPD1 receptor enhances anti-tumor efficacy of T cells in murine models of lymphoma and melanoma
.
Cell Immunol
.
2020
;
351
:
104069
.
16.
Feucht
J
,
Sun
J
,
Eyquem
J
,
Ho
YJ
,
Zhao
Z
,
Leibold
J
, et al
.
Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency
.
Nat Med
.
2019
;
25
(
1
):
82
8
.
17.
Watanabe
K
,
Terakura
S
,
Martens
AC
,
van Meerten
T
,
Uchiyama
S
,
Imai
M
, et al
.
Target antigen density governs the efficacy of anti–CD20-CD28-CD3 ζ chimeric antigen receptor–modified effector CD8+ T cells
.
J Immunol
.
2015
;
194
(
3
):
911
20
.
18.
Maude
SL
,
Frey
N
,
Shaw
PA
,
Aplenc
R
,
Barrett
DM
,
Bunin
NJ
, et al
.
Chimeric antigen receptor T cells for sustained remissions in leukemia
.
N Engl J Med
.
2014
;
371
(
16
):
1507
17
.
19.
Baeuerle
PA
,
Ding
J
,
Patel
E
,
Thorausch
N
,
Horton
H
,
Gierut
J
, et al
.
Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response
.
Nat Commun
.
2019
;
10
(
1
):
2087
.
20.
Mansilla-Soto
J
,
Eyquem
J
,
Haubner
S
,
Hamieh
M
,
Feucht
J
,
Paillon
N
, et al
.
HLA-independent T cell receptors for targeting tumors with low antigen density
.
Nat Med
.
2022
;
28
(
2
):
345
52
.
21.
Helsen
CW
,
Hammill
JA
,
Lau
VWC
,
Mwawasi
KA
,
Afsahi
A
,
Bezverbnaya
K
, et al
.
The chimeric TAC receptor co-opts the T cell receptor yielding robust anti-tumor activity without toxicity
.
Nat Commun
.
2018
;
9
(
1
):
3049
.
22.
Shah
K
,
Al-Haidari
A
,
Sun
J
,
Kazi
JU
.
T cell receptor (TCR) signaling in health and disease
.
Signal Transduct Target Ther
.
2021
;
6
(
1
):
412
.
23.
Batlevi
CL
,
Matsuki
E
,
Brentjens
RJ
,
Younes
A
.
Novel immunotherapies in lymphoid malignancies
.
Nat Rev Clin Oncol
.
2016
;
13
(
1
):
25
40
.
24.
Birtel
M
,
Voss
RH
,
Reinhard
K
,
Rengstl
B
,
Ouchan
Y
,
Michel
K
, et al
.
A TCR-like CAR promotes sensitive antigen recognition and controlled T-cell expansion upon mRNA vaccination
.
Cancer Res Commun
.
2022
;
2
(
8
):
827
41
.
25.
Eyquem
J
,
Mansilla-Soto
J
,
Giavridis
T
,
van der Stegen
SJC
,
Hamieh
M
,
Cunanan
KM
, et al
.
Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection
.
Nature
.
2017
;
543
(
7643
):
113
7
.
26.
Couzin-Frankel
J
.
Breakthrough of the year 2013. Cancer immunotherapy
.
Science
.
2013
;
342
(
6165
):
1432
3
.
27.
Wang
G
,
Zhou
X
,
Fucà
G
,
Dukhovlinova
E
,
Shou
P
,
Li
H
, et al
.
Fully human antibody VH domains to generate mono and bispecific CAR to target solid tumors
.
J Immunother Cancer
.
2021
;
9
(
4
):
e002173
.
28.
Lee
L
,
Draper
B
,
Chaplin
N
,
Philip
B
,
Chin
M
,
Galas-Filipowicz
D
, et al
.
An APRIL-based chimeric antigen receptor for dual targeting of BCMA and TACI in multiple myeloma
.
Blood
.
2018
;
131
(
7
):
746
58
.
29.
Qin
H
,
Edwards
JP
,
Zaritskaya
L
,
Gupta
A
,
Mu
CJ
,
Fry
TJ
, et al
.
Chimeric antigen receptors incorporating D domains targeting CD123 direct potent mono- and Bi-specific antitumor activity of T cells
.
Mol Ther
.
2019
;
27
(
7
):
1262
74
.
30.
Dustin
ML
,
Chakraborty
AK
,
Shaw
AS
.
Understanding the structure and function of the immunological synapse
.
Cold Spring Harb Perspect Biol
.
2010
;
2
(
10
):
a002311
.
31.
