Skip to main content

Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy

Abstract

Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for “off-the-shelf” production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.

Introduction

Asthma, a chronic lung disease affecting over 300 million people globally [1], remains a significant healthcare burden. Despite advancements in treatment, severe asthma can lead to frequent flare-ups and life-threatening complications [2, 3]. Current therapies like corticosteroids offer some relief but may not fully control the underlying inflammation or prevent exacerbations [4]. Although several FDA-approved mAb therapies, such as omalizumab and dupilumab, offer some clinical benefit in severe atopic asthma, these treatments necessitate frequent administration due to their short half-life. Furthermore, they primarily manage symptoms by reducing exacerbation frequency and severity, rather than providing a definitive cure [5].

The involvement of NK cells in asthma remains complex and not fully understood. While traditionally known for their antiviral function [6], NK cells might play a dual role in allergic asthma, either promoting or suppressing the disease [7]. Their activation is influenced by a balance of inhibitory and stimulatory signals from various receptors [8,9,10]. These receptors recognize molecules like MHC-I, cytokines, and microbial components. Human NK cells can be categorized into two subsets based on CD56 expression [11]. Studies have observed changes in NK cell phenotype and function in asthma patients. For example, patients with severe asthma exhibit increased expression of CD69 and NKG2D on their NK cells [12, 13]. Research using mouse models further highlights the complexity of NK cells’ role in asthma. Some studies suggest their involvement in allergic inflammation [14], while others indicate minimal or even a resolving effect [4, 15]. Furthermore, NK cells have been shown to produce various immune mediators like IFN-γ and TNF-α upon IgE stimulation, and they can even attack IgE-coated target cells [16]. This suggests a potential contribution of NK cells to IgE-mediated allergic responses.

Recent successes in cancer immunotherapy utilizing CAR-based therapies against tumor-specific neoantigens (e.g., HSP70) suggest the potential application of CAR technology for targeted immunotherapies in asthma. CAR technology offers an approach to engineer immune cells for antigen-specific recognition [17]. Several researchers have explored the potential of CAR-T cell therapy for allergic diseases like asthma [18,19,20,21,22]. One approach focuses on eliminating B cells that produce IgE, a key antibody in allergic reactions. Studies are investigating CARs that bind to membrane-bound IgE on B cells, offering the potential for long-term suppression with a single treatment [18, 19]. Another approach targets eosinophils, white blood cells involved in allergic inflammation. Engineered CAR-T cells can eliminate these cells and potentially suppress asthmatic symptoms [20, 21]. Additionally, research is ongoing to manipulate Tregs with CARs to suppress allergic airway inflammation [22]. While no current clinical trials test CAR therapy for asthma, these studies suggest a promising future direction for treating allergic diseases. Although CAR-T cell therapy has succeeded in various diseases, limitations like adverse effects, cytotoxicity, and T-cell exhaustion (terminal differentiation) highlight the need for alternative cell types [23,24,25].

NK cells offer several advantages over T cells for CAR-based immunotherapy in the context of asthma. Unlike T cells requiring antigen presentation, NK cells utilize innate receptors for activation. This allows CAR-NK cells to retain cytotoxic function even if the target molecule changes, potentially overcoming a limitation of CAR-T cell therapy [26]. Additionally, NK cells have a shorter lifespan in vivo, potentially reducing the risk of severe side effects associated with CAR-T cells, such as CRS and GVHD [27].

While the core structure of the CAR construct might be similar in CAR-NK and CAR-T cells, NK cells offer additional functionalities. They utilize co-stimulatory molecules like NKG2D and CD244 (2B4) to amplify their cytotoxic activity and cytokine production [27]. Furthermore, NK cells possess a broader recognition repertoire beyond the specifically targeted antigen through receptors like NCRs, NKG2D, and DNAM-1 [28, 29]. This broader recognition potential translates to a more robust and potentially more effective targeting strategy in the context of asthma.

Although research on CAR cell therapy for asthma is in its early stages, the inherent advantages of NK cells, such as readily available cell sources and potentially lower risk of complications compared to CAR-T cells, suggest a promising future direction. To comprehensively explore this potential, this review will delve into several key areas. First, we’ll examine the evolution and production of CAR designs. Next, we’ll critically assess existing pre-clinical and clinical studies of CAR-based therapies in asthma to understand current progress and identify areas for further research. Additionally, we’ll explore strategies to optimize CAR-NK cell function, focusing on ways to modulate their natural abilities for enhanced efficacy and safety in asthma treatment. Furthermore, a comparative analysis of CAR-T and CAR-NK cell therapy will be conducted, highlighting their respective advantages and limitations in the context of asthma. Finally, we’ll discuss established and novel CAR designs with promising potential for treating severe asthma. By comprehensively addressing these objectives, this review aims to provide a valuable resource for researchers and clinicians seeking to understand the potential of CAR-NK cell therapy as a novel and promising approach to managing severe asthma.

Evolution of CAR design: From first generation to enhanced functionalities

While CAR-T cell therapy is currently a leading focus in adoptive immunotherapy with success in treating leukemia and lymphomas, CAR-NK cell therapy is emerging as a promising new area with growing preclinical research [30]. The foundation for ACT was established in 1982 by Rosenberg et al., who developed an in vitro culture system capable of activating and expanding autologous PB lymphocytes. The system included NK cells, T cells, NKT cells, and monocytes from PB, with the addition of IL-2. These cultured lymphocytes exhibited enhanced killing abilities compared to NK cells and cytotoxic T lymphocytes, earning them the name lymphokine-activated killer cells [31]. The rapid development of gene engineering in the 1980s introduced novel methods in the field of ACT. The pioneering work of Yoshihisa Kuwana in 1987 described the first CAR, a fusion protein combining the antigen-binding domain of an antibody with the signaling domain of a T cell receptor [32]. This concept was further refined by Arthur et al. in 1991 with the creation of a CAR incorporating the CD3ζ intracellular signaling domain. This iteration marked the final form of the first-generation CAR, characterized by a scFv of immunoglobulin and CD3ζ signaling domains [33]. While First-generation CARs demonstrate antigen recognition, their clinical efficacy is diminished by the inadequate activation and proliferation of the T cells expressing these CARs [34]. In the early 2000s, the significance of co-stimulators was acknowledged and incorporated into CARs, resulting in the second generation. The positive outcomes of clinical trials using second-generation CAR-T targeting CD19 and CD20 in hematopoietic malignancies led to the first FDA approval of CAR-T (Tisagenlecleucel, Kymriah, and Novartis) in 2017 [35,36,37,38]. To enhance its effectiveness, multiple domains from co-stimulators (CD28, CD27, OX40, 4-1BB, etc.) were inserted into CARs. The ongoing pursuit of improved clinical efficacy has driven the exploration of further functionalities within CAR design. Immunomodulatory factors such as cytokines, chemokines, growth factors, IFN, and TNF were added to construct the fourth generation of CARs [39, 40]. The newest generation of CAR technology, known as the fifth generation, introduces an additional co-stimulatory domain to activate novel signaling pathways within CAR cells. This innovative design integrates a co-stimulatory molecule with a fragment of a cytokine receptor, such as the IL2Rβ chain, which then interacts with STAT proteins and CD27 molecules inside the cell. This enhanced structure allows for antigen-specific activation of the JAK/STAT pathway, promoting cell proliferation and preventing premature exhaustion) of the CAR cells [41].

The function of CAR therapy is indeed determined by the structure of the CAR molecule, which acts as a bridge between the immune system and target cells [42]. This section will delve into the specific influence of each component of the CAR molecule on its functionality (Fig. 1).

Fig. 1
figure 1

Unveiling the Emergence and Development of CAR NK Cells: A Classification into Five Generations: The figure depicts a sequential representation of CAR development, categorized by their structural characteristics and costimulatory signaling molecules. 1st generation: The first-generation design, characterized by a simple ScFv-CD3ζ configuration, omits co-stimulatory signals, thereby compromising therapeutic efficacy. 2nd and 3rd generation: The inclusion of one or two costimulatory molecules has been shown to augment NK cell activation and persistence. 4th generation: This approach utilizes transgenic integration to achieve cytokine secretion, specifically IL-12, fostering a multifaceted immune response against targeted cells. 5th generation: The integration of a costimulatory signaling molecule within the cytokine receptor domain of a CAR enhances the coordinated cytolytic activity of CAR- NK cells

Extracellular target-binding domain

This domain, typically incorporating a scFv comprising a heavy chain and a light chain connected through a linker, a nanobody, or a ligand specific for the target antigen, dictates the targeting specificity of CAR cells. Beyond specificity, the binding affinity between the target-binding domain and its cognate antigen significantly influences CAR performance. Deviations from optimal binding affinity, either excessively high or low, can compromise therapeutic efficacy [43]. Consequently, additional factors such as charge density of the target antigen [44], epitope location [45], and overall target antigen density on the target cell surface [46] must also be meticulously considered during CAR design for optimal therapeutic outcomes.

Spacer or hinge domain

The hinge domain, situated extracellularly, acts as a crucial linker between the target-binding domain and the transmembrane domain of the CAR molecule. Emerging evidence suggests that the hinge domain also influences CAR function. The spatial positioning of the target antigen relative to the plasma membrane can be influenced by the hinge length, consequently affecting the binding efficiency of the CAR molecule. Therefore, for optimal design, CAR development should incorporate hinge lengths tailored to the specific target antigen for enhanced therapeutic efficacy [47].

Transmembrane domain

While the primary function of the transmembrane domain is to bind the CAR molecule to the cell membrane, recent research suggests it exerts a more multifaceted influence. This domain can impact the expression level and stability of the CAR itself, modulate the formation of the immunological synapse, and potentially be involved in the dimerization of endogenous signaling molecules [48, 49]. The transmembrane domain frequently utilized in NK cells is sourced from CD3ζ, CD8, or CD28. However, alternative transmembrane segments like NKG2D, 2B4, and DNAM-1 have also been employed for this purpose [50].

Intracellular signal domain

This part integrates both signaling and costimulatory domains to activate NK or T cells. The signaling domain, typically CD3ζ or FcεRIγ, harbors ITAMs that mimic the TCR signaling function and initiate the activation cascade within the cell. Additionally, Costimulatory domains, frequently derived from the CD28 family (CD28, ICOS) or the TNFR family (4-1BB, OX40, CD27), synergize with costimulatory molecules to amplify intracellular activation signals [51, 52].

CAR-NK therapy uses similar engineered receptors (CARs) as CAR-T cell therapy. When genetically modified with a CAR, NK cells gain targeted killing ability alongside their natural cytotoxicity [53]. Researchers are enhancing NK cell killing by replacing components of the T cell CAR (CD3z, CD28, and/or CD137) with signaling molecules naturally found on NK cells, such as 2B4, NKG2D, DAP10, or DAP12. This modification led to a boost in NK cell activation and their ability to destroy cells [54]. To enhance the design of CAR for NK cells, Li et al. conducted a study comparing a single CAR-T cell design with nine different CAR-NK cell designs. These CAR-NK cells featured four distinct transmembrane domains and various intracellular signaling domains, all aimed at targeting the same mesothelin antigen. The research showed that CAR-NK cells equipped with the NKG2D transmembrane domain, 2B4 co-stimulatory domain, and CD3ζ signaling domain displayed potent and specific cytotoxicity against the target antigen [50].

Cell sources and production of CAR-NK

Similar to the well-established workflow for autologous CAR-T therapy, other CAR-engineered immune cells such as CAR-NK, CAR-macrophage, and CAR-CIK typically follow a standardized process. This includes:

  1. 1.

    Leukapheresis to isolate and enrich patient cells.

  2. 2.

    Activation and expansion of the cells.

  3. 3.

    Genetically modifying the cells with a CAR using viral (such as Lentivirus, adeno-associated virus, and 𝛾-retrovirus) or nonviral (such as e transposon systems, CRISPR/Cas9 systems and mRNA electroporation platforms) methods.

  4. 4.

    Expanding the CAR cells outside the body (ex vivo).

  5. 5.

    Formulating and cryopreserving the final cell product.

  6. 6.

    Administering lymphodepleting treatment followed by patient reinfusion of the CAR cells [55,56,57].

Manufacturing CAR cell therapy in the traditional way has several hurdles. The process itself is lengthy, taking up to 22 days and requiring specialized labs, which significantly inflates the cost and potentially delays treatment for patients in urgent need [58]. Furthermore, Isolating cells can be tricky, with a risk of contamination from other blood cells. Additionally, not all these cells readily accept the CAR modification, leading to inconsistencies [59]. The extended time these cells spend growing outside the body (ex vivo) and the processes of freezing and transport can further diminish their effectiveness. Moreover, CAR cell therapy may not be suitable for everyone. Patients who received stem cell transplants may have an altered immune system that reduces the treatment’s efficacy [60, 61]. New developments offer a promising solution to the challenges of CAR cell therapy. Using CAR products derived from donors who are either fully compatible (HLA-identical) or partially compatible (HLA-haplotype matched) with the patient offers a way to overcome these issues [62].

Several cell sources can be used to generate NK cells for CAR-NK therapy, including PBMCs, hPSCs, iPSCs, UCB, and the NK line cell line which are compared in Table 1. PBMCs are a traditional source of NK cells for CAR-NK therapy. These NK cells are typically mature (CD56dimCD16bright) and exhibit consistent characteristics across individuals [63]. They also express high levels of activating receptors (NKG2D, NKp44, NKp46) that enhance their ability [64] However, there are some key limitations associated with using PB-NK cells. Firstly, the limited availability of NK cells in PB (around 10–15%) presents a challenge for collection and ex vivo expansion, potentially hindering large-scale production. Secondly, PB-NK cells exist in various stages of maturity, resulting in a diverse range of receptor expression profiles. This inherent heterogeneity makes it difficult to guarantee the standardization and stability of the final CAR-NK cell product, which is crucial for consistent therapeutic outcomes [65]. In addition, The precise eradication of T cells is essential to minimize the impact of GVHD [66]. Furthermore, incorporating the CAR gene into PB-NK cells is less efficient compared to other cell sources. Prolonged expansion of PB-NK in the lab (in vitro) can shorten telomeres, which are structures on chromosomes that influence cell lifespan. Finally, freezing PB-NK cells (cryopreservation) further reduces their viability and effectiveness [67, 68].

Table 1 Comparation between different sources of NK cells for CAR NK cell production

Another source of NK cells is UCB. UCB is a valuable source of stem cells found in the umbilical cord and placenta after childbirth. These stem cells are unique in their ability to differentiate into various cell types, including blood cells, nerve cells, and immune cells [69]. UCB is rich not only in HSCs but also in immune cells like T cells and NK cells [70]. This makes it a promising source material for developing CAR-T and CAR-NK cell therapies [71, 72]. Compared to adult PB, immune cells in UCB are younger and less developed. They have a greater ability to multiply and a higher proportion of NK cells (15–30%) [73].In addition, this immaturity translates to a specific profile: lower CD16 expression, higher CD56 bright expression, NKG2A positivity, and lower KIR expression [74]. Studies suggest that CAR cells derived from UCB have a higher proportion of memory cells compared to those from adult blood. This could lead to longer persistence in the body and a more sustained immune response, potentially resulting in a more durable therapeutic effect [75, 76]. Additionally, UCB-derived transplants are less likely to cause GVHD, reducing the risk of this serious side effect [77]. Nevertheless, UCB-NK cells exhibit diminished cytotoxic function in vitro due to reduced levels of adhesion molecules and activating receptors, coupled with heightened expression of the inhibitory receptor NKG2A. Moreover, there is a possibility that they may be at risk of incomplete differentiation, restricted lethality, and tumorigenicity [78].

While NK cells can be obtained from PB or UCB, there’s another option. Scientists can also use mature NK cell lines that have undergone modifications to bypass replicative senescence, a natural cellular process that limits cell division [79]. These lines, like NK-92, NKL, HANK1, IMC-1, SNK-6, and SNT-8, can be easily expanded in large quantities to provide enough cells for treatments. Clonal NK cell lines offer advantages for research due to their rapid growth, homogeneity of product, and lack of expression of inhibitory receptors especially inhibitory KIRs and NKG2A [80, 81]. Additionally, the inherent absence of T cells in NK-92 cell lines eliminates the risk of GVHD following infusion [82]. Moreover, The NK-92 cell line exhibits remarkable cryopreservation tolerance, demonstrating high post-thaw viability and functional retention [83]. However, they are genetically abnormal and require radiation before use, limiting their survival in patients to just 48 h. In addition, they lack a key molecule (CD16) needed for ADCC [80]. It’s important to note that only NK-92 cells are currently approved for use in clinical trials, making them a valuable source for manufacturing CAR-NK therapies [84].

Unlike traditional immunotherapies that require personalized and time-consuming engineering of immune cells for each patient, “off-the-shelf” CAR-NK cells offer a game-changer. These pre-engineered cells are readily available for immediate use, eliminating the lengthy and expensive customization process. This approach boasts several advantages: faster treatment initiation, overcoming challenges associated with obtaining high-quality cells from individual patients, and a standardized manufacturing process that ensures consistent and predictable treatment effectiveness [85]. Allogenic UCAR cell therapy offers a promising alternative for patients with insufficient immune cells. Unlike autologous CAR cells derived from the patient themselves, UCARs are engineered from healthy donor immune cells. However, to prevent GVHD due to mismatched immune systems, UCARs require additional gene editing. While convenient as an off-the-shelf product, there are limitations. Extensive gene editing raises the risk of mutations and requires advanced technical expertise. Additionally, clinical trials have shown limited persistence and expansion of allogeneic UCAR cells within patients. This challenge could be overcome by finding ways to reduce the recipient’s immune rejection response or by minimizing the immunogenicity of the infused cells [86]. Therefore, Both autologous and allogeneic NK cell transfers struggle with low numbers of infused cells, requiring optimization protocols to increase cell population size for clinical use [87].