Barden
M
,
Holzinger
A
,
Velas
L
,
Mezősi-Csaplár
M
,
Szöőr
Á
,
Vereb
G
, et al
.
CAR and TCR form individual signaling synapses and do not cross-activate, however, can co-operate in T cell activation
.
Front Immunol
.
2023
;
14
:
1110482
.
32.
Majzner
RG
,
Rietberg
SP
,
Sotillo
E
,
Dong
R
,
Vachharajani
VT
,
Labanieh
L
, et al
.
Tuning the antigen density requirement for CAR T-cell activity
.
Cancer Discov
.
2020
;
10
(
5
):
702
23
.
33.
Kawalekar
OU
,
O' Connor
RS
,
Fraietta
JA
,
Guo
L
,
McGettigan
SE
,
Posey
AD
Jr
, et al
.
Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells
.
Immunity
.
2016
;
44
(
3
):
712
90
.
34.
Seitz
CM
,
Mittelstaet
J
,
Atar
D
,
Hau
J
,
Reiter
S
,
Illi
C
, et al
.
Novel adapter CAR-T cell technology for precisely controllable multiplex cancer targeting
.
Oncoimmunology
.
2021
;
10
(
1
):
2003532
.
35.
Rodgers
DT
,
Mazagova
M
,
Hampton
EN
,
Cao
Y
,
Ramadoss
NS
,
Hardy
IR
, et al
.
Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies
.
Proc Natl Acad Sci USA
.
2016
;
113
(
4
):
E459
68
.
36.
Cartellieri
M
,
Loff
S
,
von Bonin
M
,
Bejestani
EP
,
Ehninger
A
,
Feldmann
A
, et al
.
Unicar: a novel modular retargeting platform technology for CAR T cells
.
Blood
.
2015
;
126
(
23
):
5549
.
37.
Sun
L
,
Gao
F
,
Gao
Z
,
Ao
L
,
Li
N
,
Ma
S
, et al
.
Shed antigen-induced blocking effect on CAR-T cells targeting Glypican-3 in Hepatocellular Carcinoma
.
J Immunother Cancer
.
2021
;
9
(
4
):
e001875
.
38.
Schmidt
MM
,
Thurber
GM
,
Wittrup
KD
.
Kinetics of anti-carcinoembryonic antigen antibody internalization: effects of affinity, bivalency, and stability
.
Cancer Immunol Immunother
.
2008
;
57
(
12
):
1879
90
.
39.
Thurber
GM
,
Schmidt
MM
,
Wittrup
KD
.
Antibody tumor penetration: transport opposed by systemic and antigen-mediated clearance
.
Adv Drug Deliv Rev
.
2008
;
60
(
12
):
1421
34
.
40.
Freskgård
PO
,
Urich
E
.
Antibody therapies in CNS diseases
.
Neuropharmacology
.
2017
;
120
:
38
55
.
41.
Wahl
AS
,
Correa
D
,
Imobersteg
S
,
Maurer
MA
,
Kaiser
J
,
Augath
MA
, et al
.
Targeting therapeutic antibodies to the CNS: a comparative study of intrathecal, intravenous, and subcutaneous anti-nogo a antibody treatment after stroke in rats
.
Neurotherapeutics
.
2020
;
17
(
3
):
1153
9
.
42.
Perissinotti
AJ
,
Reeves
DJ
.
Role of intrathecal rituximab and trastuzumab in the management of leptomeningeal carcinomatosis
.
Ann Pharmacother
.
2010
;
44
(
10
):
1633
40
.
43.
Fry
TJ
,
Shah
NN
,
Orentas
RJ
,
Stetler-Stevenson
M
,
Yuan
CM
,
Ramakrishna
S
, et al
.
CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy
.
Nat Med
.
2018
;
24
(
1
):
20
8
.
44.
Larson
SM
,
Walthers
CM
,
Ji
B
,
Ghafouri
SN
,
Naparstek
J
,
Trent
J
, et al
.
CD19/CD20 bispecific chimeric antigen receptor (CAR) in naive/memory T cells for the treatment of relapsed or refractory non-hodgkin lymphoma
.
Cancer Discov
.
2023
;
13
(
3
):
580
97
.
45.
Schneider
D
,
Xiong
Y
,
Wu
D
,
Hu
P
,
Alabanza
L
,
Steimle
B
, et al
.
Trispecific CD19-CD20-CD22-targeting duoCAR-T cells eliminate antigen-heterogeneous B cell tumors in preclinical models
.
Sci Transl Med
.
2021
;
13
(
586
):
eabc6401
.
46.
Cleary
KLS
,
Chan
HTC
,
James
S
,
Glennie
MJ
,
Cragg
MS
.
Antibody distance from the cell membrane regulates antibody effector mechanisms
.
J Immunol
.
2017
;
198
(
10
):
3999
4011
.
47.
Al-Aghbar
MA
,
Jainarayanan
AK
,
Dustin
ML
,
Roffler
SR
.
The interplay between membrane topology and mechanical forces in regulating T cell receptor activity
.
Commun Biol
.
2022
;
5
(
1
):
40
.
48.
Dickopf
S
,
Georges
GJ
,
Brinkmann
U
.
Format and geometries matter: structure-based design defines the functionality of bispecific antibodies
.
Comput Struct Biotechnol J
.
2020
;
18
:
1221
7
.
49.
Haso
W
,
Lee
DW
,
Shah
NN
,
Stetler-Stevenson
M
,
Yuan
CM
,
Pastan
IH
, et al
.
Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia
.
Blood
.
2013
;
121
(
7
):
1165
74
.
50.
Watanabe
N
,
Bajgain
P
,
Sukumaran
S
,
Ansari
S
,
Heslop
HE
,
Rooney
CM
, et al
.
Fine-tuning the CAR spacer improves T-cell potency
.
Oncoimmunology
.
2016
;
5
(
12
):
e1253656
.
51.
Klein
JS
,
Gnanapragasam
PNP
,
Galimidi
RP
,
Foglesong
CP
,
West
AP
Jr
,
Bjorkman
PJ
.
Examination of the contributions of size and avidity to the neutralization mechanisms of the anti-HIV antibodies b12 and 4E10
.
Proc Natl Acad Sci USA
.
2009
;
106
(
18
):
7385
90
.
52.
Jin
J
,
Cheng
J
,
Huang
M
,
Luo
H
,
Zhou
J
.
Fueling chimeric antigen receptor T cells with cytokines
.
Am J Cancer Res
.
2020
;
10
(
12
):
4038
55
.
53.
Atar
D
,
Mast
AS
,
Scheuermann
S
,
Ruoff
L
,
Seitz
CM
,
Schlegel
P
.
Adapter CAR T cell therapy for the treatment of B-lineage lymphomas
.
Biomedicines
.
2022
;
10
(
10
):
2420
.
54.
Benmebarek
M-R
,
Cadilha
BL
,
Herrmann
M
,
Lesch
S
,
Schmitt
S
,
Stoiber
S
, et al
.
A modular and controllable T cell therapy platform for acute myeloid leukemia
.
Leukemia
.
2021
;
35
(
8
):
2243
57
.
55.
Stock
S
,
Benmebarek
MR
,
Kluever
AK
,
Darowski
D
,
Jost
C
,
Stubenrauch
KG
, et al
.
Chimeric antigen receptor T cells engineered to recognize the P329G-mutated Fc part of effector-silenced tumor antigen-targeting human IgG1 antibodies enable modular targeting of solid tumors
.
J Immunother Cancer
.
2022
;
10
(
7
):
e005054
.
56.
Ehninger
A
,
Kramer
M
,
Röllig
C
,
Thiede
C
,
Bornhäuser
M
,
von Bonin
M
, et al
.
Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia
.
Blood Cancer J
.
2014
;
4
(
6
):
e218
.
57.
Munshi
NC
,
Anderson
LD
Jr
,
Shah
N
,
Madduri
D
,
Berdeja
J
,
Lonial
S
, et al
.
Idecabtagene vicleucel in relapsed and refractory multiple myeloma
.
N Engl J Med
.
2021
;
384
(
8
):
705
16
.
58.
Seidel
UJE
,
Schlegel
P
,
Grosse-Hovest
L
,
Hofmann
M
,
Aulwurm
S
,
Pyz
E
, et al
.
Reduction of minimal residual disease in pediatric B-lineage acute lymphoblastic leukemia by an fc-optimized CD19 antibody
.
Mol Ther
.
2016
;
24
(
9
):
1634
43
.
59.
Yuraszeck
T
,
Kasichayanula
S
,
Benjamin
JE
.
Translation and clinical development of bispecific T-cell engaging antibodies for cancer treatment
.
Clin Pharmacol Ther
.
2017
;
101
(
5
):
634
45
.
60.
Berinstein
NL
,
Grillo-López
AJ
,
White
CA
,
Bence-Bruckler
I
,
Maloney
D
,
Czuczman
M
, et al
.
Association of serum Rituximab (IDEC-C2B8) concentration and anti-tumor response in the treatment of recurrent low-grade or follicular non-Hodgkin's lymphoma
.
Ann Oncol
.
1998
;
9
(
9
):
995
1001
.
61.