Another source, hPSCs, encompassing both human hiPSCs and hESCs, possess the unique capabilities of self-renewal and differentiation into various cell types [88]. iPSCs offer another promising approach for off-the-shelf CAR-NK cell therapy which facilitates the ex vivo expansion of NK cells by over 1,000-fold, achieving a final cell product with a purity exceeding 90% [89]. These cells, created by reprogramming adult cells, have the potential to become an unlimited source of CAR therapy due to their ability to multiply endlessly [90]. Originally developed by Takahashi using Sox2, Oct3/4, c-Myc, and Klf4 [91], iPSCs share similar properties with embryonic stem cells, including indefinite self-renewal and the ability to differentiate into various cell types. This technology offers a convenient way to obtain pluripotent stem cells without using embryos [92]. The process of generating iPSC-CAR-NK cells involves modifying the iPSCs in their undifferentiated state before guiding them to become mature and homogenous populations of CD45⁺CD56⁺ NK cells through a two-step differentiation process. First, they are stimulated by vascular endothelial growth factor, stem cell factor, and bone morphogenic protein 4 to differentiate into HSCs. Then, these stem cells are further differentiated into NK cells using stem cell factor, FLT3L, IL-3, IL-7, and IL-15 [65, 93]. Finally, the differentiated iPSC-CAR-NK cells are expanded in large numbers for clinical use by co-culturing with artificial APC (K562 and aAPC cells expressing mbIL15/mbIL21 + 41BBL) [94]. Studies show that Phenotypically, the iPSC-derived NK cells express specific markers, including NKG2D, CD56, CD16, NKG2A, NKG2C, and KIR which are also found on natural NK cells and may even exhibit stronger cytotoxicity effects than primary NK cells [95, 96]. Additionally, they appear to be more efficient at expressing CAR structures, making them a promising avenue for developing off-the-shelf CAR-NK cell therapy [93]. Despite the potential of iPSC-derived CAR-NK cells, several crucial issues necessitate cautious evaluation before widespread clinical application. First, heterogeneity among different iPSC lines presents a challenge. Variations in differentiation capacity can lead to inconsistencies in the final cell product. Second, the robust proliferative capacity and pluripotency of iPSCs raise safety concerns. Genetic variations within these cells have the potential to be amplified during differentiation and expansion, potentially compromising the stability and safety of the cell therapy product and even increasing the risk of oncogenicity. Furthermore, iPSCs may exhibit residual DNA methylation patterns reflecting their original somatic cell source. This phenomenon, termed “epigenetic memory,” could potentially influence their differentiation trajectory, favoring the development of cell lineages distinct from those of the donor cells. Finally, the immunogenicity of CAR-NK cells derived from iPSCs is a concern. These cells may be recognized and eliminated by the recipient’s immune system, potentially reducing their therapeutic efficacy [69, 97, 98]. While amenable to genetic engineering for enhanced function, iPSC-derived NK cells exhibit a diminished ADCC response due to low expression of the critical Fc receptor CD16. This limitation can be potentially addressed through the integration of a high-affinity CD16 variant, restoring the efficacy of the ADCC mechanism [99].

When CARS comes to NK cells produced from the differentiation of stem cells, there are two methods to consider. One option is to introduce CARs to the stem cells first and then differentiate them into CAR-expressing NK cells. The other approach involves introducing CARs into the already differentiated NK cells, following a process similar to that seen in primary NK cells or NK cell lines. The challenge lies in the fact that NK cells are diverse and resistant to direct transduction, transfection, or nucleofection in vitro. This difficulty could lead to high costs associated with obtaining CAR-NK cells. NK cells obtained from stem cell differentiation, on the other hand, are more uniform and exhibit superior transduction efficiency. Nonetheless, it is crucial to verify whether the integrated CAR has any impact on NK cell differentiation, a consideration that does not arise when engineering NK cells directly [100].

A new approach for off-the-shelf CAR therapy involves generating the CAR molecules directly inside a patient’s body. This approach uses specially designed vectors to deliver the CAR gene precisely to specific immune cells. Ideal vectors for this purpose must be skilled delivery agents. They need to be highly specific, targeting only the desired immune cells and avoiding unintended changes elsewhere in the body. Additionally, they must efficiently deliver the CAR gene and promote either long-term or short-term expression of the protein depending on the treatment plan. Finally, practicality and safety are critical considerations. The vector production process should be readily scalable for large-scale production, and the final formulation must be stable for long-term storage as an off-the-shelf treatment, with affordability ensuring broad patient access. Notably, safety remains a key concern, and the vectors themselves must possess a well-established safety profile [69, 101].

CAR-NK cell engineering utilizes various gene transduction methods, each with its own advantages and limitations. Retroviral and lentiviral vectors offer efficient delivery but raise concerns about genotoxicity and reduced NK cell viability. Electroporation and liposomal transfection provide a faster and less variable approach, but the introduced genes are short-lived. DNA transposons offer a promising alternative with stable integration, low immunogenicity, and affordability, but face hurdles related to low efficiency and potential cell death during the DNA delivery process (Fig. 2) [102, 103].

Fig. 2
figure 2

CAR-NK cells production: This figure summarizes the diverse approaches to CAR-NK cell production. The traditional ex vivo method involves isolating NK cells from sources like peripheral blood (PBMCs), umbilical cord blood (UCB), or NK cell lines, followed by activation, gene modification with CAR vectors, expansion, cryopreservation, and administration. While readily available, PBMCs have limitations in NK cell quantity and function. UCB offers younger and more proliferative NK cells, but concerns remain about their in vitro activity and differentiation. Cell lines provide rapid growth but have limitations related to genetic abnormalities and pre-treatment needs. Additionally, human pluripotent stem cells (hPSCs) including induced pluripotent stem cells (iPSCs) are emerging as a promising source due to their self-renewal and potential for generating large numbers of homogenous and highly cytotoxic CAR-NK cells. The figure also highlights a novel in vivo approach that delivers CAR genes directly into a patient’s body, bypassing the need for ex vivo manipulation. This comprehensive overview emphasizes the importance of cell source selection and the potential of in vivo gene delivery for future advancements in CAR-NK cell therapy

Since CAR-T cells are living cell therapy, strict quality checks are essential throughout production to ensure a safe and effective final product. This involves controlling the production materials (specifically cells and gene editing vectors), ongoing monitoring and testing during production, final product release testing, validating the entire manufacturing process, and performing stability studies. These studies assess factors like CAR expression levels, lymphocyte types present, cell purity, cell viability, and ratios, effectiveness in lab tests (in vitro potency), and contamination control (including sterility testing, mycoplasma checks, checks for replication-competent viruses, rapid detection of microbes, endotoxin testing, and others.) [59, 104,105,106].

Preclinical and clinical study on CAR therapy in asthma

Current asthma treatments, while effective for many, have limitations. Despite managing symptoms in a majority of patients, some remain resistant to medications like anti-inflammatory drugs and bronchodilators [22]. Additionally, long-term use of inhaled corticosteroids can lead to undesirable side effects such as high blood pressure and osteoporosis [107]. This lack of a definitive cure, particularly for severe asthma, necessitates the exploration of alternative treatment avenues.

IgE-mediated allergic asthma, affecting over half of all allergic asthmatics, poses a significant health burden with uncontrolled symptoms despite available medications. A survey among asthmatic patients showed that 75% of them prefer non-drug treatments for their asthma. Therefore, a treatment method that has a longer and more stable effect is desirable. In recent years, the use of cell therapy based on CAR therapy has had many successful results in the field of cancer and other diseases. It seems that the use of this therapeutic method in the treatment of asthma and AHR can be welcomed by everyone [18].

The presence of mIgE enables T Cells to target and eliminate cells involved in IgE production, including germinal center B cells, plasmablasts, plasma cells, and memory B cells [108, 109]. Achieving high specificity for FcεRIα-based CARs requires meeting four key criteria. First, precise mIgE binding strength for T cell activation and target cell elimination [110]. Second, efficient discrimination against healthy FcεRI-expressing cells to avoid side effects [111, 112]. Third, distinction between FcεRI-bound and FcεRII-bound IgE for specificity and to prevent non-specific immune activation, and Finally, high-affinity binding to membrane-anchored mIgE while minimizing competition from circulating IgE [113, 114]. To address this, a study using low-affinity FcεRIα-based CARs was conducted, demonstrating their ability to induce selective T cell responses against cells expressing transmembrane IgE but not free IgE or FcεRI/FcεRII-bound IgE. These CARs possess the specificity and potency required for targeting IgE-expressing B cells in ACT for asthma and allergic diseases. Adejuwon [18] constructed FcεRIα-based CAR-T cells of both the 2nd and 3rd generation. These CARs were then introduced into primary human CD8 + T cells via lentiviral transduction, paving the way for in vitro assessment of their longevity and ability to selectively destroy IgE-expressing B cells. Second generation CARs effectively eliminated target cells.

Driven by the goal of developing a single-dose therapeutic approach for IgE suppression in allergic disease, Christie et al. constructed CARs targeting the EMPD of mIgE. This specific binding domain exhibits exclusive expression on all IgE-producing B cell populations, potentially enabling long-lasting IgE suppression with a single administration. EMPD’s exclusive expression on IgE + B cells allows CARs to target and eliminate them while avoiding competition from circulating free IgE due to EMPD’s absence on secreted forms. One particular CAR, 2E3E10, exhibited remarkable effectiveness against both U266 and Daudi cells expressing mIgE, triggering both potent cytotoxicity and IFN-γ production. This was particularly noteworthy given the relatively low expression level of mIgE on U266 cells. The results of this study demonstrate the feasibility of using EMPD-specific CARs to reprogram T cells to target and eliminate mIgE-expressing B cells, suggesting a potential therapeutic strategy for allergic diseases such as asthma [19].

SEA, a significant phenotype of refractory asthma,, is driven by eosinophilic inflammation [21]. Targeting eosinophils and related cytokines(IL-4,IL-5 and IL-13) shows promise, but long-term use limits effectiveness and affordability [115]. A recent article by Jin et al. developed a promising approach for asthma cure using long-lasting CAR-T cells targeting IL-5Rα + eosinophils [20]. IL-5Rα is highly expressed on the surface of mature eosinophils and progenitors. By binding IL-5Rα to IL-5, it causes the release of various cytokines and intensification of inflammation [116]. These CAR-T cells, dubbed Immortal-like and Functional IL-5 CAR-T cells, demonstrate remarkable persistence and efficacy in suppressing asthma symptoms. They engineered IL-5 CAR-T to produce an IL-4 mutein, a modified version of IL-4 that effectively blocks the signaling of both IL-4 and IL-13. In multiple models of asthma, a single administration of IL-5 CAR-T cells in immunocompetent mice, without any prior conditioning, effectively eliminated pathological eosinophils and blocked the actions of IL-4/IL-13. This resulted in long-lasting suppression of type 2 inflammation and asthmatic symptoms. In line with this study, Chen et al. developed a CCAR-T utilizing IL-5-CD28-CD3ζ receptor. In this work, an IL-5-based CCAR configuration was created that does not rely on scFv for binding to the target. This system harnesses T cells to selectively eliminate eosinophils. IL-5 CCAR-T cells effectively neutralized eosinophils in vitro and mouse asthma models (the HDM and OVA-stimulated. Additionally, these IL-5-anchored CCAR-T cells had a lasting impact on reducing eosinophils, and IL-5 levels, and preventing airway inflammation for three months, surpassing the typical active therapeutic period of single mAb-injections which is four weeks [21].

CD4 + Tregs balance allergic airway inflammation [117]. Skuljec et al. used CARs to redirect Tregs to lung epithelium in asthmatic mice. CAR-Tregs accumulated in lungs and lymph nodes, reducing AHR and eosinophils. They also inhibited IL-5 production, mucus production, and Th2 cytokine levels, while increasing TGF-β1 and IL-10 expression. In comparison to unmodified Treg cells, CAR-modified Tregs demonstrated remarkable superiority in managing asthma, highlighting the importance of specific Treg cell activation in the affected organ [22].

According to www.clinicaltrials.gov accessed on 28 April 2024, there are currently no ongoing clinical trials in asthma to test the effectiveness of CAR-based therapy—preclinical studies on the applications of CARs in Asthma are summarized in Table 2.

Fungal asthma, a specific type of asthma, is primarily caused by AF. This fungus triggers the release of IL-25 and IL-33 cytokines from respiratory epithelial cells through the action of various proteases. These cytokines, in turn, stimulate eosinophils and contribute to AHR, leading to inflammation in the airways [118, 119]. Dectin-1, a type C lectin receptor on immune cells like lymphocytes, macrophages, and bronchial epithelial cells, strengthens immune responses by recognizing beta-glucans on the surface of AF [120, 121]. Dectin-1 CAR T cells show promise for fungal airway inflammation treatment. This strategy has the potential to be more targeted and have fewer side effects compared to traditional drug therapies. . Seif et al.‘s engineered CD8 + Af-CAR-T cells with dectin-1 protected mice from lung damage, preserved alveolar structure and reduced necrosis around the bronchi, suggesting promise for future asthma therapies [122]. Kumarasan et al. developed a Dectin-CAR that specifically recognizes laminarin, a component of AF, and restricts fungal growth. This CAR also significantly enhances IFN-γ production by over 6-fold [123]. Targeting Dectin-1 with CAR-NK cells holds promise for treating asthma, but needs more optimization, human trials, and safety evaluation.

Table 2 Summary of preclinical studies on the applications of CARs in Asthma

NK cell, subtypes, activation, and function mechanisms

Unique among innate lymphoid cells, NK cells play a vital role in immunoregulation and cytotoxicity within the immune system [17]. NK cells develop from HSCs in the bone marrow through a series of differentiation steps [124]. NK cells present in the blood, lymph nodes, and tissues and classified in two main subpopulations based on the levels of CD56 and CD16: CD56brightCD16- and CD56dimCD16+. These cells lack CD3 expression [125].

CD56brightCD16⁻ NK cells are a distinct subset of NK cells found predominantly in secondary lymphoid organs. Though lacking expression of KIRs, they exhibit potent immunomodulatory functions by producing a diverse array of cytokines, including IFN-γ, IL-10, and IL-2. In contrast, CD56dimCD16 + NK is highly cytotoxic and induces ADCC. They exhibit abundant expression of perforin and granzyme B, cytotoxic molecules crucial for their cytolytic function. Furthermore, KIRs expressed on NK cells play a central role in self- and non-self-recognition, contributing to their precise targeting of infected or transformed cells. This group comprises 90% of circulating NK cells and is also found in the spleen and lungs. Compared to other tissues, the lungs harbor a high proportion of NK cells, particularly the CD56dimCD16 + subset which circulate between the lungs and blood [126, 127]. CD56dimCD16 + NK cells are typically associated with rapid responses in acute inflammation, and CD56brightCD16- NK cells can contribute to immune regulation in chronic settings. Additionally, CD56dimCD16 + NK cells demonstrate an increased capacity to produce cytokines when encountering target cells. Despite significant progress in characterizing NK cell subsets, the precise boundaries between them, are still being elucidated [128, 129].

NK cells act as early responders in the immune system, playing a critical role in defending against viral infections and fighting against cancer. NK cells are a key part of the immune system’s defense against cancer, particularly hematopoietic tumors. They can eliminate abnormal cells without prior sensitization, unlike other immune cells. Unlike T cell-based therapies, which have shown limited success, NK cells offer a promising alternative due to their inherent ability to directly kill cancer cells through various mechanisms and promote anti-tumor immune response [130]. They have a similar function to CD8 + T cells. They augment the immune system by stimulating other immune cells [131, 132]. NK cells constantly integrate signals from numerous surface receptors (Fig. 3). These receptors act as a finely tuned switchboard, sending both inhibitory and activating signals that determine the cell’s response. This dynamic interplay allows NK cells to rapidly adapt and respond to environmental changes [133]. Most of the inhibitory receptors on NK cells recognize MHC class I molecules. These include two main groups: KIRs with inhibitory motifs and the CD94-NKG2A heterodimer. PD-1, TIGIT, LAG-3, and CD96 are other inhibitory receptors [134, 135]. Many activating receptors exist for NK cells which NCRs (NKp46, NKp44, NKp30, etc.) and CD16 can be mentioned in this list [10]. MICA and MICB, molecules related to MHC class I proteins, activate NK cells through the NKG2D receptor which is one of several factors influencing their activity. These interactions play a crucial role in recognizing and eliminating inflammated cells. MICA and MICB are present on the surface of virus-infected and stressed cells as well as in soluble forms [136]. Furthermore, the NKG2D receptor initiates NK cell activation utilizing the adapter molecule DAP10 [137]. Conversely, upon binding to the appropriate ligands in NK cells, NCRs enlist adapter molecules DAP12, CD3ζ, or FcεRIγ which carry the ITAM. These adapter molecules trigger the commencement of activation signaling cascades. CD16 is unique in its ability to trigger some responses in resting NK cells without additional signals. However, it primarily activates them in collaboration with other receptors. The receptor DNAM-1, a member of the Immunoglobulin superfamily, has the capability to stimulate NK cells through its interaction with CD112 [137]. Furthermore, IL-15, IL-12, IFN type 1, and DCs are considered very strong NK cell activators [138].

Fig. 3
figure 3

NK cells receptors: NK cells rely on a combination of activating and inhibitory receptors to determine their actions. Additionally, receptors specific to interleukins (signaling molecules) allow NK cells to respond to interleukins’ effects. Finally, receptors for chemotactic and adhesive factors guide NK cell movement and attachment, further influencing their activities

NK cells produce appropriate immune responses through producing chemokines such as chemokine CCL 3, CCL 5, and CXCL 8 and cytokines such as TNF-α, IFN-γ, GM-CSF, IL-5, 13, 10 or interacting with cells such as macrophages, T cells, B cells, granulocytes, and DCs [126, 139,140,141]. IFN-γ is the main cytokine of NK cells, which is produced through stimulation of cytokines such as IL-12 and IL-18 and plays a significant role in treating asthma [142].

Recent studies clarify that Th2-polarizing DCs and immature DC cells are killed by NK cells through helper molecule DNAX 1 (DNAM-1) and NKp30. As a result, the number of DCs involved in Th2 immune responses is reduced [143]. Furthermore, the combined action of both NK cell subsets exerts its cytotoxicity on DCs by way of direct contact with eosinophils and leads to their apoptosis through a different pathway from caspases and degranulation of NK cells [12, 144, 145]. Additionally, In the presence of lipoxin A4, NK cells dial down the production of superoxide anions, key players in inflammatory processes [145].

In the lung, NK cells can trigger apoptosis in neutrophils, promoting the resolution of acute inflammation. This process is mediated by the caspase pathway, involving the Fas and NKp46 receptors on neutrophils and NK cells [146]. While, neutrophils play an important role in the maturation of NK cells [147].

NK cells activate plasmacytoid DCs through NKp46, exerting anti-inflammatory functions. In mice, lung plasmacytoid DCs suppress T-cell division and effector T-cell generation induced by myeloid DCs, thereby regulating lung inflammation [126].

Research suggests decreased activity of lung NK cells compared to their counterparts in PB. This adaptation might serve a crucial purpose. Given the lung’s constant exposure to allergens, the subdued NK cell response could help prevent excessive inflammation and tissue damage [148].

While the number of IL-4 producer -NK cells increases in allergic asthma the number of NK cells producing IFN-γ decreases. In contrast, in neutrophilic asthma, the level of IFN-γ and granzyme B secreted from NK cells is increased [126]. Moreover, during severe asthma, the ratio of NK cells to CD4 + T cells within the lungs is reduced. NK cell numbers in the bloodstream are also lower. In addition, findings reveal the impaired ability of NK cells to trigger eosinophil cell death in patients with severe asthma [12, 13]. The research discussed above, unveils a critical dysfunction in asthma–impaired NK cells. These immune cells are not only reduced in number but also show abnormal reactions to allergens, potentially hindering the fight against asthma. The reduced NK cell activity highlights the potential of drug or cell therapies to improve asthmatic patients’ immune response by increasing NK cell populations and enhancing their abilities.

Unveiling the difference: A comparative analysis of Treg and NK cell

NK cells and Treg cells play crucial roles in immune responses to allergens, interacting with APCs to orchestrate the response. While NK cells make up a significant 5–20% of circulating lymphocytes, Treg cells constitute less than 5% [149].

NK cells originate from lymphoid progenitor cells within the bone marrow and mature in secondary lymphoid organs. Subsequently, they populate diverse tissues such as PB, lung, liver, skin, bone marrow, and kidney. In contrast, Treg cells expand in the thymus and PB and express TCR [150, 151]. Compared to Tregs, the human lung harbors a significantly higher amount of NK cells, which is one of the body’s primary reservoirs for this critical immune cell population [152].

NK cells comprise two main groups differentiated by CD56 expression: CD56dim and CD56bright. The dominant population, CD56dim NK cells, circulate within the blood and exhibit robust cytotoxic activity, directly eliminating infected or tumor cells. In contrast, CD56bright NK cells, residing primarily in lymphoid tissues, contribute to immune defense by secreting cytokines and chemokines. According to the direct killing function of NK cells, in inflammatory or infectious reactions, all these cells perform killing while only some T cells act as cytolytic [153, 154]. Treg cells, as a subset of CD4 + T cells, have high expression of the IL-2 receptor α chain (CD25) [155]. CD4 + CD25 + Tregs can be further classified into two main subsets based on their origin: thymus-derived Tregs and peripherally-derived Tregs. Studies demonstrated that Treg cells derived from the periphery, display the ability to regulate the immune response to foreign antigens [156]. Additionally, NK cells differ from Treg cells in the absence of CD3 [157].

While both NK cells and T cells utilize MHCs on APCs for recognition, NK cells exhibit greater flexibility. This flexibility stems from their ability to recognize receptors directly on target cells through non-MHC-dependent mechanisms [140, 158]. Unlike Treg cells, which recognize antigens with rearranged receptors in a clonal form, NK cells express random combinations of active and inhibitory receptors to recognize antigens by integrating them [151]. In contrast to Tregs, which primarily utilize the αβTCR and TLRs for recognition, NK cells possess a diverse cluster of activating (e.g., NKp44, NKp30, NKp46) and inhibitory receptors (e.g., NKG2A, KIR3DL1, KIR2DL1, KIR3DL2) [155].

In general, NK cells have a broader range of potential targets through the recognition of diverse antigens. However, their recognition lacks the precision and specificity achieved by Treg cells. Compared to T cells, whose population can double within 10 h upon activation, NK cells exhibit slower proliferation, requiring approximately 1.5 days to achieve the same feat [159, 160]. NK cells generally have a shorter lifespan and rely on immediate responses. On the contrary, T cells can form specialized memory cells which persist for long-term immune protection [160, 161].

Both cells utilize perforin granzymes to induce target cell death. Additionally, they express apoptotic ligands such as FASL. However, unlike phagocytic cells, neither NK cells nor Treg cells digest debris from eliminated cells [155]. Distinct from T cells, NK cells can kill by natural mechanisms and ADCC [162].

NK cells just like Treg cells can produce anti-inflammatory cytokines including IL-10 and IFN-γ [152]. Studies have shown that Treg cells suppress immune response and promote tolerance to foreign antigens by secreting IL-10 and TGF-β [163]. Among the types of NK cells, this function is performed by CD56dimCD16+, which secretes fewer cytokines than T cells [129, 164]. Low cytokine secretion can counteract the harmful effects of TGF-β in asthmatic patients.

In addition, unlike T cells, these cells are easy to transduce [88]. As the last comparison, GVHD is rarely reported in treatment with NK cells. This may be partially explained by the reduced proliferative and cytokine secretion capacity of NK cells compared to T cells. A study by Ruggeri et al. using mouse models confirms this finding, demonstrating that NK cells can eliminate APCs, thereby exerting a protective effect against GVHD [165] (Table 3). However, other factors such as differences in target cell recognition and immune regulation require further investigation [166].

Table 3 A comparative analysis of Treg and NK cell

The strength and challenges of NK cells as immunotherapy candidates

In the previous chapter, we discussed certain significant characteristics of NK cells. In this section, we will delve into the details of utilizing these features in CAR-NK, as well as the challenges and prospects that lie ahead.

NCR

NCRs are one of the activating receptors on the surface of NK cells which induce NK cells to kill targets and produce cytokines [167]. The NCR family comprises NKp30 (CD337), NKp44(CD336), and NKp46 (CD335), are determined by the genetic sequences NCR3, NCR2, and NCR1, respectively [168]. A comparison of the different NCRs comprises is presented in Table 4 NK cells with high NCR surface density exhibit stronger cytotoxicity against target cells [169].

NKp30 plays a multifaceted role in the immune response. Encoded by the NCR3 gene, this single-unit transmembrane protein with an immunoglobulin-like domain exists in three forms (a, b, and c) due to alternative splicing, each with distinct functions [170]. Found on both resting and activated NK cells, NKp30 goes beyond just targeting infected or cancerous cells [171]. It influences the immune response by regulating cytokine production. NKp30a and NKp30b promote the release of immunostimulatory Th1 cytokines like IFN-γ and TNF-α, while NKp30c acts as an immunoregulator by inducing IL-10 production [172,173,174].

NKp44, another member of the NCR family, shares a similar structure with a single external immunoglobulin-like domain but differs in its expression pattern [168]. Unlike NKp30 and NKp46 which are present on both resting and activated NK cells, NKp44 expression is typically induced only after activation [171]. This controlled expression helps maintain a balanced immune response.

While NKp30 and NKp44 are single-unit transmembrane proteins with one extracellular domain, NKp46 is unique with two such domains [168]. Interestingly, NKp46 is present on both resting and activated NK cells, allowing for quicker responses [171]. To trigger NK cell activation, NCRs rely on adaptor molecules containing an ITAM signaling. NKp46 and NKp30 partner with CD3ζ and FcRγ, while NKp44 utilizes DAP-12 for signal transmission [175]. Importantly, NKp46 and NKp30 have the potential to deliver stronger activating signals due to their adaptor complexes containing more ITAMs compared to NKp44 [176].

NK cells in severe asthma show lower NCR expression, particularly NKp30 and NKp46 [177]. Linking NCRs to weaker anti-inflammatory function. NCR1 may regulate allergic responses by reducing airway inflammation [178]. Additionally, Studies suggest NCRs might play a role in resolving acute inflammation. Human NK cells can induce neutrophil apoptosis through the NKp46 and Fas pathway in a laboratory setting [146]. This mechanism could potentially help reduce asthma. Elhaik Goldman et al. link NCR1 to reduced allergic lung inflammation [179]. NCR1-deficient mice showed worse symptoms, suggesting it dampens allergies by controlling granulocyte infiltration into the airways and IgE production.NCRs’ potent immune activation potential makes them promising for CAR-T therapy. Studies by Divanji et al. showed enhanced cancer cell cytotoxicity with CAR-T cells expressing NKp30, NKp44, NKp46 [131]. Interestingly, subsequent research by the same group revealed that even though NKp44 and NKp46 CARs lack specific signaling domains, they can still interact with and activate their natural adaptor molecules within NK cells [180]. Kasahara et al. explored incorporating NKp44’s antigen-binding domain into CAR design for broad target recognition in CAR-T cells [181]. This highlights NCRs’ potential in CAR therapy, but further research is needed for CAR-NK applications in diseases like asthma.

Dap 10, Dap 12, and NKG2D

NKG2D is a critical activating receptor in NK cells which has a short cytoplasmic tail that lacks the ability to initiate intracellular signaling [182, 183]. NKG2D, located on the cell membrane, requires adaptor proteins for stability and signal transduction [184]. These adaptor proteins, such as Dap10 and Dap12, trigger distinct signaling pathways. DAP10 utilizes a YxxM motif to activate PI3K [185, 186], whereas DAP12 employs an ITAM motif to recruit and activate ZAP70-Syk, ultimately leading to cytokine release and cell killing [187, 188].

As some studies show promising effects of Dap10 and Dap 12 such as improved persistence, cytokine secretion, and cytotoxicity in CAR-T therapy [189,190,191], some others show their encouraging effects in CAR-NK therapy [183, 192]. Xiao et al. conducted a study on NK cells using a novel CAR with NKG2D and DAP12 components. The study demonstrated that NK cells with DAP12-containing NKG2D-CAR had higher cytotoxicity and IFN-γ release compared to NK cells with CD3ζ-containing NKG2D-CAR [183]. The incorporation of DAP12 signaling in CAR designs has been associated with a triad of adverse consequences: NK cell exhaustion, hyperactivation, and CRS. These limitations necessitate the investigation of alternative co-stimulatory domains, such as DAP10, for safer and more effective CAR-NK cell therapies [193]. NKG2D-DAP10 increases NK cytotoxicity according to Wilton et al.‘s research [192]. In addition, pent et al. employed DAP10 and DAP12 in the development of CAR-NK cells, utilizing them as the principal intracellular domains for the transmission of signals from NK stimulatory receptors. As a result, DAP10,12 CAR-NK cells may exhibit increased lifespan, enhanced cytotoxic, and suppress allergic responses through IFN-γ production.

IL-10

NK cells are a source of IL-10, but this anti-inflammatory cytokine is also generated by various immune cell types such as TH1, TH2, TH17, Teff cells, FoxP3 − and FoxP3 + Treg cells, B cells, and myeloid cell subsets [194, 195]. IL-10 exerts its effects through interaction with a specific receptor complex composed of IL-10Rα and IL-10Rβ subunits. Ligand binding triggers the Jak/STAT pathway, with Jak1 and Tyk2 kinases phosphorylating STAT3 [196, 197]. Although STAT3 is essential for this process, research suggests that STATE1 and STATE5 also contribute to IL-10 signaling [198]. Nuclear translocation of phosphorylated STAT3 subsequently induces the expression of anti-inflammatory genes, including SOCS-3 [199]. SOCS-3 acts as a negative regulator, inhibiting the production of pro-inflammatory cytokines and antigen presentation by downregulating key signaling pathways like NF-κB and MAPKs [200]. Additionally, IL-10 can activate the p38 MAPK pathway, leading to the induction of HO-1, a protein with further anti-inflammatory properties. In conclusion, IL-10 employs a multifaceted signaling network involving the Jak/STAT pathway and HO-1 induction to suppress inflammation and maintain immune homeostasis [199].

IL-10 is a key anti-inflammatory cytokine in asthma, reducing lung damage and disease severity [201, 202]. TH cell-derived IL-10 plays a crucial role in establishing tolerance to allergens and resolving allergic inflammation, with impaired production linked to severe asthma [203, 204]. Studies show IL-10 limits TH2 cells, reduces eosinophilic inflammation, and mitigates AHR. Genetic variations in IL-10 expression are linked to asthma risk and severity [195]. IL-10 also exerts a regulatory influence on Th2 lymphocyte production of IL-4 and IL-5, thereby modulating IgE-mediated mast cell activation. Animal studies further corroborate these anti-inflammatory effects [205].

Beyond TH cells, recent research highlights the immunomodulatory role of IL-10 from other immune cell populations. Treg Foxp3 cells contribute to suppressing allergic lung inflammation through IL-10 production [206]. Additionally, IL-10-producing ILC2s promote epithelial barrier integrity by suppressing pro-inflammatory mediators like IL-6 and IL-8, thereby mitigating tissue damage during allergic reactions [207]. Myelomonocytic cells exposed to IL-10 upregulate IL-1 receptor antagonists, leading to diminished eosinophil recruitment and reduced AHR [208]. However, despite these compelling findings, a comprehensive understanding of the precise mechanisms by which IL-10 regulates the diverse phenotypes of asthma remains elusive [202].

Building on the promise of CAR-T cell therapy, researchers are exploring the potential of incorporating IL-10 to enhance their efficacy and broaden their application. Mohseni et al. engineered HLA-A2 CAR-Tregs to express IL-10, boosting their suppressive ability [198]. This finding aligns with Huang et al.‘s work, where CAR-T cells engineered for IL-10 secretion exhibited improved proliferation and effector function [209]. Expanding on this concept, Chen et al. pioneered the use of IL-10R as a therapeutic target in AML by engineering CAR-T cells to recognize AML cells expressing IL-10R as the antigen-binding domain. The study observed an increase in Treg cells and strongly supported the therapeutic potential of incorporating IL-10 into CAR-T cell therapy [210]. Overall, in asthma, CAR-NK cells could offer a novel anti-inflammatory approach by leveraging their ability to produce IL-10 (Fig. 4).

KIRs

KIRs contribute to the regulation of NK cell activity. These highly polymorphic molecules are expressed on the surface of CD56dimCD16 + NK cells and transduce signals that modulate cytotoxic function [126, 127, 211]. Two types of KIR are inhibitory and activating receptors. Inhibitory and activating KIRs interact with HLA-I molecules to regulate NK cell responses [212].

Inhibitory KIRs expressed by NK cells suppress cytotoxicity through binding of NK cells and MHC-1 on healthy cells [213]. One of the mechanisms to enhance the cytotoxic activity of CAR-NK is silencing inhibitory KIRs. KIR-blocking antibodies are one of the approaches to silence inhibitory KIRs [214].

Unlike most KIR receptors with clear-cut inhibitory or activating functions, KIR2DL4 presents a unique case. While it possesses structural features associated with both types, only its activating function has been definitively established [215, 216]. KIR2DL4 (CD158d), expressed on the surface of NK cells recognizes HLA-G as its ligand [215, 217]. HLA-G polymorphisms are associated with increased susceptibility to AHR and asthma development [218]. Interestingly, research suggests HLA-G expression can increase in various tissues during inflammation [219]. This includes the bronchial epithelium (lining) of asthmatics’ lungs [218, 220]. Recent research suggests a more nuanced understanding of HLA-G function. This raises the intriguing possibility of an analogous interaction between NK cells and HLA-G in the lung microenvironment, potentially influencing asthma development.

KIR2DL4 exhibits a unique genetic polymorphism. Two common variants exist within the transmembrane region, with the 9 A-TM allele containing a single nucleotide deletion in exon 6. This deletion results in a defective receptor incapable of inducing IFN-γ secretion upon ligand binding [216]. IFN-γ variations may impact asthma risk. Firstly by affecting Th1/Th2 balance, as previously reviewed by Szabo et al. [221], and secondly, through NK cell-derived IFN-γ in tolerogenic DC generation [222]. This suggests a more complex role for KIR2DL4 in allergy.

When KIR2DL4 binds to HLA-G, the ITIM motif within KIR2DL4 recruits phosphatases like SHP-1 and SHP-2, leading to the subsequent inhibition of CD16/FcγRIIIa signaling in human NK cells [223]. This CD16 inhibition aligns with the self-missing hypothesis, as CD16 is involved in ADCC. However, KIR2DL4 activation can also trigger the release of pro-inflammatory cytokines like TNF-α, IL-1α, IL-1β, IL-6, and IL-8 from NK cells, highlighting its potential role in promoting inflammatory responses [224, 225]. This activating signal appears to be mediated by FcεRIγ and is independent of the ITIM motif [224, 226, 227]. Soluble HLA-G induces similar cytokine secretion, possibly via endosomal KIR2DL4 binding and a DNA-dependent protein kinase subunit, AKT, and NF-κB [225, 228].

Activating KIRs interact non-covalently with ITAM-containing DAP-12. When the activating KIR binds to DAP-12, it triggers a chain reaction within the NK cell. This reaction involves the phosphorylation of various molecules downstream of the initial interaction, ultimately leading to NK cell activation [229, 230]. Researchers are exploring further enhancements to CAR-NK cells by leveraging activating KIRs. These approaches include stimulating them using mAb or specialized molecules called NK Cell Engagers [231]. However, a major hurdle lies in the high degree of similarity (97–98%) between the external regions of activating and inhibitory KIRs [230]. Designing protein tools for specific targeting of activating KIRs is challenging. A promising strategy uses peptide-MHC complexes recognized by activating KIR2DS2 [232]. Studies have shown that introducing these complexes through a DNA vaccine approach can be effective in activating CAR-NK cells in mice engineered to express specific KIRs [233]. Interestingly, allelic variations in KIR2DS2 itself don’t affect its ability to recognize specific peptide-MHC complexes [232]. This suggests a single complex can activate NK cells regardless of the KIR2DS2 isoform they express. Since all isoforms respond, targeting specific isoforms for therapy offers no additional benefit.

In conclusion, KIRs, particularly KIR2DL4, play a complex and multifaceted role in regulating NK cell activity and potentially influencing asthma development. Silencing inhibitory KIRs may enhance CAR-NK cells, but KIR2DL4-HLA-G interaction is complex. Activating KIRs, like KIR2DS2, hold promise for improved CAR-NK therapy.Future investigations in humans are necessary to determine the effectiveness of this approach and its potential impact on improving CAR-NK cell-based treatments for asthma and other diseases.

NKG2A

NKG2A (CD159a) plays a critical role in regulating the activity of NK cells, acting as an inhibitory immune checkpoint receptor. It recognizes HLA-class I and E molecules on healthy cells, preventing NK cells from attacking them and thereby preventing autoimmune reactions [234]. This recognition system allows NK cells to distinguish “self” from “non-self” cells within the body [235]. NKG2A overexpression leads to NK cell exhaustion (dysfunction) [235, 236]. Blocking NKG2A might restore NK cell activity [237]. High NKG2A suppresses NK cells. This effect is likely mediated by the repression of STAT1, NF-κB, and p38 MAPK signaling pathways, which are thought to be key mechanisms in NK cell activation [238].