Munoz
J
,
Jaglowski
S
,
McKinney
MS
,
Isufi
I
,
Stiff
PJ
,
Sachs
J
, et al
.
A phase 1 study of ACTR087 in combination with rituximab, in subjects with relapsed or refractory CD20-positive B-cell lymphoma
.
Blood
.
2019
;
134
(
Suppl ment_1
):
244
.
62.
Picca
A
,
Psimaras
D
.
Chapter 31 - neurological complications of immunotherapy and monoclonal antibody therapy
. In:
Newton
HB
,
Malkin
MG
, editors.
Neurological complications of systemic cancer and antineoplastic therapy
. 2nd ed.
Academic Press
;
2022
. p.
521
36
.
63.
Lu
R-M
,
Hwang
YC
,
Liu
IJ
,
Lee
CC
,
Tsai
HZ
,
Li
HJ
, et al
.
Development of therapeutic antibodies for the treatment of diseases
.
J Biomed Sci
.
2020
;
27
(
1
):
1
.
64.
Bander
NH
.
Antibody-drug conjugate target selection: critical factors
.
Methods Mol Biol
.
2013
;
1045
:
29
40
.
65.
Roßkopf
S
,
Eichholz
KM
,
Winterberg
D
,
Diemer
KJ
,
Lutz
S
,
Münnich
IA
, et al
.
Enhancing CDC and ADCC of CD19 antibodies by combining Fc protein-engineering with Fc glyco-engineering
.
Antibodies
.
2020
;
9
(
4
):
63
.
66.
Lazar
GA
,
Dang
W
,
Karki
S
,
Vafa
O
,
Peng
JS
,
Hyun
L
, et al
.
Engineered antibody Fc variants with enhanced effector function
.
Proc Natl Acad Sci USA
.
2006
;
103
(
11
):
4005
10
.
67.
Lund
J
,
Winter
G
,
Jones
PT
,
Pound
JD
,
Tanaka
T
,
Walker
MR
, et al
.
Human Fc gamma RI and Fc gamma RII interact with distinct but overlapping sites on human IgG
.
J Immunol
.
1991
;
147
(
8
):
2657
62
.
68.
Hudecek
M
,
Sommermeyer
D
,
Kosasih
PL
,
Silva-Benedict
A
,
Liu
L
,
Rader
C
, et al
.
The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity
.
Cancer Immunol Res
.
2015
;
3
(
2
):
125
35
.
69.
Cao
X
,
Chen
J
,
Li
B
,
Dang
J
,
Zhang
W
,
Zhong
X
, et al
.
Promoting antibody-dependent cellular phagocytosis for effective macrophage-based cancer immunotherapy
.
Sci Adv
.
2022
;
8
(
11
):
eabl9171
.
70.
Lei
A
,
Yu
H
,
Lu
S
,
Lu
H
,
Ding
X
,
Tan
T
, et al
.
A second-generation M1-polarized CAR macrophage with antitumor efficacy
.
Nat Immunol
.
2024
;
25
(
1
):
102
16
.
71.
Zhang
L
,
Tian
L
,
Dai
X
,
Yu
H
,
Wang
J
,
Lei
A
, et al
.
Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions
.
J Hematol Oncol
.
2020
;
13
(
1
):
153
.
72.
Li
P
,
Jiang
N
,
Nagarajan
S
,
Wohlhueter
R
,
Selvaraj
P
,
Zhu
C
.
Affinity and kinetic analysis of Fcgamma receptor IIIa (CD16a) binding to IgG ligands
.
J Biol Chem
.
2007
;
282
(
9
):
6210
21
.
73.
Derer
S
,
Glorius
P
,
Schlaeth
M
,
Lohse
S
,
Klausz
K
,
Muchhal
U
, et al
.
Increasing FcγRIIa affinity of an FcγRIII-optimized anti-EGFR antibody restores neutrophil-mediated cytotoxicity
.
mAbs
.
2014
;
6
(
2
):
409
21
.
74.
Peipp
M
,
Lammerts van Bueren
JJ
,
Schneider-Merck
T
,
Bleeker
WWK
,
Dechant
M
,
Beyer
T
, et al
.
Antibody fucosylation differentially impacts cytotoxicity mediated by NK and PMN effector cells
.
Blood
.
2008
;
112
(
6
):
2390
9
.
75.
Ratner
M
.
Genentech’s glyco-engineered antibody to succeed Rituxan
.
Nat Biotechnol
.
2014
;
32
(
1
):
6
7
.
76.
Umaña
P
,
Jean-Mairet
J
,
Moudry
R
,
Amstutz
H
,
Bailey
JE
.
Engineered glycoforms of an antineuroblastoma IgG1 with optimized antibody-dependent cellular cytotoxic activity
.
Nat Biotechnol
.
1999
;
17
(
2
):
176
80
.
77.
Bosch
F
,
Cantin
G
,
Cortelezzi
A
,
Knauf
W
,
Tiab
M
,
Turgut
M
, et al
.
Obinutuzumab plus fludarabine and cyclophosphamide in previously untreated, fit patients with chronic lymphocytic leukemia: a subgroup analysis of the GREEN study
.
Leukemia
.
2020
;
34
(
2
):
441
50
.
78.
Salles
G
,
Duell
J
,
González Barca
E
,
Tournilhac
O
,
Jurczak
W
,
Liberati
AM
, et al
.
Tafasitamab plus lenalidomide in relapsed or refractory diffuse large B-cell lymphoma (L-MIND): a multicentre, prospective, single-arm, phase 2 study
.
Lancet Oncol
.
2020
;
21
(
7
):
978
88
.
79.
Moreno
C
,
Greil
R
,
Demirkan
F
,
Tedeschi
A
,
Anz
B
,
Larratt
L
, et al
.
Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial
.
Lancet Oncol
.
2019
;
20
(
1
):
43
56
.
80.
Yu
X
,
Orr
CM
,
Chan
HTC
,
James
S
,
Penfold
CA
,
Kim
J
, et al
.
Reducing affinity as a strategy to boost immunomodulatory antibody agonism
.
Nature
.
2023
;
614
(
7948
):
539
47
.
81.
Liu
T
,
Jiang
S
,
Teng
X
,
Zhong
L
,
Liu
M
,
Jin
Y
, et al
.
A comparison of panitumumab and cetuximab in the treatment of KRAS wild-type metastatic colorectal cancer: a systematic review and meta-analysis
.
Immunopharmacol Immunotoxicol
.
2023
;
45
(
1
):
1
9
.
82.
Wang
B
,
Gallolu Kankanamalage
S
,
Dong
J
,
Liu
Y
.
Optimization of therapeutic antibodies
.
Antib Ther
.
2021
;
4
(
1
):
45
54
.
83.
Adams
GP
,
Schier
R
,
McCall
AM
,
Simmons
HH
,
Horak
EM
,
Alpaugh
RK
, et al
.
High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules
.
Cancer Res
.
2001
;
61
(
12
):
4750
5
.
84.
Kim
GP
,
Grothey
A
.
Targeting colorectal cancer with human anti-EGFR monoclonocal antibodies: focus on panitumumab
.
Biologics
.
2008
;
2
(
2
):
223
8
.
85.
Labrijn
AF
,
Buijsse
AO
,
van den Bremer
ETJ
,
Verwilligen
AYW
,
Bleeker
WK
,
Thorpe
SJ
, et al
.
Therapeutic IgG4 antibodies engage in Fab-arm exchange with endogenous human IgG4 in vivo
.
Nat Biotechnol
.
2009
;
27
(
8
):
767
71
.
86.
Xiao
Q
,
Zhang
X
,
Tu
L
,
Cao
J
,
Hinrichs
CS
,
Su
X
.
Size-dependent activation of CAR-T cells
.
Sci Immunol
.
2022
;
7
(
74
):
eabl3995
.
87.
Abraham
AR
,
Maghsoudlou
P
,
Copland
DA
,
Nicholson
LB
,
Dick
AD
.
CAR-Treg cell therapies and their future potential in treating ocular autoimmune conditions
.
Front Ophthalmol
.
2023
;
3
.
88.
Marin
D
,
Li
Y
,
Basar
R
,
Rafei
H
,
Daher
M
,
Dou
J
, et al
.
Safety, efficacy and determinants of response of allogeneic CD19-specific CAR-NK cells in CD19+ B cell tumors: a phase 1/2 trial
.
Nat Med
.
2024
;
30
(
3
):
772
84
.
89.
Heczey
A
,
Xu
X
,
Courtney
AN
,
Tian
G
,
Barragan
GA
,
Guo
L
, et al
.
Anti-GD2 CAR-NKT cells in relapsed or refractory neuroblastoma: updated phase 1 trial interim results
.
Nat Med
.
2023
;
29
(
6
):
1379
88
.
90.
Liu
R
,
Oldham
RJ
,
Teal
E
,
Beers
SA
,
Cragg
MS
.
Fc-engineering for modulated effector functions-improving antibodies for cancer treatment
.
Antibodies
.
2020
;
9
(
4
):
64
.
91.