NKG2A has a multifaceted role in regulating NK cell function. CD56bright NK cells, characterized by low KIR expression but high NKG2A levels, exhibit diminished cytotoxicity and IFN-γ production compared to CD56dim NK cells with high KIR expression and lower NKG2A [4, 239]. Furthermore, NKG2A may be involved in various immune-mediated diseases, including autoimmune diseases, inflammatory conditions, parasitic infections, and transplant rejection [240, 241].

NKG2A blockade enhances NK cell function. Studies demonstrate that blocking NKG2A with antibodies like Monalizumab can restore cytotoxicity, IFN-γ production, and ADCC in NK cells isolated from patients [242]. This finding is further supported by gene editing approaches that eliminate NKG2A, confirming its role as an inhibitory checkpoint [243]. However, the impact of NKG2A blockade appears to be context-dependent. While Grote et al. observed limited effects on CD276-CAR NK-92 cell cytotoxicity against melanoma upon NKG2A knockout or blocking it with an antibody [244], a significant increase in cytotoxicity was seen against AML cells when combined with knockouts of CBL-B and TIGIT. This suggests the potential benefit of targeting multiple inhibitory pathways for optimal efficacy [245]. Additionally, studies employing CRISPR/Cas9 to disrupt NKG2A demonstrated enhanced cytotoxic activity of CD33-CAR-NK cells [246]. Besides CRISPR-mediated CARs, Combining allogeneic CAR-NK therapy with anti-NKG2A antibodies has also been proposed to overcome NKG2A’s inhibitory role [247]. The potential for translating these findings to asthma treatment warrants further investigation, particularly considering the potential benefits of enhanced NK cell cytotoxicity and IFN-γ production.

TGF-β

TGF-β is a small signaling protein with a surprisingly large impact on human health which exists in three isoforms (TGF-β1, TGF-β2, and TGF-β3) [248]. Among immune cells, NK cells stand out alongside eosinophils and epithelial cells as significant producers of TGF-β, comparable to monocytes [249]. It acts as an immunosuppressive cytokine, dampening T, B, and NK cell activity to maintain immune balance [250]. Beyond the immune system, TGF-β arranges cellular processes by attracting repair cells (macrophages and fibroblasts), stimulating the formation of structural tissue support (fibronectin), and even influencing ASM cell growth [248]. Research further reveals TGF-β’s impact on essential biological processes, regulating cell proliferation, differentiation, and death, as well as influencing the extracellular matrix and stem cell fate, ultimately affecting development, tissue repair, and immune responses across multiple organs [251]. Excessive TGF-β activity can suppress the immune system, hindering target cell elimination and potentially worsening asthma, a disease marked by immune dysregulation [252].

TGF-β signaling, via Smad protein activation, regulates gene expression. A specific Smad3 gene variation is linked to asthma susceptibility [253, 254]. TGF-β plays a complex and multifaceted role in asthma [255]. TGF-β exhibits pro-fibrotic properties, promoting the growth of ASM cells and the deposition of extracellular matrix by fibroblasts [251]. This excessive tissue remodeling ultimately leads to structural changes that constrict the airways in asthma [248]. Biopsies and BAL fluid from asthmatic patients reveal elevated levels of TGF-β, further supporting its involvement in the disease [256]. TGF-β contributes to airway remodeling in asthma by promoting EMT, a process where airway epithelial cells transform into scar-forming fibroblasts [256]. This suggests a potential dysfunction in epithelial repair mechanisms in asthmatic patients [257, 258]. TGF-β secreted by eosinophils induces airway constriction and AHR, as well as promoting goblet cell metaplasia, excessive mucus production, tissue injury, and structural alterations in the airways [259]. In contrast, some studies suggest TGF-β might also have anti-inflammatory properties. This is relevant to asthma, where a weak immune response with low IL-10 and TGF-β production is linked to severe cases [252]. The combined action of TGF-β and IL-10 enables the regulation of pro-inflammatory cells and the mitigation of exaggerated inflammatory reactions [260]. In stark contrast to NK cells, the release of IL-10 and TGF-β by Treg cells in humans plays a pivotal role in effectively dampening airway inflammation [261]. Moreover, excessive TGF-β production by NK cells in asthma might contribute to the overall disease process [260].

In terms of considering the interaction of NK cells with TGF-β, it has been demonstrated to suppress the cytotoxic responses of NK cells. Findings were observed in a study involving human NK cells, where the introduction of TGF-β to IL-2/15-stimulated NK cells led to a decrease in the expression of IFN-γ, granzyme B. In research conducted by Trotta et al., it was found that TGF-β suppresses NK cell activation induced by CD16 binding. The authors proposed that the inhibitory impact on NK-cell ADCC responses is facilitated by Smad3, which acts downstream of TGF-β signaling through its receptor. These effects were reversed upon the inhibition or knockdown of the TGF-β receptor [262].

The potential therapeutic implications of these inhibitory findings are intriguing. Panek et al.‘s research using experimental models demonstrates that blocking specific pathways downstream of TGF-β signaling can effectively reduce airway fibrosis [248]. Additionally, research on SARS-CoV-2 infection suggests that inhibiting TGF-β might restore NK cell function, potentially offering new avenues for treatment [263]. Moreover, Co-administration of TGF-β antagonists with NK cells has demonstrated the ability to maintain their cytotoxicity and sustain the expression levels of activating NK receptors such as NKG2D and CD16. The utilization of Fresolimumab, an antibody that neutralizes TGF-β, and galunisertib, an inhibitor targeting TGF-βRI, has been highlighted for their safety and tolerability profiles in human subjects [82].

In a strategically separated investigation, CRISPR/Cas9 disrupts TGF-βR2 in NK cells [264], enhancing their resilience against target cells and resistance to TGF-β-mediated immunosuppression, leading to stronger anti-tumor effects in models of AML and glioblastoma [265].

In a comparable approach, CAR-NK cells were engineered to overexpress a dnTGF-βR, which exhibits a strong affinity for TGF-β without initiating subsequent signaling cascades. This alteration effectively alleviates the inhibitory impacts of TGF-β, consequently enhancing NK cell cytotoxicity and significantly augmenting the release of perforin, granzyme, and IFN-γ [266, 267].

These approaches are also employed in CAR-T cells to obstruct the impact of TGF-β by integrating the CAR with the dnTGF-βRII which acts as a decoy receptor for the cytokine, thereby inhibiting the initiation of its downstream signaling pathway [268]. CAR-T cells use dnTGF-βRII to block TGF-β, enhancing their antitumor effect [155, 269]. Given the advantages of developing TGF-β resistant CAR-T cells, a phase 1 clinical trial was carried out to evaluate the efficiency of CAR-T cell therapy modified with dnTGF-βRII in cancer [270].

In summary, researches suggest a complex role for TGF-β in asthma. Its presence seems to be dose and time-dependent, potentially contributing to airway fibrosis and cough. Unfortunately, TGF-β suppresses NK cells. CAR studies suggest removing TGF-β may benefit asthma. Future research could engineer CAR cells to target TGF-β or block its production in asthmatic cells, offering a new treatment approach (Fig. 5).

CCLs

CCLs such as CCL3 (MIP-1α ), CCL4 (MIP-1), and CCL5 termed RANTES have a role in the pathogenesis of asthma by inducing type 2 inflammation [271]. CCL3 is expressed highly by macrophages, NK cells, neutrophils, and other immune cells in inflammatory conditions [272]. CCL3 could potentially function as a significant genetic modulator for T-lymphocytes, macrophages, and chemo-attractants responsible for mononuclear cells [273]. Conversely, increased expression of CCR5, a CCL3 receptor, can stimulate T cells to produce more IL-2, potentially mitigating asthma symptoms [274].

CCL4 is produced by a wide range of cells including monocytes, NK cells, neutrophils, T and B lymphocytes, as well as epithelial and endothelial cells lining tissues, and fibroblasts within connective tissue [275, 276]. CCL4 acts against eosinophils after releasing from an activated form of them, especially in inflammatory conditions of airways [277]. On the other hand, some studies demonstrated that CCL4 can exacerbate asthma and AHR [271]. Immune cells such as monocytes, NK cells, and T cells are attracted to the inflamed area by binding CCL4 to its receptors CCR5 and CCR8 [278]. A study by Capelli et al. found significant differences in CCL4 levels between the BAL of chronic bronchitis patients and control subjects [279]. Additionally, Dupilumab treatment reduced CCL4 expression in nasal polyps, potentially mitigating excessive inflammation [280].

CCL5 is a potent chemoattractant for monocytes, T helper cells, DC cells, eosinophils, and NK cells at the site of inflammation. CCL5 exhibits a higher binding affinity for CCR1 and CCR5 compared to CCR3. Th1 cells usually have CCR1 and CCR5 on their surface, while Th2 cells mostly express CCR3 [281, 282]. Elevated concentrations of CCL5 have been documented in individuals diagnosed with asthma [283]. The use of antibodies to specifically target CCL5 in a murine model of allergic airway disease has revealed the capacity to hinder inflammation within the airways [284]. Additionally, Allard et al.‘s findings demonstrate that CCL5 activates monocytes in the exacerbation of rhinovirus-induced asthma [285]. As a result, Reducing CCL2, CXCL8, and CXCL10 in CAR-NK cells via gene editing might be beneficial due to their inflammatory effects.

ADCC and CD16

NK cells utilize a variety of effector mechanisms in order to eradicate target cells. Of particular importance is the involvement of CD16A (FcγRIIIA), a low-affinity Fc receptor expressed on their surface. CD16A specifically recognizes the Fc region of antibodies pre-bound to target cells, initiating ADCC. ADCC is a mechanism by which antibodies induce the elimination of target cells through non-phagocytic means [286]. ADCC can be mediated by several human antibody classes: IgG, IgA, and IgE [287]. This process empowers NK cells to function as potent cytotoxic effectors against cells targeted for destruction by antibodies [167, 288].

ADCC represents a critical mechanism for NK cells to eliminate virally infected cells. Furthermore, in vitro ADCC capacity has been linked to susceptibility to asthma following respiratory infections [289]. As prominent ADCC effectors, NK cells utilize three primary mechanisms to eliminate target cells: cytotoxic granule exocytosis, Fas ligand signaling, and release of perforins and granzymes. The release of perforin and granzymes from granules represents the principal and best-characterized mechanism employed in ADCC [290, 291]. While various myeloid lineage cells can mediate ADCC, NK cells appear to be the key effectors in vivo. The clinical effectiveness of many targeted mAb therapies has been shown to be dependent on NK cell activity [292]. This highlights the critical role of NK cells and their high expression of activating CD16A in ADCC, making them a target for optimizing antibody interactions in the context of immunotherapy.

Screening for ADCC activity could identify patients needing alternative immunotherapy or interventions to boost their ADCC [293]. CAR-NK cells co-expressing CD16A and CD64 (FcγRI) show promise, demonstrating enhanced and sustained ADCC [294] (Fig. 6).

Perforin and granzyme

NK cells eliminate stressed, transformed, or infected cells, including damaged epithelial cells found in asthmatic lungs by releasing cytolytic granules that consist of perforin and granzymes [295, 296]. These granules work in a coordinated fashion: perforin creates pores in the target cell membrane, while granzymes infiltrate the cell and trigger apoptosis through multiple pathways. Granzyme B can achieve this in two ways: by activating a special suicide program (caspase-3 cleavage) or through other methods not depending on this program [297, 298]. Notably, research suggests a link between impaired NK cell cytotoxicity and the development of severe asthma, highlighting the importance of this process in maintaining lung health [13].

Perforin-deficient mice develop asthma-like symptoms (Mathias et al.), suggesting its critical role in preventing the disease [299].

Benralizumab, a therapeutic agent for severe eosinophilic asthma, exerts its multifaceted effects through binding to their IL-5Rα, blocking survival signals and leveraging NK cells to eliminate them [300]. This activation triggers ADCC, prompting NK cells to release perforin and granzyme, ultimately inducing apoptosis in both eosinophils and basophils [252, 300, 301]. Recent in vitro investigations provide additional insights into this mechanism, elucidating the essential function of an eosinophil-NK cell synapse in the induction of eosinophil apoptosis by benralizumab. In a significant breakthrough, Dagher et al. revealed that benralizumab can eliminate eosinophils through macrophages via two distinct pathways: directly engulfing eosinophils in a process called ADCP and stimulating macrophages to produce TNF, a molecule known to trigger cell death. Eosinophils upexpress TNFR1 during apoptosis. Interestingly, the presence of NK cells further amplified TNFR1-mediated apoptosis by releasing another signaling molecule, IFN-γ [302].

NK cells targeting eosinophils show promise for eosinophilic asthma treatment, offering a potential alternative to IL-5 biologics with fewer side effects [13]. Further research on enhancing NK cell function is warranted (Fig. 6).

Fas/FasL

Fas (CD95, APO-1), a type I transmembrane protein belonging to the TNFR superfamily, harbors a cytoplasmic death domain. Its ligand, FasL, a 40 kDa protein appertaining to the TNF superfamily, triggers apoptosis. Fas-FasL pathway triggers cell death in activated immune cells, maintaining immune balance by eliminating potentially harmful lymphocytes and preventing tissue damage [303, 304].

While NK cells express a diverse array of death ligands, only FasL and TRAIL have been demonstrably implicated in direct target cell cytotoxicity during NK-mediated killing in humans and mice [305, 306]. Upon ligand binding, Fas triggers distinct signaling pathways categorized as apoptotic and non-apoptotic. Mechanistically, in an apoptotic way, Fas engagement by FasL recruits FADD, which subsequently binds procaspase-8. This interaction leads to the formation of the DISC and activation of caspase-8. Activated caspase-8 then cleaves and activates downstream caspases, such as caspase-3 and caspase-7, ultimately culminating in apoptosis [307, 308]. The activation of nonapoptotic pathways by Th2 cells using Fas-mediated mechanisms might be involved in the progression of pulmonary inflammation. The regulation of Fas signaling within Th2 cells is crucial in the inhibition of type 2 inflammatory responses. Distinguishing Th2 cell Fas signaling is challenging. Fas-FasL antagonists might harm other immune cells. More specific therapies require further research [309].

FasL inhibition resulted in enhanced T cell activation, with a dose-dependent and time-dependent increase in IL-5, IL-9, GM-CSF production, and airway eosinophilia. Paradoxically, activated T cells suppressed eosinophilic airway inflammation and allergic asthma, likely through a subsequent reduction in cytokine production [309, 310]. FasL restricts the proliferation and expansion of antigen-specific T cells, consequently diminishing the production of Th2 cytokines. [311]

NK cells may play a role in asthma by regulating neutrophil activity in the lungs [146]. Asthmatic patients show airway neutrophilia, suggesting disrupted communication between these cells [312]. NK cells from asthmatic patients also exhibit abnormal function [313].

CAR-NK cells targeting the Fas-FasL pathway offer promise for new asthma treatments. Studies show engineering CAR-NK cells with a modified Fas receptor lacking the death domain (ΔFAS) improves their persistence [314]. CAR-NK cells targeting FasL offer a new approach for asthma by inducing Fas-mediated apoptosis in immune cells, potentially reducing inflammation Building on the success of CAR cell therapies that exploit the Fas/FasL pathway to enhance cytotoxicity [100, 315] (Fig. 6).

IFN-γ

NK cells and NKT cells are crucial producers of IFN-γ during the innate immune response. In contrast, CD8 + and CD4 + T cells become significant sources of IFN-γ during the adaptive immune response [199]. The secreted IFN-γ binds to its receptor IFNGR on cells, regulating the immune response. IFN-γ can also induce APCs to release IL-12, reactivating IFN-γ production. This forms a positive feedback loop in inflamed settings and can reduce asthmatic inflammation [316]. T-bet, a transcription factor from the T-box family (TBX21 gene) promotes IFN-γ production in Th1 cells and NK cells, but not Th2 cells [316]. Introducing T-bet into Th2 cells via transduction turns them into IFN-γ-producing Th1 cells [317]. This process could be a promising approach to mitigating asthma by potentially suppressing the release of excessive Th2 cytokines.

IFN-γ has several signaling pathways. In the Canonical pathway, IFN-γ locks onto its receptor, which triggers a domino effect. Two proteins, JAK1 and JAK2, get activated and modify a specific site on another protein known as STAT1. This activated STAT1 pairs up with another STAT1, and together they move to the nucleus. There, they bind to their specific gene (GAS). As a result, GAS increases the production of proteins that fight infection [318]. Meanwhile, SOCS hinders JAK and STAT activation. In addition, PIAS restrains gene transcription induced by IFN-γ [319]. During a non-canonical pathway, it activates a series of proteins including STAT1, PI3K, and Akt. This activation eventually leads to a protein named mTOR playing a role in the IFN response. Additionally, mTOR works with p70S6 kinase to increase the production of IFN-γ [320, 321].

Th1 IFN-γ suppresses asthma by inhibiting Th2 cells [322]. Production of IFN-γ by NK cells has the potential to induce the polarization of CD4 + T-cells towards a Th1 phenotype [323]. In addition, IFN-γ inhibits mucus, chitinases, and eosinophilia through the airway epithelium independently of Th2 cell activation [324], and might favor allergic inhibition by reducing IgE production [325]. Patients diagnosed with allergic asthma demonstrate elevated levels of IL-4-producing NK cells and reduced levels of IFN-γ-producing NK cells circulating in the bloodstream [326]. Both the IFN-γ + NK cells and the concentration of IFN-γ exhibited a notable decrease in individuals with severe asthma in comparison to those in good health. This finding indicates that NK cells derived from severe asthma patients may possess an inherent deficiency that results in diminished levels of IFN-γ expression and secretion [4]. Therefore, enhancing the ability of NK cells to produce IFN-γ holds promise as a therapeutic strategy for alleviating the symptoms of asthma [323]. Numerous studies support IFN-γ’s inhibitory role in asthma pathogenesis. In a study by Renzi et al., infants with lower levels of IFN-γ in their PB experienced more severe asthma symptoms. Additionally, the ratio of IFN-γ to IL-4 was lower in infants requiring medication for their symptoms [327, 328]. Another research shows that decreased levels of IFN-γ + CD4 + cells and lower concentrations of IFN-γ were found in individuals diagnosed with allergic asthma [329]. Additionally, Ex vivo studies suggest impaired IFN-γ responses in asthma patients compared to healthy individuals during viral infection [330]. In contrast, the transcriptomic analysis showed that TNF-α and IFN-γ exposure affects gene expressions in ASM, reducing corticosteroid sensitivity and potentially worsening pediatric asthma symptoms [194]. NK cells’ cytotoxic properties via IFN-γ elimination of target cells in asthma suggest potential for enhanced efficacy through CAR therapy [331, 332] (Fig. 6).