Koh
CH
,
Lee
S
,
Kwak
M
,
Kim
BS
,
Chung
Y
.
CD8 T-cell subsets: heterogeneity, functions, and therapeutic potential
.
Exp Mol Med
.
2023
;
55
(
11
):
2287
99
.
92.
Luckheeram
RV
,
Zhou
R
,
Verma
AD
,
Xia
B
.
CD4⁺T cells: differentiation and functions
.
Clin Dev Immunol
.
2012
;
2012
:
925135
.
93.
Wolf
NK
,
Kissiov
DU
,
Raulet
DH
.
Roles of natural killer cells in immunity to cancer, and applications to immunotherapy
.
Nat Rev Immunol
.
2023
;
23
(
2
):
90
105
.
94.
Look
A
,
Burns
D
,
Tews
I
,
Roghanian
A
,
Mansour
S
.
Towards a better understanding of human iNKT cell subpopulations for improved clinical outcomes
.
Front Immunol
.
2023
;
14
:
1176724
.
95.
Gruijs
M
,
Sewnath
CAN
,
van Egmond
M
.
Therapeutic exploitation of neutrophils to fight cancer
.
Semin Immunol
.
2021
;
57
:
101581
.
96.
Mantovani
A
,
Allavena
P
,
Marchesi
F
,
Garlanda
C
.
Macrophages as tools and targets in cancer therapy
.
Nat Rev Drug Discov
.
2022
;
21
(
11
):
799
820
.
97.
Chmielewski
M
,
Abken
H
.
CAR T cells transform to trucks: chimeric antigen receptor-redirected T cells engineered to deliver inducible IL-12 modulate the tumour stroma to combat cancer
.
Cancer Immunol Immunother
.
2012
;
61
(
8
):
1269
77
.
98.
Guo
T
,
Ma
D
,
Lu
TK
.
Sense-and-Respond payload delivery using a novel antigen-inducible promoter improves suboptimal CAR-T activation
.
ACS Synth Biol
.
2022
;
11
(
4
):
1440
53
.
99.
Rydzek
J
,
Nerreter
T
,
Peng
H
,
Jutz
S
,
Leitner
J
,
Steinberger
P
, et al
.
Chimeric antigen receptor library screening using a novel NF-κB/NFAT reporter cell platform
.
Mol Ther
.
2019
;
27
(
2
):
287
99
.
100.
Kankeu Fonkoua
LA
,
Sirpilla
O
,
Sakemura
R
,
Siegler
EL
,
Kenderian
SS
.
CAR T cell therapy and the tumor microenvironment: current challenges and opportunities
.
Mol Ther Oncolytics
.
2022
;
25
:
69
77
.
101.
Li
L
,
Li
Q
,
Yan
ZX
,
Sheng
LS
,
Fu
D
,
Xu
P
, et al
.
Transgenic expression of IL-7 regulates CAR-T cell metabolism and enhances in vivo persistence against tumor cells
.
Sci Rep
.
2022
;
12
(
1
):
12506
.
102.
Alizadeh
D
,
Wong
RA
,
Yang
X
,
Wang
D
,
Pecoraro
JR
,
Kuo
CF
, et al
.
IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype
.
Cancer Immunol Res
.
2019
;
7
(
5
):
759
72
.
103.
Du
L
,
Nai
Y
,
Shen
M
,
Li
T
,
Huang
J
,
Han
X
, 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
;
8
:
675179
.
104.
Kueberuwa
G
,
Kalaitsidou
M
,
Cheadle
E
,
Hawkins
RE
,
Gilham
DE
.
CD19 CAR T cells expressing IL-12 eradicate lymphoma in fully lymphoreplete mice through induction of host immunity
.
Mol Ther Oncolytics
.
2018
;
8
:
41
51
.
105.
Svoboda
J
,
Gerson
JN
,
Landsburg
DJ
,
Chong
EA
,
Barta
SK
,
Dwivedy Nasta
S
, et al
.
Interleukin-18 secreting autologous anti-CD19 CAR T-cells (huCART19-IL18) in patients with non-hodgkin lymphomas relapsed or refractory to prior CAR T-cell therapy
.
Blood
.
2022
;
140
(
Suppl 1
):
4612
4
.
106.
Kubin
M
,
Kamoun
M
,
Trinchieri
G
.
Interleukin 12 synergizes with B7/CD28 interaction in inducing efficient proliferation and cytokine production of human T cells
.
J Exp Med
.
1994
;
180
(
1
):
211
22
.
107.
Ferlazzo
G
,
Pack
M
,
Thomas
D
,
Paludan
C
,
Schmid
D
,
Strowig
T
, et al
.