DNAM-1

DNAM-1 (CD226), a transmembrane glycoprotein of the immunoglobulin superfamily, plays a pivotal role in immune regulation [333, 334]. In NK cells, DNAM-1 functions as a key activating receptor. Deficiency in DNAM-1 impairs essential NK cell processes like proliferation, activation, and effector functions [335,336,337]. It works alongside other activating receptors like NKG2D and NCRs to trigger cell killing by binding specific ligands [335]. DNAM-1 signals activation through its interaction with two ligands, PVR (CD155) and Nectin-2 (CD112) [338]. Notably, DNAM-1 functions as a co-stimulatory receptor, harboring an ITAM-like motif that amplifies signaling pathways initiated by ITAMs within immune cells [339].

Intriguingly, the immunoregulatory role of DNAM-1 extends beyond NK cell activation and might hold relevance in asthma and AHR. Studies suggest that DNAM-1 signaling promotes the activation, proliferation, and differentiation of Th1 and Th17 cells which are considered to be protective in asthma [336]. Additionally, DNAM-1 in CD4 + T cells promotes IFN-γ production while its deficiency favors the production of pro-inflammatory IL-4 and IL-13 cytokines [340, 341]. Similarly, DNAM-1 + CD8 + T cells exhibit enhanced production of IFN-γ and granzyme B, further supporting their potential role in reducing inflammation [342].

DNAM-1’s versatility makes it a target for CAR-NK engineering. Studies show engineered NK cells with activated DNAM-1 exhibit direct cytotoxicity [343], supporting the potential of CAR-NK for targeting tumor antigens [344, 345]. CAR-NK-92 cells engineered with a DNAM-1 intracellular domain exhibit enhanced cytotoxicity compared to CAR-NK cells incorporating a CD28 costimulatory domain. CAR-NK-92 cells, which possess both the DNAM-1 and 2B4 costimulatory domains, demonstrated the most elevated cytolytic activity [346]. Adoptive transfer of NK cells with increased DNAM-1 levels can be very helpful in the treatment of asthma, considering that these cells play an important role in the production of cytokines and intensification of the overreaction of the airways.

DNAM-1 CAR-NK cells may outcompete inhibitory receptors like TIGIT and CD96 for binding to PVR and Nectin-2 ligands, potentially leading to enhanced activation and improved cytotoxicity. Therefore, incorporating DNAM-1 into CAR-NK presents a promising avenue for alleviating asthma symptoms, further investigation is necessary to elucidate potential complications and definitively assess its efficacy.

TNF-α

TNF-α, encoded by a gene on chromosome 6, is a key cytokine implicated in inflammatory processes [347]. Structurally, this homotrimeric protein of 157 amino acids is primarily produced by activated macrophages, T-lymphocytes, and NK cells [348]. TNF-α can augment NK cell cytotoxicity [349], and its pleiotropic effects on various biological processes make its influence on immune regulation in asthma multifaceted [350]. Notably, research suggests TNF-α can modulate both pro-inflammatory and anti-inflammatory mechanisms [351]. TNF-α exists in both soluble (sTNF-α) and transmembrane (tmTNF-α) forms. Initially synthesized as tmTNF-α, it requires cleavage by TNF-α-converting enzyme for release as the active sTNF-α [352]. This cytokine exerts its effects by binding to two distinct TNF receptors, TNFR1 and TNFR2 [350]. The interaction of TNF-α with these receptors, particularly TNFR1, triggers signaling cascades that mediate diverse biological processes [348]. Notably, TNFR1 signaling is primarily linked to pro-inflammatory and apoptotic pathways, while TNFR2 signaling is crucial for mononuclear cell activation and proliferation [353, 354].

Emerging evidence suggests a multifaceted role for TNF-α in asthma pathogenesis. The airways of individuals with neutrophilic and severe asthma exhibit elevated TNF-α levels [355]. TNF-α promotes the production of Th2 cytokines (IL-4, IL-5, IL-13), which further enhance ASM contraction [356, 357]. Furthermore, TNF-α, IFN-γ, and IL-1β may increase CD38 expression (cyclic ADP ribose hydrolase) in ASMs, potentially leading to increased intracellular Ca2 + signaling and AHR induction [358]. TNF-α also synergizes with IL-17 cytokines to recruit neutrophils, contributing to neutrophilic inflammation [115, 359]. TNFR1-mediated hyperactivation of the NF-κB pathway is associated with upregulation of pro-inflammatory genes and airway inflammation in asthma patients [360]. Conversely, TNFR2 signaling demonstrates an anti-inflammatory effect by suppressing Th2 and Th17 cell differentiation and decreasing cytokine levels [361, 362]. In contrast to the prevailing view, certain research indicates that TNF-α binding to TNFR2 can enhance T cell activation, proliferation, and survival during airway inflammation [363]. Notably, TNF-α appears to exert these effects primarily on CD8 + Teff cells [364,365,366,367,368,369,370,371,372]. These seemingly contradictory findings highlight the complex interplay between TNF-α and its receptors in asthma. TNFR1 drives inflammation, while TNFR2’s therapeutic potential requires further exploration.

CAR-NK cells engineered to target TNF-α may offer therapeutic potential for asthma. These cells could eliminate mucus-producing goblet cells or deplete inflammatory immune cells, potentially improving symptoms [348, 373,374,375,376,377,378,379,380,381,382,383,384,385]. However, the precise role of TNF-α in asthma and CAR-NK therapy warrants further investigation.

Table 4 A comparative analysis of the various NCR components
Fig. 4
figure 4

NK Cells Promote Healing (Anti-inflammatory Actions): NK cells have surprising anti-inflammatory capabilities through Nkp46 receptor and IL-10. (A) By Nkp46 receptor They can connect with plasmacytoid dendritic cells, triggering responses that dampen inflammation. (B) Via IL-10 production, NK cells can suppress inflammation in three ways. STAT3 pathway reduces inflammatory molecule production (Nf-kB gene expression) through inhibition of MAPK pathway of NfkB. p38/MAPK pathway helps increase the production of HO-1, a molecule with anti-inflammatory properties. PI3K/AKT pathway reduce pro-inflammatory signals and caspase-3 to induce anti-inflammatory responses

Fig. 5
figure 5

TGF-β CAR NK cells: methods for engineering NK cells to overcome the suppressive effects of TGF-β. These engineered cells are known as TGF-β CAR NK cells. One method uses CRISPR/Cas9 technology to delete the gene responsible for making the TGF-β receptor on the NK cell. Without this receptor, the TGF-β molecule can’t bind and suppress the NK cell’s activity. As a result, these engineered NK cells produce more interferon gamma and become more cytotoxic. Another method builds a new receptor molecule for the NK cell. It uses the ectodomain of the TGF-β receptor, but combines it with the endo domain of NKG2D. This allows the engineered NK cell to recognize target cells but bypasses the TGF-β suppression. This approach improves NK cell function and reduces the inhibitory effects of TGF-β. dnTGF-β CAR-NK cells lack both the functional receptor and the ability to respond to TGF-β signals. Consequently, the TGF-β molecule has no suppressive effect, and these engineered NK cells show increased production of interferon gamma, perforin, and granzymes

Fig. 6
figure 6

NK cell’s cytotoxicity: The image describes how NK cells fight against target cells within the inflamed airways of an asthmatic patients. (A) NK cells utilizes CD16A to recognize Fc region of antibodies pre-bound to target cells, initiating ADCC. ADCC induce the elimination of target cells through FAS ligand signaling and release of perforin/granzymes. (B) NK cells release IFN-γ, which is a signaling molecule. It binds to receptors on target cells, activating a protein named STAT1.Then, Two STAT1 molecules combine and bind to DNA, promoting the production of proteins crucial for fighting against allergens by specific gene known as GAS. Also, IFN-γ can activate additional pathways involving proteins such as mTOR. This loop ultimately leads to increased IFN-γ production, further amplifying the immune response. (C) NK cells contain specialized granules filled with perforin and granzyme B. Perforin creates pores in the target cell’s membrane. Granzyme B enters the target cell through the pores, initiating apoptosis. (D) FasL binds to Fas receptors on target cells. This Binding triggers recruitment of FADD, which activates procaspase-8 into caspase-8. Caspase-8 directly cleaves target proteins, inducing apoptosis. Notably, caspase-8 can also activate caspase-3, another cell death executioner to robust more apoptosis response

CAR-NK vs. CAR-T: Unveiling potential, challenges, and solutions

NK cells offer several advantages over T cells for CAR-based immunotherapy. Unlike T cells, which require antigen presentation for activation, NK cells are activated through their innate receptors. This allows CAR-NK cells to retain their cytotoxic function even if the target loses or downregulates its antigen, Additionally, NK cell-based therapies are associated with a lower risk of severe side effects compared to CAR-T cell therapy. This is because NK cells have a limited lifespan in the body, reducing the chance of complications like CRS, ICANS, and GVHD [27]. These advantages position CAR-NK cells as a promising next-generation cellular therapy. A detailed description of the comparison of CAR-T vs. CAR-NK is discussed below (Table 5).

Table 5 Comparison of CAR-T vs. CAR-NK

Source of immune cells

A critical advantage of CAR-NK cells over CAR-T cells lies in the source of allogeneic immune effector cells. CAR-T cell therapy necessitates the utilization of autologous T lymphocytes, rendering patients who have undergone prior lymphodepletion therapies ineligible due to insufficient T cell counts. This limitation can significantly delay, or even preclude, treatment initiation. Studies have demonstrated that a substantial proportion of patients fail to meet the minimum T cell collection threshold for CAR-T cell manufacturing [386].

CAR-NK cells offer a compelling alternative in this regard. Unlike T cells, NK cells exhibit reduced alloreactivity and minimal risk of GVHD due to their MHC-independent activation pathways. This characteristic enables the development of off-the-shelf CAR NK cell products. Allogeneic NK cells can be sourced from diverse origins, including PB [63], UCB [75], hESCs, iPSCs [93], or cell lines like NK-92 [80].

Manufacture and cost

While CAR-T has revolutionized asthma treatment [18,19,20,21, 387], access remains limited due to high costs and complex, personalized manufacturing [388]. Recent research explores cost reduction through distributed, automated CAR-T manufacturing, demonstrating significant savings compared to centralized processes [389,390,391]. An alternative approach is the development of off-the-shelf CAR-T therapies. These therapies involve the large-scale production of genetically modified cells at a central location, enabling them to be used allogeneically for multiple patients [392].

In contrast to CAR-T cells, CAR-NK cells exhibit a naturally diminished propensity to induce GVHD, enabling their allogeneic application without the need for complex gene editing interventions [393]. Similar to CAR-T cells, cytokines can independently induce NK cell proliferation to levels sufficient for clinical application [394, 395]. While feeder cells significantly augment this expansion, their complete removal from cultures remains challenging, posing potential safety risks [396,397,398,399]. To address this, researchers are exploring alternative feeder-free methods. One promising approach utilizes dissolvable, polymer-based microspheres that gradually release growth factors and nutrients, thereby facilitating NK cell expansion [400].

Delivery approach

Delivering CAR constructs to T and NK cells utilizes distinct approaches due to their inherent biological differences. CAR-T cell manufacturing traditionally relies on viral vectors, such as retrovirus, adeno-associated virus vectors, and especially lentivirus, particularly VSV-G pseudotyped lentiviral vectors, to achieve efficient and sustained CAR expression [102]. However, NK cells present a challenge due to their low VSV-G receptor expression, hindering lentiviral transduction [401]. A promising solution lies in the Baboon envelope pseudotyped lentiviral vector, which significantly enhances NK cell transduction and even facilitates the delivery of CRISPR components for potential gene editing [402]. While other lentiviral envelope options like gibbon ape leukemia virus envelope and feline endogenous retrovirus envelope protein (RD114-TR) exist, BaEV currently appears to be the most effective vector for NK cell transduction [102, 403, 404]. Safety concerns regarding insertional mutagenesis associated with some viral vectors require further investigation in the context of CAR-T therapy [405], although long-term studies with CAR-T therapy haven’t reported such malignancies [288].

Beyond viral vectors, research explores non-viral delivery for CAR-T/NK cells due to limitations of viral vectors such as immunogenicity, cost [102, 406]. These methods (Sleeping Beauty, PiggyBac transposons) offer a safer integration profile but lower efficiency [102]. Alternatively, mRNA CAR constructs with electroporation or lipid nanoparticles achieve high, but transient, CAR expression [407]. DNA nano vectors represent another promising strategy, enabling non-integrating CAR expression with minimal immunogenicity and genotoxicity [408]. CRISPR-Cas9 unlocks targeted CAR-NK engineering, including CAR integration and inhibitory gene deletion [409]. Novel delivery methods like biomaterial vehicles and microrobots explore ex vivo-independent approaches [410, 411]. The field is rapidly evolving, with BaEV lentiviral vectors showing promise for CAR-NK, while non-viral and innovative delivery methods hold potential for safer and more versatile CAR-T and CAR-NK therapies.

CAR Structure

CAR-NK and CAR-T cells share a fundamental CAR structure with three key domains [412]. However, a key distinction lies in their co-stimulatory molecules. CAR-T cells typically utilize CD28 or 4-1BB, while CAR-NK cells leverage NKG2D and 2B4 [27].

Persistent

The field of ACT faces a crucial challenge: ensuring the long-term persistence of infused immune effector cells. The conventional view suggests that NK cells have a lower potential for long-term persistence compared to T cells [413]. This limited lifespan is believed to contribute to the modest clinical success observed in NK cell-based therapies [414, 415]. Consequently, CAR-NK cell development has primarily focused on enhancing cytotoxic function, with less emphasis placed on promoting persistence [183, 416].

However, research offers a promising approach to improving CAR-NK cell persistence. Engineering NK cells to express immunostimulatory cytokines like IL-15 has demonstrated enhanced survival [417, 418]. Moreover, short-term exposure to a combination of IL-12, IL-15, and IL-18 can generate memory-like NK cells exhibiting improved persistence, potentially leading to more efficacious CAR-NK cell therapy [419, 420]. In contrast, CAR-T cell development has successfully incorporated costimulatory domains into their design, enriching memory-like CAR-T cells and leading to enhanced persistence and durable treatment responses in both preclinical models and clinical trials [421]. Furthermore, engineering CAR-T cells to express immunostimulatory cytokines such as IL-7, IL-12, IL-15, IL-18, IL-21, and IL-23, creating so-called “armored CARs,” has shown promise in improving persistence and is currently being evaluated in ongoing clinical trials [422, 423].

Inducible promoters offer a promising strategy to enhance the functionality, safety, and specificity of CAR cells. These promoters are designed to activate only upon a specific trigger, such as recognition of an antigen, metabolite, drug, or a particular cell signaling pathway [424]. NFAT promoters control CAR-T cell transgene expression [425], while NF-κB promoters in CAR-NK cells trigger IL-12 secretion upon activation, boosting cytotoxicity and monocyte recruitment [426].

Pre-conditioning regimens using lymphodepletion is a crucial factor influencing the persistence of infused T cells in CAR-T therapy, and this approach may hold similar importance for CAR-NK therapy [427]. Typically, a fludarabine/cyclophosphamide combination is used prior to CAR-NK infusion to minimize host rejection of the infused NK cells [428]. Studies in unmodified NK cell therapy suggest that high-dose lymphodepletion promotes in vivo NK cell expansion and persistence, highlighting its potential role in enhancing CAR-NK cell therapy efficacy [64]. CAR-T therapy shows promise with selective lymphodepletion using CD52 antibodies and knockout CAR-T cells [429]. Whether such an approach can be applied to CAR-NK therapy remains to be explored.

Expansion

For CAR-T cells, prolonged ex vivo expansion can be detrimental, leading to a state of exhaustion that negatively impacts their memory phenotype, longevity, and proliferative capacity upon reintroduction [430, 431]. Achieving sufficient numbers of CAR-T cells for therapy hinges on the ex vivo expansion duration. Traditional protocols cultivate T cells for 9–14 days, but recent trends suggest potential benefits of shortening this period. Studies indicate that a shorter ex vivo culture may limit excessive T-cell differentiation, potentially enhancing efficacy [432]. However, cytokines like IL-2, commonly used for expansion, can induce exhaustion and decrease persistence. Conversely, expansion with IL15 or a combination of IL-2, IL-4, and TGF-β resulted in less differentiated CAR-T [433,434,435]. Similarly, studies suggest that incorporating IL-4, IL-7, and IL-21 promotes stemness and inhibits exhaustion [436].

A crucial stage in CAR-NK cell production is expanding the transduced cells. However, limited NK cell proliferation capacity hinders achieving sufficient numbers. Telomere shortening restricts their proliferative potential, but incorporating IL-21 into the culture medium can address this [437]. Co-culturing NK cells with feeder cells expressing IL-15, 4-1BB ligand, and MICA also enhances proliferation and viability [438]. While cytokines optimize ex vivo expansion, specific combinations like IL-2, 15, and 18 with anti-CD3 and CD52 beads expedite expansion [27, 439]. Notably, IL-15 alone, without IL-2, enhances cytotoxicity and expansion without activating regulatory T cells [27, 395]. IL-21 also promotes NK cell proliferation, particularly in synergy with feeder cells [440].

Antigen loss

A critical challenge shared by both CAR-T and CAR-NK cell therapies is antigen loss, which can lead to treatment failure [441]. Several strategies have been explored to address antigen loss in CAR-T therapy, including the development of dual-targeted CAR T cells and sequential administration of CAR-T cell products targeting distinct antigens [442,443,444,445,446,447]. However, these approaches are not without limitations, as documented cases of relapse due to antigen loss in patients receiving dual-targeted CAR-T cell therapy are highlighted [442,443,444]. Preclinical models have shown promise for additional approaches, including multi-antigen targeting using CARs incorporating ankyrin repeat domains, and engineering CAR-T cells to modulate the endogenous immune system. These approaches all hold promise for overcoming antigen loss and limiting relapse in CAR-T therapy [448,449,450].

CAR-NK cells may possess a potential advantage in overcoming antigen loss compared to CAR-T cells. Their intrinsic cytotoxic arsenal, mediated by natural cytotoxicity NCR and death receptor ligands, offers a built-in resistance mechanism against CAR target antigen loss [214, 451]. Moreover, intracellular signaling of NK cells (NKG2D and DNAM-1) can contribute to the identification of the target cell independent of CAR, antigen-loss does not affect the efficacy of CAR-modified NK cells [452, 453]. Li et al. [454] explored the use of an antigen-specific inhibitory KIR-based receptor that effectively inhibited aCAR-mediated trogocytosis and the associated immunomodulatory consequences. This approach successfully preserved the ability of NK cells expressing the same antigen to exert key effector functions against target cells [454]. Beyond inhibitory KIR antagonism, cytokine treatment strategies are explored to reduce inhibitory KIR levels on NK cells, thereby enhancing CAR-NK cell activity against HLA-A/B/C + targets. Studies indicate superior KIR downregulation with IL-12, IL-15, and IL-18 compared to IL-15 alone [455].

Cytotoxicity

A key area of research is optimizing the cytotoxicity of CAR-T and CAR-NK cells. One strategy involves engineering costimulatory domains into the CAR construct to enhance the activating intracellular signaling of the modified cells. While the TCR serves as the primary signaling receptor for T cell cytotoxicity [411], various transmembrane costimulatory molecules, such as CD28, 4-1BB, ICOS, and OX-40, can amplify downstream signaling pathways when incorporated within the costimulatory domain of the CAR. Each of these costimulatory molecules influences distinct signaling cascades that impact CAR-T cell cytotoxicity, proliferation, and persistence [456, 457]. In contrast, NK cells utilize a complex network of activating and inhibitory receptors that recruit specific downstream signaling mediators. Fine-tuning these signaling cues determines the balance between NK cell activation and inhibition [411]. Studies suggest that tailoring CAR engineering specifically for NK cells by incorporating NK-specific signaling domains, such as 2B4 and DAP10/DAP12, within the costimulatory domain, can enhance CAR-NK cell efficacy and IFN-γ secretion compared to CAR constructs designed for T cells [457, 458].

Side effects

Despite the remarkable response rates achieved with CAR-T cell therapy, a significant drawback lies in the potential for severe side effects [459]. These complications are primarily attributed to the potent cytokine storm triggered upon CAR-T cell activation [460, 461]. CRS and ICANS are prominent examples, manifesting as fever, multi-organ failure, and neurological symptoms [461, 462]. Management of these cytokine-mediated toxicities often necessitates corticosteroids and, in severe cases, anti-cytokine antibodies [463]. This cytotoxicity profile stands in stark contrast to that observed with CAR-NK cells. Early clinical trials with CAR-NK cells have not reported CRS or neurotoxicity, potentially due to a distinct cytokine release pattern upon activation [27]. CAR-T cell activation triggers the release of a pro-inflammatory cytokine storm, including TNF-α, IL-1β, IL-2, and IL-6 [462]. Conversely, CAR-NK cells appear to secrete a less inflammatory cytokine profile, with GM-CSF being a notable example [452]. This suggests that CAR-NK cells might offer a potentially safer cytotoxicity profile compared to CAR-T cells, with a reduced risk of severe side effects. Further research is warranted to definitively compare the direct cytotoxic potential of these cell types, but the emerging evidence suggests a promising avenue for CAR-NK cell therapy with potentially improved tolerability.

Improving CAR-NK therapy safety and efficacy

Use of immune checkpoints like PD-L2 and CTLA-4

The immune checkpoint molecule PD-L2, which is a ligand for PD-1, plays a role in regulating inflammation in asthma. Studies suggest that a deficiency in PD-L2 leads to increased activity of immune cells such as eosinophils and lymphocytes, potentially contributing to excessive response to allergens in asthmatic patients [464, 465]. Studies by Akbari et al. suggest that PD-L2 deficiency may worsen AHR, inflammation, and IL-4 secretion [466]. PD-L2 on CAR-T cells represents a promising strategy for future asthma treatment. Its well-established immunosuppressive properties suggest it could effectively dampen the inflammatory response characteristic of asthma.

Liu et al. investigated CTLA-4 as a checkpoint in CAR macrophages targeting M1 microglia, implicated in neuroinflammation. Engineered CAR-macrophages with the CTLA-4 showed promise in reducing M1 microglia activity [467]. A separate study suggests that CAR-T cells engineered with CTLA-4 in their cytoplasmic tail lead to a significant reduction in CRS, a common complication of CAR cell therapy. Zhou et al. suggest CTLA-4 incorporation in CAR-T cells promotes self-tolerance and potentially reduces trogocytosis, a process limiting CAR-T durability. This strategy could enhance CAR cells lifespan and efficacy [468]. Therefore, the usage of CTLA-4 in CAR therapy can be very useful in various aspects.

AI and ML in CAR cell therapy

Despite the remarkable achievements of CAR cell therapy, several challenges remain. These include optimizing CAR design and cell products, enhancing the time-consuming, and expensive manufacturing processes, improving response rates and remission durability, and minimizing toxicity [469, 470]. ML and AI offer exciting solutions to these challenges and are revolutionizing the field of medical research [471].

AI holds immense promise for CAR T-cell therapy, particularly in tackling the persistent challenge of target identification. Multi-omics approaches, encompassing a wide range of biological data including genomics, epigenomics, transcriptomics, T cell receptor repertoire profiling, proteomics, metabolomics, and microbiomics, generate vast datasets. AI algorithms can effectively explore these datasets to identify promising targets for CAR cell design [470, 472, 473]. Furthermore, AI facilitates the characterization of CAR cell phenotype. By employing multifaceted ML algorithms, researchers can analyze extensive datasets of the functional and structural characteristics of CAR cells, leading to a deeper understanding of their therapeutic potential [474].

To address manufacturing obstacles, the development of an automated AI-facilitated CAR cell manufacturing platform is underway. AI can analyze the platform’s accumulated data, providing valuable insights and driving decisions for process improvement [469, 475]. Beyond manufacturing, AI offers the potential to enhance CAR cell function in several ways. First, AI can analyze vast datasets generated from advanced techniques like scRNA-seq and TCR-seq to identify crucial genetic markers associated with treatment response. ML algorithms can then leverage these markers to predict individual patient responses, paving the way for personalized treatment strategies [476, 477]. Second, AI has the potential to improve CAR cell activation [478] and enhance gene editing methodologies for CAR cell design [479].

Moreover, AI can identify and manage adverse events like CRS [480,481,482]. AI-powered methods can analyze cytokine profiles and leverage existing clinical data to rapidly diagnose CRS, significantly aiding clinicians [480]. AI can also address the challenge of differentiating CAR cells from other blood cells. One study achieved a remarkable 99.63% accuracy in CAR cell identification using an AI model trained on microscopy images [483].

Developing robust AI-driven CAR cell prediction models necessitates the utilization of extensive omics and clinical datasets. The establishment of comprehensive multi-omics datasets is critical for the successful implementation of AI approaches [484]. Data limitations arise from evolving clinical practices (e.g., CAR dosage adjustments) [485], hindering AI integration in CAR cell therapy. Careful consideration and robust data collection strategies are crucial for successful integration.

Novel CAR Designs

The field of CAR engineering is actively exploring modifications to existing strategies. This relentless research and investigation aims to overcome limitations and develop a more selective and potent approach to CAR cell therapy [486]. Having reviewed the established CAR designs, we will now turn our attention to some promising new directions in CAR design (Fig. 7).

Fig. 7
figure 7

Novel CAR design: This figure explores novel CAR designs beyond established approaches. Researchers are investigating strategies to improve efficacy, safety, and target recognition. These include bispecific/dual CARs for multi-antigen targeting, TRUCK CARs for targeted cytokine release, armored CARs for co-expressing beneficial proteins, and nanobody/FHVH CARs for reduced immunogenicity. Suicide genes provide a safety measure for rapid cell elimination, while inducible and switchable CARs offer precise control over activation. Inhibitory-CARs address NK cell fratricide, and TCR-CARs broaden target recognition. Finally, “ON and OFF” CARs utilize drugs to regulate activity precisely. This comprehensive overview highlights the dynamic field of CAR design, constantly innovating to enhance therapeutic potential

Bispecific and dual car

To improve the efficacy of CAR cell therapy, researchers are exploring dual or multi-targeting functionalities within the CAR design. This can take two main approaches: First, multi-epitope targeting, where the CAR recognizes multiple distinct sites (epitopes) on the same target antigen, potentially strengthening the binding and enhancing cytotoxicity activity. Second, bispecific or multi-specific CARs can be engineered to recognize entirely different antigens on the target cell surface. This strategy aims to improve antigen recognition and prevent evasion [486].

TRUCK CAR

The TRUCK approach represents a novel strategy in CAR cell therapy. This approach involves engineering CAR cells to secrete specific transgenic cytokines upon CAR engagement with target antigens. Examples of such cytokines include IL-7 [487], IL-12 [488], IL-15 [489], IL-18 [332], IL-23 [490], and IL-33 [491]. This targeted cytokine release at the antigen site offers a two-pronged benefit: first, it can provide an auto-stimulatory effect, enhancing the activity of the CAR cells themselves. Second, it can activate other immune cell types, leading to a more robust anti-asthma response [492].

Armored CAR

Armored CAR cells are designed to co-express different proteins with the CAR, for example, antibodies or their fragments [493]. The multifaceted nature of armored CAR NK cells, with their ability to produce a wide range of cytokines, proteins, and enzymes, holds significant promise for the development of next-generation CAR NK cells with improved therapeutic efficacy [214].

Nanobody and FHVH CAR

To enhance CAR cell therapy, researchers are exploring alternative targeting domains with reduced immunogenicity and improved expression within cells. These alternatives move beyond the conventional scFv by utilizing shorter extracellular fragments. One promising approach involves single-domain variable heavy-chain antibodies derived from camelid antibodies (also known as nanobodies) [494]. Alternatively, a FHVH can be employed, minimizing the risk of an immune response against the targeting domain in patients [495].

Suicide gene

While NK cells generally exhibit a safer profile compared to T cells in cell-based therapies, additional safety measures can be crucial for emergency situations [496]. One strategy for achieving rapid termination of such therapies involves incorporating a suicide gene into the therapeutic transgene. In the context of NK cells, researchers have explored the iCasp9 safety switch for this purpose. This system utilizes a modified caspase-9 molecule fused to the human FK506 binding protein. This system allows for the selective elimination (apoptosis) of effector cells by introducing a small-molecule drug. CAR cells engineered with iCasp9 have demonstrated effectiveness in controlling cytotoxicity under unfavorable conditions [27, 75, 497].

Inducible CAR

The ability to control gene expression with precise timing and magnitude is crucial in various biological applications. One approach involves inducible expression systems, where gene expression can be triggered by specific cellular signals and transcription factors. Examples include synthetic Notch signaling [498, 499] STAT5, AP-1, and NFκB [500]. These systems offer researchers a powerful tool to manipulate gene expression in a tightly regulated manner. Alternatively, hypoxia-inducible factor-1α can be utilized to restrict gene expression to specific cellular environments with low oxygen levels [501]. This approach provides a strategy for targeted gene activation in response to distinct physiological conditions.

Switchable CAR

sCAR represents a novel approach to enhance the precision and safety of CAR cell therapy. Unlike conventional CAR-T cells that directly target antigens, sCAR cells lack this recognition domain. Instead, they rely on a specific “switch” molecule to control their activation. Two main strategies have emerged in sCAR design. The first utilizes an antibody-based switch where the sCAR recognizes the Fab fragment of an antigen-specific antibody pre-bound to the cell, offering precise control over activation and targeting [502,503,504]. Alternatively, an adaptor protein switch can be employed, such as zipFv, which combines a scFv with a leucine zipper fragment. This adaptor protein bridges the gap between the sCAR and the antigen-specific antibody, enabling controlled activation of sCAR cells. This innovative approach allows for a more refined control over CAR cell activity, potentially improving the safety and efficacy of CAR cell therapy [505].

Inhibitory-CAR

To address the challenge of NK cell fratricide mediated by trogocytosis, a novel strategy utilizes iCARs targeting NK-cell-specific inhibitory receptors. These iCARs essentially deliver a “don’t kill me” signal to NK cells. The co-expression of antigen-targeting activating CARs alongside these self-recognizing iCARs has demonstrated efficacy in preventing trogocytosis-mediated NK fratricide and enhancing the overall activity of CAR-NK cells [454].

TCR-CAR

TCR-CAR-NK cells represent a novel approach to engineering NK cells for target recognition. Unlike conventional NK cells, which recognize targets independently of MHC-I molecules, TCR-CAR-NK cells leverage TCRs to broaden their targeting capabilities. TCRs are specialized surface molecules that enable T cells to recognize specific antigen fragments presented by MHC-I molecules on target cells. By engineering NK cells with TCRs, researchers aim to equip them with the ability to recognize targets based on their specific surface protein expression (proteome) [214]. Studies have demonstrated the feasibility of this approach. Walseng et al. successfully transduced NK-92 cells with a TCR-CAR, enabling these engineered cells to react against MHC-I-positive target cells [506]. In essence, the integration of TCRs with CAR-NK cells offers a promising strategy for enhancing the target recognition potential of NK cells.

On and off CAR

ON and OFF-controllable CAR signaling systems utilize established drugs to regulate CAR activity precisely. Multiple strategies exist: lenalidomide can trigger CAR degradation for swift termination [507], dasatinib can induce gradual downregulation potentially mitigating T cell exhaustion [508], and researchers are exploring drug-induced proteolytic cleavage for rapid CAR inactivation [509, 510]. Drug-regulatable expression systems offer precise control over gene expression but face limitations like unintended “leakiness” in the off state, reduced expression compared to always-on systems, and a limited range between on and off states. Researchers are constantly innovating to address these challenges. A recent development, signal neutralization by an inhibitable protease platform, demonstrates significant improvements – it achieves a tight shut-off state, a wider range of expression control, and even greater potency compared to traditional, always-on systems in various pre-clinical models. This advancement highlights the ongoing efforts to refine drug-regulatable expression systems for broader applications [509].

Logic gate CAR

To improve the specificity of a targeting system, researchers are exploring the use of biological circuits with Boolean logic (Fig. 8). These circuits demand the engagement of multiple specific targets in a defined order for activation. An AND-gate design requires two or more targets for activation, achievable by separating signaling and costimulatory components into distinct entities [511].

Fig. 8
figure 8

Logic gate CAR: This figure explores the concept of Boolean logic circuits for enhanced target specificity in CAR-NK cell therapy. AND-gate and AND-NOT-gate designs require engagement of multiple targets for activation, offering high specificity but with challenges like complex transgene integration, potential immunogenicity, and vulnerability to antigen escape. The simpler OR-gate logic can be achieved through co-infusion of distinct CAR products or tandem targeting systems within a single construct. Overall, this figure highlights the ongoing advancements in CAR design towards achieving precise and safe targeting strategies

Both receptors in the AND-gate design must bind their specific ligands to trigger a full-fledged cellular response against the target. An alternative approach to achieve AND-gate logic involves sequential detection of multiple targets, as exemplified by synthetic Notch or synthetic intramembrane proteolysis receptor systems [499, 512, 513]. These designs utilize two gene expression cassettes. The first cassette constitutively expresses a synthetic receptor containing a transcription factor. Upon ligand (target A) binding, this receptor undergoes cleavage, releasing the transcription factor. The freed transcription factor then translocates to the nucleus and drives the inducible expression of a conventional targeting system (e.g., CAR) specific for a second target (antigen B). This sequential activation strategy ensures that only when both targets are present can a full response be initiated. However, a potential limitation of AND-gate designs is the risk of off-target effects. If healthy tissue expresses the target recognized by the inducible system (e.g., CAR) and is co-localized with the primary target cells, off-target activation can still occur [498].

Researchers are exploring AND-NOT-gate logic as an alternative strategy to enhance target specificity [514, 515]. Unlike AND-gate systems, AND-NOT gates require the presence of a specific target molecule (antigen A) and the simultaneous absence of another target (antigen B) to trigger a response. This is achieved by co-expressing two distinct signaling modules: one that activates upon binding antigen A and another that inhibits upon binding antigen B. The inhibitory module effectively overrides the activation signal if antigen B is present, preventing an unwanted response. An example of this approach involves co-expressing an activation module targeting a specific molecule with an inhibition module targeting a common cellular marker (e.g., HLA-A2). This AND-NOT-gate strategy offers the potential for highly selective targeting of cells lacking the inhibitory marker, thereby increasing targeting specificity while mitigating the risk of off-target activation [515, 516].

While AND- and AND-NOT-gated systems offer promising advantages in terms of target specificity, these designs also face noteworthy challenges. First, these circuits necessitate the co-expression of multiple transgenes. The resulting increase in the genetic payload size can significantly hinder the efficiency of transgene integration within target cells [517]. Second, early attempts at constructing these computational circuits using synthetic biology, such as the synthetic Notch system, often relied on non-human components like viral transcription factors. The potential immunogenicity of these foreign components presents a significant hurdle for translating such designs into clinical applications. Finally, these systems rely on the presence or absence of multiple specific targets in a defined combination. This increased complexity raises the probability of antigen escape, as the target entity needs only to modify the expression pattern of one of the multiple targets to evade detection [518].

Achieving OR-gate logic in target recognition can be accomplished through various strategies. A straightforward approach involves the co-infusion of distinct targeting systems (e.g., CAR cell products) against multiple antigens, essentially creating a ‘cocktail treatment.’ Clinical trials have demonstrated the efficacy of this approach, with studies showing improved survival rates in patients receiving combined targeting of EGFR and CD133 or CD19 and CD20. However, this method increases the workload for researchers and necessitates further investigation into potential toxicity associated with administering multiple targeting agents [519, 520].

Another strategy employs tandem targeting systems, which incorporate multiple antigen recognition domains (e.g., scFvs) within a single construct. Activation can occur upon binding to any of the targeted antigens. Tandem systems targeting CD19/CD20 and CD19/CD22 have shown promising results in pre-clinical models [521, 522]. Furthermore, designs incorporating even more targets, such as CD19/CD20/CD22, have demonstrated therapeutic potential against antigen-heterogeneous targets. However, with an increasing number of targets, safety considerations become paramount [523, 524].