Distinct roles of IL-12 and IL-15 in human natural killer cell activation by dendritic cells from secondary lymphoid organs
.
Proc Natl Acad Sci USA
.
2004
;
101
(
47
):
16606
11
.
108.
Lu
X
.
Impact of IL-12 in cancer
.
Curr Cancer Drug Targets
.
2017
;
17
(
8
):
682
97
.
109.
Narayan
V
,
Barber-Rotenberg
JS
,
Jung
IY
,
Lacey
SF
,
Rech
AJ
,
Davis
MM
, et al
.
PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial
.
Nat Med
.
2022
;
28
(
4
):
724
34
.
110.
Liu
X
,
Zhang
Y
,
Li
K
,
Liu
Y
,
Xu
J
,
Ma
J
, et al
.
A novel dominant-negative PD-1 armored anti-CD19 CAR T cell is safe and effective against refractory/relapsed B cell lymphoma
.
Transl Oncol
.
2021
;
14
(
7
):
101085
.
111.
Agarwal
S
,
Aznar
MA
,
Rech
AJ
,
Good
CR
,
Kuramitsu
S
,
Da
T
, et al
.
Deletion of the inhibitory co-receptor CTLA-4 enhances and invigorates chimeric antigen receptor T cells
.
Immunity
.
2023
;
56
(
10
):
2388
407.e9
.
112.
Zhang
J
,
Hu
Y
,
Yang
J
,
Li
W
,
Zhang
M
,
Wang
Q
, et al
.
Non-viral, specifically targeted CAR-T cells achieve high safety and efficacy in B-NHL
.
Nature
.
2022
;
609
(
7926
):
369
74
.
113.
Liu
X
,
Ranganathan
R
,
Jiang
S
,
Fang
C
,
Sun
J
,
Kim
S
, et al
.
A chimeric switch-receptor targeting PD1 augments the efficacy of second-generation CAR T cells in advanced solid tumors
.
Cancer Res
.
2016
;
76
(
6
):
1578
90
.
114.
Noh
K-E
,
Lee
JH
,
Choi
SY
,
Jung
NC
,
Nam
JH
,
Oh
JS
, et al
.
TGF-β/IL-7 chimeric switch receptor-expressing CAR-T cells inhibit recurrence of CD19-positive B cell lymphoma
.
Int J Mol Sci
.
2021
;
22
(
16
):
8706
.
115.
Flanagan
RJ
,
Jones
AL
.
Fab antibody fragments: some applications in clinical toxicology
.
Drug Saf
.
2004
;
27
(
14
):
1115
33
.
116.
Adams
R
,
Griffin
L
,
Compson
JE
,
Jairaj
M
,
Baker
T
,
Ceska
T
, et al
.
Extending the half-life of a fab fragment through generation of a humanized anti-human serum albumin Fv domain: an investigation into the correlation between affinity and serum half-life
.
mAbs
.
2016
;
8
(
7
):
1336
46
.
117.
Adachi
K
,
Kano
Y
,
Nagai
T
,
Okuyama
N
,
Sakoda
Y
,
Tamada
K
.
IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor
.
Nat Biotechnol
.
2018
;
36
(
4
):
346
51
.
118.
Locke
FL
,
Filosto
S
,
Chou
J
,
Vardhanabhuti
S
,
Perbost
R
,
Dreger
P
, et al
.
Impact of tumor microenvironment on efficacy of anti-CD19 CAR T cell therapy or chemotherapy and transplant in large B cell lymphoma
.
Nat Med
.
2024
;
30
(
2
):
507
18
.
119.
Boyman
O
,
Purton
JF
,
Surh
CD
,
Sprent
J
.
Cytokines and T-cell homeostasis
.
Curr Opin Immunol
.
2007
;
19
(
3
):
320
6
.
120.
Lei
W
,
Zhao
A
,
Liu
H
,
Yang
C
,
Wei
C
,
Guo
S
, et al
.
Safety and feasibility of anti-CD19 CAR T cells expressing inducible IL-7 and CCL19 in patients with relapsed or refractory large B-cell lymphoma
.
Cell Discov
.
2024
;
10
(
1
):
5
.
121.
Tamada
K
,
Geng
D
,
Sakoda
Y
,
Bansal
N
,
Srivastava
R
,
Li
Z
, et al
.
Redirecting gene-modified T cells toward various cancer types using tagged antibodies
.
Clin Cancer Res
.
2012
;
18
(
23
):
6436
45
.
122.
Liang
A
,
Ye
S
,
Li
P
,
Huang
J
,
Zhu
S
,
Yao
X
, et al
.