A more versatile approach is exemplified by the split, universal, and programmable (SUPRA) CAR system developed by Wong et al. This system allows for the creation of various logic gates within CAR cells. The SUPRA CAR design splits the CAR into two components: a membrane-bound zipCAR and a freely circulating zipFv. The zipFv can bind to the zipCAR via complementary leucine zippers, forming a complete functional CAR. By utilizing different zipCAR/zipFv combinations, the SUPRA system can rapidly reprogram CAR cells to achieve different logic gates. For instance, the simultaneous addition of two zipFvs creates an OR-gate, enabling CAR cells to potentially target multiple antigens. However, the limited in vivo half-life of the zipFv currently restricts the broader application of the SUPRA CAR system [42, 505].

Conclusion and future perspective

While CAR-T cell therapy has shown remarkable progress, challenges such as GVHD, complex manufacturing, and the potential for severe side effects remain. CAR-NK cells offer a compelling alternative approach to address these limitations. Their “off-the-shelf” nature, potentially lower cost, and reduced risk of GVHD and CRS position them as a next-generation cellular immunotherapy.

In this review, we aimed to suggest potential strategies to enhance CAR-NK efficacy for the management of severe asthma and establish promising CAR design. NK can express IL-10, IFN-γ, perforin-granzyme, FASL, KIR, DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. We discussed that for enhancing CAR-NK efficacy in asthma, we can modulate IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, and KIR and decrease TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR using gene-editing.

NK cells are a powerful weapon in the immune system’s arsenal, wielding an array of tools to combat threats. These tools include cytokine production (such as IL-10 and IFN-γ) to modulate immune responses, ADCC to eliminate target cells, and direct target cell lysis through the perforin-granzyme system. Additionally, NK cells express FasL to trigger apoptosis in target cells, while receptors like KIRs, DAP, and DNAM-1 control their activation. NK cells also produce cytokines like TGF-β and TNF-α, influencing immune suppression and inflammation, respectively. They even release chemokines (CCL) to recruit other immune cells and express NKG2A, a receptor that can inhibit their activation. Finally, transcription factors and the EGFR further regulate their functions. By strategically modulating these diverse NK cell functions using gene editing, CAR-NK can be a promising approach to managing severe asthma.

The field of CAR cell therapy is undergoing a revolution with a focus on designing more precise and effective therapies. AI is revolutionizing CAR cell therapy by addressing key hurdles. AI can pinpoint promising targets for CAR design by analyzing massive biological datasets. It can also delve into CAR cell characteristics to optimize their function. Additionally, AI is being used to automate manufacturing, potentially making this therapy more accessible. AI can further personalize treatments by identifying genetic markers for response, and improve cell function by enhancing activation or reducing side effects. However, successful AI integration relies on robust data collection strategies to keep pace with the evolving field of CAR cell therapy.

Researchers are exploring various avenues to achieve an efficient therapy. One approach involves engineering CARs to recognize multiple targets simultaneously (bispecific CAR) or to secrete cytokines upon target engagement (TRUCK CAR). Additionally, alternative targeting domains with reduced immunogenicity (nanobodies) and safety mechanisms like suicide genes are being investigated. Furthermore, researchers are developing innovative methods for controlling CAR activity, including inducible CARs and switchable CARs. To enhance target specificity, complex logic gates mimicking Boolean operations (AND, AND-NOT, OR) are being explored. These advancements offer promising possibilities for the future of CAR cell therapy, paving the way for safer and more targeted treatments.

Data availability

Data will be available upon request by the editors/reviewers.

Abbreviations

ACT:

Adoptive cellular therapy

ADCC:

Antibody-dependent cellular cytotoxicity

ADCP:

Antibody-dependent cellular phagocytosis

ADP:

Adenosine diphosphate

AF:

Aspergillus fumigatus

AHR:

Airway hyper-reactivity/airway hyperresponsiveness

AI:

Artificial intelligence

Akt:

Ak strain transforming

AML:

Acute myelogenous leukemia

APC:

Antigen-presenting cell

ASM:

Airway smooth muscle

BaEV:

Baboon Endogenous Retrovirus

BALF:

Bronchoalveolar lavage fluid

cAMP:

Cyclic antimicrobial peptides

CAR:

Chimeric antigen receptor

CCAR-T:

Cytokine-anchored chimeric antigen receptor T

CCL:

(C-C motif) ligand

CCL:

CC chemokine ligand

CCR:

CC chemokine receptor

COPD:

Chronic obstructive pulmonary disease

CRISPR:

Clustered regularly interspaced short palindromic repeats

CRS:

Cytokine Release Syndrome

CTL:

Cytotoxic T lymphocyte

CTLA-4:

Cytotoxic T-lymphocyte-associated antigen 4

CXCL:

Chemokine (C-X-C motif) ligand

DAP:

Diaminopimelic acid

DC:

Dendritic cell

DISC:

Death-inducing signaling complex

DNA:

Deoxyribonucleic acid

DNAM-1:

DNAX accessory molecule-1

dnTGF-βR:

Dominant-negative TGF-β receptor

EGF:

Epidermal growth factor

EGFR:

Epidermal growth factor receptor

EGFR:

Epidermal growth factor receptor

EMPD:

Extracellular membrane-proximal domain

EMT:

Epithelial-mesenchymal transformation

EPX:

Eosinophil peroxidase

FADD:

Fas-associated death domain protein

FASL:

Fas ligand

FcεR:

The high-affinity IgE receptor

FDA:

Food and Drug Administration

FHVH:

Fully human heavy-chain-only variable domain

FLT3L:

Fms-like tyrosine kinase 3 ligand FoxP3:nuclear transcription factor Forkhead box P3

GM-CSF:

Granulocyte-macrophage colony-stimulating factor

GPC3:

Glypican-3

GVHD:

Graft versus host disease

HDM:

House Dust Mite

hESC:

Human embryonic stem cell

hiPSC:

Human induced pluripotent stem cell

HSCs:

Hematopoietic stem cells

HLA:

Human leukocyte antigens

HO-1:

Heme oxygenase-1

hPSC:

Human pluripotent stem cell

ICANs:

Cell-Associated Neurotoxicity Syndrome

iCARs:

Inhibitory chimeric antigen receptors

iCasp9:

Inducible caspase-9

IFN-γ:

Interferon-gamma

IL:

Interleukin

ILC:

Innate lymphoid cell

iPSC:

Induced pluripotent stem cells

ITAM:

Immunoreceptor tyrosine-based activation motif

ITT:

Immunoglobulin tail tyrosine

Jak/STAT:

Janus kinase/signal transducers and activators of transcription

KIR:

Killer-cell Immunoglobulin-like Receptor

LAG-3:

Lymphocyte activation gene-3

mAb:

Monoclonal antibodies

MAPK:

Mitogen-activated protein kinase

MHC:

Major histocompatibility complex

MICA and B:

Major histocompatibility complex class I chain-A and B

mIgE:

Transmembrane IgE

MIP-1:

Macrophage inflammatory protein-1

ML:

Machine learning

mTOR:

The mammalian target of rapamycin

NCR:

Natural cytotoxicity receptor

NFAT:

Nuclear factor of activated T-cells

NF-κB:

Nuclear factor kappa B

NK:

Natural killer

NKG2D:

Natural killer group 2D

NSCLC:

Non-Small Cell Lung Cancer

OVA:

Ovalbumin

PB:

Peripheral blood

PBMC:

Peripheral blood mononuclear cells

PD:

Programmed cell death protein

PD-1:

Death protein 1

PI3K:

Phosphatidylinositol 3-kinase

PVR:

Poliovirus receptor

PVRL2:

Poliovirus receptor-related 2

RANTES:

Regulated upon activation normal T cell expressed and secreted

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus 2

ScFv:

Single-chain variable fragment

SEA:

Severe eosinophilic asthma

SOCS-3:

Suppressor of cytokine signaling 3

SUPRA-CAR:

Split universal and programmable chimeric antigen receptor

T reg:

Regulatory T cells

T-bet:

T-box transcription factor TBX21

TCR:

T-cell receptor

TEFF:

Effector T cell

TF:

Tissue factor

TGF-β:

Transforming growth factor-beta

TGF-βR:

Transforming growth factor-beta receptor

Th2:

T-helper cell type 2

TIGIT:

T cell immunoreceptor with immunoglobulin and ITIM domain

TKI:

Tyrosine kinase inhibitor

TLR:

Toll-like receptor

TNBC:

Triple-negative breast cancer

TNF:

Tumor necrosis factor

TNFR:

Tumor necrosis factor receptor

TNF-α:

Tumor necrosis factor-alpha

TRUCK:

T cells Redirected for antigen-unrestricted Cytokine-initiated Killing

TSLP:

Thymic stromal lymphopoietin

Tyk2:

Tyrosine kinase 2

UCAR:

Universal chimeric antigen receptor

UCB:

Umbilical cord blood

VSV-G:

Vesicular-stomatitis-virus-G protein

ZAP70-Syk:

Zeta chain of T cell receptor-associated protein kinase 70- Spleen tyrosine kinase

References

  1. Vos T, Lim SS, Abbafati C, Abbas KM, Abbasi M, Abbasifard M, et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the global burden of Disease Study 2019. Lancet. 2020;396(10258):1204–22.

    Article  Google Scholar 

  2. Lankarani KB, Honarvar B, Athari SS. The mechanisms underlying helicobacter pylori-mediated protection against allergic asthma. Tanaffos 2017;16(4):251–259.

  3. Athari SS, Athari SM. The importance of eosinophil, platelet and dendritic cell in asthma. Asian Pac J Trop Dis 2014;4(1):41–47.

  4. Devulder J, Chenivesse C, Ledroit V, Fry S, Lobert P-E, Hober D et al. Aberrant anti-viral response of natural killer cells in severe asthma. Eur Respir J. 2020;55(5).

  5. Dudek RP, Ma Z. CAR T cells for treating severe atopic allergic diseases. Del J Public Health. 2023;9(4):12.

    Article  Google Scholar 

  6. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9(5):503–10.

    Article  CAS  PubMed  Google Scholar 

  7. Erten G, Aktas E, Deniz G. Natural killer cells in allergic inflammation. T cell regulation in Allergy. Asthma Atopic Skin Dis. 2008;94:48–57.

    CAS  Google Scholar 

  8. Mandelboim O, Lieberman N, Lev M, Paul L, Arnon TI, Bushkin Y, et al. Recognition of haemagglutinins on virus-infected cells by NKp46 activates lysis by human NK cells. Nature. 2001;409(6823):1055–60.

    Article  CAS  PubMed  Google Scholar 

  9. Adib-Conquy M, Scott‐Algara D, Cavaillon JM, Souza‐Fonseca‐Guimaraes F. TLR‐mediated activation of NK cells and their role in bacterial/viral immune responses in mammals. Immunol Cell Biol. 2014;92(3):256–62.

    Article  CAS  PubMed  Google Scholar 

  10. Long EO, Sik Kim H, Liu D, Peterson ME, Rajagopalan S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol. 2013;31:227–58.

    Article  CAS  PubMed  Google Scholar 

  11. Caligiuri MA. Human natural killer cells. Blood. J Am Soc Hematol. 2008;112(3):461–9.

    CAS  Google Scholar 

  12. Barnig C, Cernadas M, Dutile S, Liu X, Perrella MA, Kazani S, et al. Lipoxin A4 regulates natural killer cell and type 2 innate lymphoid cell activation in asthma. Sci Transl Med. 2013;5(174):ra17426–26.

    Article  Google Scholar 

  13. Duvall MG, Barnig C, Cernadas M, Ricklefs I, Krishnamoorthy N, Grossman NL, et al. Natural killer cell–mediated inflammation resolution is disabled in severe asthma. Sci Immunol. 2017;2(9):eaam5446.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Korsgren M, Persson CG, Sundler F, Bjerke T, Hansson T, Chambers BJ, et al. Natural killer cells determine development of allergen-induced eosinophilic airway inflammation in mice. J Exp Med. 1999;189(3):553–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Haspeslagh E, Van Helden MJ, Deswarte K, De Prijck S, Van Moorleghem J, Boon L, et al. Role of NKp46 + natural killer cells in house dust mite-driven asthma. EMBO Mol Med. 2018;10(4):e8657.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Arase N, Arase H, Hirano S, Yokosuka T, Sakurai D, Saito T. IgE-mediated activation of NK cells through FcγRIII. J Immunol. 2003;170(6):3054–8.

    Article  CAS  PubMed  Google Scholar 

  17. Zhang B, Yang M, Zhang W, Liu N, Wang D, Jing L, et al. Chimeric antigen receptor-based natural killer cell immunotherapy in cancer: from bench to bedside. Cell Death Dis. 2024;15(1):50.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Adejuwon AO. Redirecting T cell specificity to IgE-expressing B cell for the management of severe allergic asthma. University of Delaware; 2017.

  19. Christie KM, Fay BL, Ma ZM. A novel application of chimeric antigen receptor (CAR) T cells for treating severe allergic diseases. J Immunol. 2021;206(1Supplement):6607–6607.

    Article  Google Scholar 

  20. Jin G, Liu Y, He Z, Wang L, Zhao X, Li Z et al. One infusion of engineered long-lasting and multifunctional T cells cures asthma in mice. bioRxiv. 2023:2023.11. 06.565713.

  21. Chen S, Chen G, Xu F, Sun B, Chen X, Hu W, et al. Treatment of allergic eosinophilic asthma through engineered IL-5-anchored chimeric antigen receptor T cells. Cell Discovery. 2022;8(1):80.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Skuljec J, Chmielewski M, Happle C, Habener A, Busse M, Abken H, et al. Chimeric antigen receptor-redirected regulatory T cells suppress experimental allergic airway inflammation, a model of asthma. Front Immunol. 2017;8:247521.

    Article  Google Scholar 

  23. Nastoupil LJ, Jain MD, Feng L, Spiegel JY, Ghobadi A, Lin Y, 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;38(27):3119.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, Ingaramo M, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015;21(6):581–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhang B-L, Qin D-Y, Mo Z-M, Li Y, Wei W, Wang Y-S, et al. Hurdles of CAR-T cell-based cancer immunotherapy directed against solid tumors. Sci China Life Sci. 2016;59:340–8.

    Article  CAS  PubMed  Google Scholar 

  26. Moscarelli J, Zahavi D, Maynard R, Weiner LM. The next generation of cellular immunotherapy: chimeric antigen receptor-natural killer cells. Transplantation Cell Therapy. 2022;28(10):650–6.

    Article  CAS  Google Scholar 

  27. Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R, et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med. 2020;382(6):545–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Whalen KA, Rakhra K, Mehta NK, Steinle A, Michaelson JS, Baeuerle PA, editors. Engaging natural killer cells for cancer therapy via NKG2D, CD16A and other receptors. Taylor & Francis; 2023. MAbs.

  29. Hu Z, Xu X, Wei H. The adverse impact of tumor microenvironment on NK-cell. Front Immunol. 2021;12:633361.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang Z-H, Li W, Dong H, Han F. Current state of NK cell-mediated immunotherapy in chronic lymphocytic leukemia. Front Oncol. 2023;12:1077436.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Fagan E, Eddleston A. Immunotherapy for cancer: the use of lymphokine activated killer (LAK) cells. Gut. 1987;28(2):113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kuwana Y, Asakura Y, Utsunomiya N, Nakanishi M, Arata Y, Itoh S, et al. Expression of chimeric receptor composed of immunoglobulin-derived V resions and T-cell receptor-derived C regions. Biochem Biophys Res Commun. 1987;149(3):960–8.

    Article  CAS  PubMed  Google Scholar 

  33. Irving BA, Weiss A. The cytoplasmic domain of the T cell receptor ζ chain is sufficient to couple to receptor-associated signal transduction pathways. Cell. 1991;64(5):891–901.

    Article  CAS  PubMed  Google Scholar 

  34. Harding FA, McArthur JG, Gross JA, Raulet DH, Allison JP. CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones. Nature. 1992;356(6370):607–9.

    Article  CAS  PubMed  Google Scholar 

  35. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  37. 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;365(8):725–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Liu D, Zhao J, Song Y. Engineering switchable and programmable universal CARs for CAR T therapy. J Hematol Oncol. 2019;12:1–9.

    Article  Google Scholar 

  40. Liu Q, Li J, Zheng H, Yang S, Hua Y, Huang N, et al. Adoptive cellular immunotherapy for solid neoplasms beyond CAR-T. Mol Cancer. 2023;22(1):28.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Esmaeilzadeh A, Tahmasebi S, Athari SS. Chimeric antigen receptor -T cell therapy: Applications and challenges in treatment of allergy and asthma. Biomed Pharmacother. 2020;123:109685.

  42. Zhang P, Zhang G, Wan X. Challenges and new technologies in adoptive cell therapy. J Hematol Oncol. 2023;16(1):97.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Liu X, Jiang S, Fang C, Yang S, Olalere D, Pequignot EC, et al. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res. 2015;75(17):3596–607.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen J, Qiu S, Li W, Wang K, Zhang Y, Yang H, et al. Tuning charge density of chimeric antigen receptor optimizes tonic signaling and CAR-T cell fitness. Cell Res. 2023;33(5):341–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Grada Z, Hegde M, Byrd T, Shaffer DR, Ghazi A, Brawley VS et al. TanCAR: a novel bispecific chimeric antigen receptor for cancer immunotherapy. Mol Therapy-Nucleic Acids. 2013;2.

  46. Katsarou A, Sjöstrand M, Naik J, Mansilla-Soto J, Kefala D, Kladis G, et al. Combining a CAR and a chimeric costimulatory receptor enhances T cell sensitivity to low antigen density and promotes persistence. Sci Transl Med. 2021;13(623):eabh1962.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood cancer J. 2021;11(4):69.

    Article  PubMed  PubMed Central  Google Scholar 

  48. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hirabayashi K, Du H, Xu Y, Shou P, Zhou X, Fucá G, et al. Dual-targeting CAR-T cells with optimal co-stimulation and metabolic fitness enhance antitumor activity and prevent escape in solid tumors. Nat cancer. 2021;2(9):904–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Li Y, Hermanson DL, Moriarity BS, Kaufman DS. Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell. 2018;23(2):181–92. e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang H, Li F, Cao J, Wang X, Cheng H, Qi K, et al. A chimeric antigen receptor with antigen-independent OX40 signaling mediates potent antitumor activity. Sci Transl Med. 2021;13(578):eaba7308.