Safety and efficacy of a novel anti-CD20 chimeric antigen receptor (CAR)-T cell therapy in relapsed/refractory (r/r) B-cell non-Hodgkin lymphoma (B-NHL) patients after failing CD19 CAR-T therapy
.
J Clin Oncol
.
2021
;
39
(
15_Suppl l
):
2508
.
123.
Spiegel
JY
,
Patel
S
,
Muffly
L
,
Hossain
NM
,
Oak
J
,
Baird
JH
, 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
;
27
(
8
):
1419
31
.
124.
Wang
V
,
Gauthier
M
,
Decot
V
,
Reppel
L
,
Bensoussan
D
.
Systematic review on CAR-T cell clinical trials up to 2022: academic center input
.
Cancers
.
2023
;
15
(
4
):
1003
.
125.
Kantarjian
H
,
Stein
A
,
Gökbuget
N
,
Fielding
AK
,
Schuh
AC
,
Ribera
JM
, et al
.
Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia
.
N Engl J Med
.
2017
;
376
(
9
):
836
47
.
126.
Kantarjian
HM
,
DeAngelo
DJ
,
Stelljes
M
,
Martinelli
G
,
Liedtke
M
,
Stock
W
, et al
.
Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia
.
N Engl J Med
.
2016
;
375
(
8
):
740
53
.
127.
Chong
EA
,
Alanio
C
,
Svoboda
J
,
Nasta
SD
,
Landsburg
DJ
,
Lacey
SF
, et al
.
Pembrolizumab for B-cell lymphomas relapsing after or refractory to CD19-directed CAR T-cell therapy
.
Blood
.
2022
;
139
(
7
):
1026
38
.
128.
Müller
F
,
Taubmann
J
,
Bucci
L
,
Wilhelm
A
,
Bergmann
C
,
Völkl
S
, et al
.
CD19 CAR T-cell therapy in autoimmune disease — a case series with follow-up
.
N Engl J Med
.
2024
;
390
(
8
):
687
700
.
129.
Ueda
T
,
Shiina
S
,
Iriguchi
S
,
Terakura
S
,
Kawai
Y
,
Kabai
R
, et al
.
Optimization of the proliferation and persistency of CAR T cells derived from human induced pluripotent stem cells
.
Nat Biomed Eng
.
2023
;
7
(
1
):
24
37
.
130.
Glienke
W
,
Dragon
AC
,
Zimmermann
K
,
Martyniszyn-Eiben
A
,
Mertens
M
,
Abken
H
, et al
.
GMP-compliant manufacturing of TRUCKs: CAR T cells targeting GD(2) and releasing inducible IL-18
.
Front Immunol
.
2022
;
13
:
839783
.
131.
Shum
T
,
Omer
B
,
Tashiro
H
,
Kruse
RL
,
Wagner
DL
,
Parikh
K
, et al
.
Constitutive signaling from an engineered IL7 receptor promotes durable tumor elimination by tumor-redirected T cells
.
Cancer Discov
.
2017
;
7
(
11
):
1238
47
.
132.
Choe
JH
,
Watchmaker
PB
,
Simic
MS
,
Gilbert
RD
,
Li
AW
,
Krasnow
NA
, et al
.
SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma
.
Sci Transl Med
.
2021
;
13
(
591
):
eabe7378
.
133.
Blaeschke
F
,
Chen
YY
,
Apathy
R
,
Daniel
B
,
Chen
AY
,
Chen
PA
, et al
.
Modular pooled discovery of synthetic knockin sequences to program durable cell therapies
.
Cell
.
2023
;
186
(
19
):
4216
34.e33
.
134.
Wu
C-Y
,
Roybal
KT
,
Puchner
EM
,
Onuffer
J
,
Lim
WA
.
Remote control of therapeutic T cells through a small molecule–gated chimeric receptor
.
Science
.
2015
;
350
(
6258
):
aab4077
.
135.
Leung
WH
,
Gay
J
,
Martin
U
,
Garrett
TE
,
Horton
HM
,
Certo
MT
, et al
.
Sensitive and adaptable pharmacological control of CAR T cells through extracellular receptor dimerization
.
JCI Insight
.
2019
;
5
(
11
):
e124430
.
136.
Cooper
TM
,
Wu
V
,
Wilson
A
,
Appelbaum
J
,
Jarjour
J
,
Gustafson
J
, et al
.
Pediatric and young adult leukemia adoptive therapy (PLAT)-08: a phase 1 study of SC-DARIC33 in pediatric and young adults with relapsed or refractory CD33+ AML
.
J Clin Oncol
.
2022
;
40
(
16_Suppl l
):
TPS7078
.