    Article  CAS  PubMed  Google Scholar 

  52. Kawalekar OU, O’Connor RS, Fraietta JA, Guo L, McGettigan SE, Posey AD, et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. 2016;44(2):380–90.

    Article  CAS  PubMed  Google Scholar 

  53. Mehta RS, Rezvani K. Chimeric antigen receptor expressing natural killer cells for the immunotherapy of cancer. Front Immunol. 2018;9:330897.

    Article  Google Scholar 

  54. Wang G, Wang W. Advanced cell therapies for glioblastoma. Front Immunol. 2022;13:904133.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Circosta P, Donini C, Gallo S, Giraudo L, Gammaitoni L, Rotolo R, et al. Full chimaeric CAR. CIK from patients engrafted after allogeneic haematopoietic cell transplant: Feasibility, anti-leukaemic potential and alloreactivity across major human leukocyte antigen barriers. Br J Haematol. 2023;200(1):64–9.

    Article  CAS  PubMed  Google Scholar 

  56. Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020;38(8):947–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Müller S, Bexte T, Gebel V, Hartmann J, Schambach A, Modlich U, et al. High cytotoxic efficiency of lentivirally and alpharetrovirally engineered CD19-specific chimeric antigen receptor natural killer cells against acute lymphoblastic leukemia. Front Immunol. 2020;10:485159.

    Article  Google Scholar 

  58. Watanabe N, Mo F, McKenna MK. Impact of manufacturing procedures on CAR T cell functionality. Front Immunol. 2022;13:876339.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Stock S, Schmitt M, Sellner L. Optimizing manufacturing protocols of chimeric antigen receptor T cells for improved anticancer immunotherapy. Int J Mol Sci. 2019;20(24):6223.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Smith C, Corvino D, Beagley L, Rehan S, Neller MA, Crooks P, et al. T cell repertoire remodeling following post-transplant T cell therapy coincides with clinical response. J Clin Investig. 2019;129(11):5020–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Massey J, Jackson K, Singh M, Withers B, Hendrawan K, Zaunders J, et al. Haematopoietic stem cell transplantation results in extensive remodelling of the clonal T cell repertoire in multiple sclerosis. Front Immunol. 2022;13:798300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lonez C, Breman E, Allogeneic CAR-T, Therapy. Technologies: Has Promise Been Met? Cells. 2024;13(2):146.

    CAS  PubMed  Google Scholar 

  63. Strauss-Albee DM, Fukuyama J, Liang EC, Yao Y, Jarrell JA, Drake AL, et al. Human NK cell repertoire diversity reflects immune experience and correlates with viral susceptibility. Sci Transl Med. 2015;7(297):ra297115–297115.

    Article  Google Scholar 

  64. Marofi F, Saleh MM, Rahman HS, Suksatan W, Al-Gazally ME, Abdelbasset WK, et al. CAR-engineered NK cells; a promising therapeutic option for treatment of hematological malignancies. Stem Cell Res Ther. 2021;12(1):374.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Zhang Y, Zhou W, Yang J, Yang J, Wang W. Chimeric antigen receptor engineered natural killer cells for cancer therapy. Experimental Hematol Oncol. 2023;12(1):70.

    Article  CAS  Google Scholar 

  66. Simonetta F, Alvarez M, Negrin RS. Natural killer cells in graft-versus-host-disease after allogeneic hematopoietic cell transplantation. Front Immunol. 2017;8:258411.

    Article  Google Scholar 

  67. Heipertz EL, Zynda ER, Stav-Noraas TE, Hungler AD, Boucher SE, Kaur N, et al. Current perspectives on off-the-shelf allogeneic NK and CAR-NK cell therapies. Front Immunol. 2021;12:732135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lamers-Kok N, Panella D, Georgoudaki A-M, Liu H, Özkazanc D, Kučerová L, et al. Natural killer cells in clinical development as non-engineered, engineered, and combination therapies. J Hematol Oncol. 2022;15(1):164.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Chen Z, Hu Y, Mei H. Advances in CAR-Engineered Immune Cell Generation: Engineering approaches and Sourcing strategies. Adv Sci. 2023;10(35):2303215.

    Article  Google Scholar 

  70. Liu D-D, Hong W-C, Qiu K-Y, Li X-Y, Liu Y, Zhu L-W, et al. Umbilical cord blood: a promising source for allogeneic CAR-T cells. Front Oncol. 2022;12:944248.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. 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. 2024:1–13.

  72. Yu T, Luo C. Cord blood-derived CD19-specific chimeric antigen receptor T cells: an off-the-shelf promising therapeutic option for treatment of diffuse large B-cell lymphoma. Front Immunol. 2023;14:1139482.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Wang J, Metheny L. Umbilical cord blood derived cellular therapy: advances in clinical development. Front Oncol. 2023;13:1167266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Sabbah M, Jondreville L, Lacan C, Norol F, Vieillard V, Roos-Weil D, et al. CAR-NK cells: a chimeric hope or a promising therapy? Cancers. 2022;14(15):3839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Liu E, Tong Y, Dotti G, Shaim H, Savoldo B, Mukherjee M, et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia. 2018;32(2):520–31.

    Article  CAS  PubMed  Google Scholar 

  76. Caël B, Galaine J, Bardey I, Marton C, Fredon M, Biichle S, et al. Umbilical cord blood as a source of less differentiated T cells to produce CD123 CAR-T cells. Cancers. 2022;14(13):3168.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Brunstein CG, Miller JS, McKenna DH, Hippen KL, DeFor TE, Sumstad D, et al. Umbilical cord blood–derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical effect. Blood J Am Soc Hematol. 2016;127(8):1044–51.

    CAS  Google Scholar 

  78. Sarvaria A, Jawdat D, Madrigal JA, Saudemont A. Umbilical cord blood natural killer cells, their characteristics, and potential clinical applications. Front Immunol. 2017;8:255264.

    Article  Google Scholar 

  79. Klingemann H. The NK-92 cell line—30 years later: its impact on natural killer cell research and treatment of cancer. Cytotherapy. 2023;25(5):451–7.

    Article  CAS  PubMed  Google Scholar 

  80. Suck G, Odendahl M, Nowakowska P, Seidl C, Wels WS, Klingemann HG, et al. NK-92: an ‘off-the-shelf therapeutic’for adoptive natural killer cell-based cancer immunotherapy. Cancer Immunol Immunother. 2016;65:485–92.

    Article  CAS  PubMed  Google Scholar 

  81. Guo F, Zhang Y, Cui J. Manufacturing CAR-NK against tumors: who is the ideal supplier? Chin J Cancer Res. 2024;36(1):1.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Yilmaz A, Cui H, Caligiuri MA, Yu J. Chimeric antigen receptor-engineered natural killer cells for cancer immunotherapy. J Hematol Oncol. 2020;13:1–22.

    Article  Google Scholar 

  83. Shankar K, Capitini CM, Saha K. Genome engineering of induced pluripotent stem cells to manufacture natural killer cell therapies. Stem Cell Res Ther. 2020;11:1–14.

    Article  Google Scholar 

  84. Zhang J, Zheng H, Diao Y. Natural killer cells and current applications of chimeric antigen receptor-modified NK-92 cells in tumor immunotherapy. Int J Mol Sci. 2019;20(2):317.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Morgan MA, Büning H, Schambach A. Use of cell and genome modification technologies to generate improved off-the-shelf CAR T and CAR NK cells. Front Immunol. 2020;11:557760.

    Article  Google Scholar 

  86. Lin H, Cheng J, Mu W, Zhou J, Zhu L. Advances in universal CAR-T cell therapy. Front Immunol. 2021;12:744823.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kang S, Gao X, Zhang L, Yang E, Li Y, Yu L. The advances and challenges of NK cell-based cancer immunotherapy. Curr Oncol. 2021;28(2):1077–93.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Zhang L, Meng Y, Feng X, Han Z. CAR-NK cells for cancer immunotherapy: from bench to bedside. Biomark Res. 2022;10(1):12.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Fang F, Wang W, Chen M, Tian Z, Xiao W. Technical advances in NK cell-based cellular immunotherapy. Cancer Biology Med. 2019;16(4):647.

    Article  CAS  Google Scholar 

  90. Wang Z, McWilliams-Koeppen HP, Reza H, Ostberg JR, Chen W, Wang X, et al. 3D-organoid culture supports differentiation of human CAR + iPSCs into highly functional CAR T cells. Cell Stem Cell. 2022;29(4):515–27. e8.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76.

    Article  CAS  PubMed  Google Scholar 

  92. Choi K-D, Yu J, Smuga-Otto K, Salvagiotto G, Rehrauer W, Vodyanik M, et al. Hematopoietic and endothelial differentiation of human induced pluripotent stem cells. Stem Cells. 2009;27(3):559–67.

    Article  CAS  PubMed  Google Scholar 

  93. Knorr DA, Ni Z, Hermanson D, Hexum MK, Bendzick L, Cooper LJ, et al. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Translational Med. 2013;2(4):274–83.

    Article  CAS  Google Scholar 

  94. Zhu H, Kaufman DS. An improved method to produce clinical-scale natural killer cells from human pluripotent stem cells. Springer; 2019.

  95. Cichocki F, Bjordahl R, Gaidarova S, Mahmood S, Abujarour R, Wang H, et al. iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti–PD-1 therapy. Sci Transl Med. 2020;12(568):eaaz5618.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Lin X, Sun Y, Dong X, Liu Z, Sugimura R, Xie G. IPSC-derived CAR-NK cells for cancer immunotherapy. Biomed Pharmacother. 2023;165:115123.

    Article  CAS  PubMed  Google Scholar 

  97. Xue D, Lu S, Zhang H, Zhang L, Dai Z, Kaufman DS, et al. Induced pluripotent stem cell-derived engineered T cells, natural killer cells, macrophages, and dendritic cells in immunotherapy. Trends Biotechnol. 2023;41(7):907–22.

    Article  CAS  PubMed  Google Scholar 

  98. Merkle FT, Ghosh S, Kamitaki N, Mitchell J, Avior Y, Mello C, et al. Human pluripotent stem cells recurrently acquire and expand dominant negative P53 mutations. Nature. 2017;545(7653):229–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Zhu H, Blum RH, Bjordahl R, Gaidarova S, Rogers P, Lee TT, et al. Pluripotent stem cell–derived NK cells with high-affinity noncleavable CD16a mediate improved antitumor activity. Blood J Am Soc Hematol. 2020;135(6):399–410.

    Google Scholar 

  100. Pang Z, Wang Z, Li F, Feng C, Mu X. Current progress of CAR-NK therapy in cancer treatment. Cancers. 2022;14(17):4318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Michels A, Ho N, Buchholz CJ. Precision medicine: in vivo CAR therapy as a showcase for receptor-targeted vector platforms. Mol Ther. 2022;30(7):2401–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Schmidt P, Raftery MJ, Pecher G. Engineering NK cells for CAR therapy—recent advances in gene transfer methodology. Front Immunol. 2021;11:611163.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Wang W, Liu Y, He Z, Li L, Liu S, Jiang M, et al. Breakthrough of solid tumor treatment: CAR-NK immunotherapy. Cell Death Discovery. 2024;10(1):40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Li Y, Huo Y, Yu L, Wang J. Quality control and nonclinical research on CAR-T cell products: general principles and key issues. Engineering. 2019;5(1):122–31.

    Article  CAS  Google Scholar 

  105. Dai X, Mei Y, Cai D, Han W. Standardizing CAR-T therapy: getting it scaled up. Biotechnol Adv. 2019;37(1):239–45.

    Article  CAS  PubMed  Google Scholar 

  106. Tumaini B, Lee DW, Lin T, Castiello L, Stroncek DF, Mackall C, et al. Simplified process for the production of anti–CD19-CAR–engineered T cells. Cytotherapy. 2013;15(11):1406–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Committee GIAE, editor. Global strategy for asthma management and prevention (revised 2002). NHLBI/WHO Workshop Report; 2006, GINA; 2006.

  108. Talay O, Yan D, Brightbill HD, Straney EE, Zhou M, Ladi E, et al. IgE + memory B cells and plasma cells generated through a germinal-center pathway. Nat Immunol. 2012;13(4):396.

    Article  CAS  PubMed  Google Scholar 

  109. Wu LC, Zarrin AA. The production and regulation of IgE by the immune system. Nat Rev Immunol. 2014;14(4):247–59.

    Article  CAS  PubMed  Google Scholar 

  110. Krogsgaard M, Prado N, Adams EJ, He X-l, Chow D-C, Wilson DB, et al. Evidence that structural rearrangements and/or flexibility during TCR binding can contribute to T cell activation. Mol Cell. 2003;12(6):1367–78.

    Article  CAS  PubMed  Google Scholar 

  111. Kraft S, Kinet J-P. New developments in FcεRI regulation, function and inhibition. Nat Rev Immunol. 2007;7(5):365–78.

    Article  CAS  PubMed  Google Scholar 

  112. Katona IM, Urban J Jr, Scher I, Kanellopoulos-Langevin C, Finkelman F. Induction of an IgE response in mice by Nippostrongylus brasiliensis: characterization of lymphoid cells with intracytoplasmic or surface IgE. 1983.

  113. Geha RS, Jabara HH, Brodeur SR. The regulation of immunoglobulin E class-switch recombination. Nat Rev Immunol. 2003;3(9):721–32.

    Article  CAS  PubMed  Google Scholar 

  114. Maeda K, Burton G, Padgett DA, Conrad DH, Huff T, Masuda A et al. Murine follicular dendritic cells and low affinity Fc receptors for IgE (Fc epsilon RII). Journal of immunology (Baltimore, Md: 1950). 1992;148(8):2340-7.

  115. Brusselle GG, Koppelman GH. Biologic therapies for severe asthma. N Engl J Med. 2022;386(2):157–71.

    Article  CAS  PubMed  Google Scholar 

  116. Klion AD, Rothenberg ME. Advances in eosinophilic diseases in 2018. J Allergy Clin Immunol. 2019;144(6):1490–4.

    Article  PubMed  Google Scholar 

  117. Strickland DH, Holt PG. T regulatory cells in childhood asthma. Trends Immunol. 2011;32(9):420–7.

    Article  CAS  PubMed  Google Scholar 

  118. Moss RB. Severe fungal asthma: a role for biologics and inhaled antifungals. J Fungi. 2023;9(1):85.

    Article  CAS  Google Scholar 

  119. Godfrey DI, Pellicci DG, Rossjohn J. NKT cells: the smoking gun in fungal-induced asthma? Nat Med. 2013;19(10):1210–1.

    Article  CAS  PubMed  Google Scholar 

  120. Di Pietro R, Dubuc V, Manguin E, Giroux-Lafond R, Bédard C, Boivin R et al. Characterization of fungal exposure and dectin-1 expression in healthy horses and horses with severe asthma. Am J Vet Res. 2022;83(6).

  121. Brown GD. Dectin-1: a signalling non-TLR pattern-recognition receptor. Nat Rev Immunol. 2006;6(1):33–43.

    Article  CAS  PubMed  Google Scholar 

  122. Seif M, Kakoschke TK, Ebel F, Bellet MM, Trinks N, Renga G, et al. CAR T cells targeting aspergillus fumigatus are effective at treating invasive pulmonary aspergillosis in preclinical models. Sci Transl Med. 2022;14(664):eabh1209.

    Article  CAS  PubMed  Google Scholar 

  123. Kumaresan PR, Manuri PR, Albert ND, Maiti S, Singh H, Mi T et al. Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection. Proceedings of the National Academy of Sciences. 2014;111(29):10660-5.

  124. Myers JA, Miller JS. Exploring the NK cell platform for cancer immunotherapy. Nat Reviews Clin Oncol. 2021;18(2):85–100.

    Article  Google Scholar 

  125. Hamilton G, Plangger A. The impact of NK cell-based therapeutics for the treatment of lung cancer for biologics: targets and therapy. Biologics: Targets and Therapy. 2021:265 – 77.

  126. Lepretre F, Gras D, Chanez P, Duez C. Natural killer cells in the lung: potential role in asthma and virus-induced exacerbation? Eur Respiratory Rev. 2023;32(169).

  127. Zhang H, Wang J, Li F. Modulation of natural killer cell exhaustion in the lungs: the key components from lung microenvironment and lung tumor microenvironment. Front Immunol. 2023;14:1286986.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. van de Deniz G, Akdis M. Natural killer cells in patients with allergic diseases. J Allergy Clin Immunol. 2013;132(3):527–35.

    Article  CAS  PubMed  Google Scholar 

  129. Fauriat C, Long EO, Ljunggren H-G, Bryceson YT. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood J Am Soc Hematol. 2010;115(11):2167–76.

    CAS  Google Scholar 

  130. Li T, Niu M, Zhang W, Qin S, Zhou J, Yi M. CAR-NK cells for cancer immunotherapy: recent advances and future directions. Front Immunol. 2024;15:1361194.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Diwanji N, Getts D, Wang Y. Chimeric Antigen cytotoxic receptors for in vivo Engineering of Tumor-Targeting NK cells. ImmunoHorizons. 2024;8(1):97–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Malmberg K-J, Carlsten M, Björklund A, Sohlberg E, Bryceson YT, Ljunggren H-G, editors. Natural killer cell-mediated immunosurveillance of human cancer. Seminars in immunology. Elsevier; 2017.

  133. Raulet DH, Vance RE, McMahon CW. Regulation of the natural killer cell receptor repertoire. Annu Rev Immunol. 2001;19(1):291–330.

    Article  CAS  PubMed  Google Scholar 

  134. Sivori S, Vacca P, Del Zotto G, Munari E, Mingari MC, Moretta L. Human NK cells: surface receptors, inhibitory checkpoints, and translational applications. Cell Mol Immunol. 2019;16(5):430–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Kumar S. Natural killer cell cytotoxicity and its regulation by inhibitory receptors. Immunology. 2018;154(3):383–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Champsaur M, Lanier LL. Effect of NKG2D ligand expression on host immune responses. Immunol Rev. 2010;235(1):267–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Siemaszko J, Marzec-Przyszlak A, Bogunia-Kubik K. NKG2D natural killer cell receptor—a short description and potential clinical applications. Cells. 2021;10(6):1420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Biron CA, Nguyen KB, Pien GC, Cousens LP, Salazar-Mather TP. Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annu Rev Immunol. 1999;17(1):189–220.

    Article  CAS  PubMed  Google Scholar 

  139. Schuijs MJ, Png S, Richard AC, Tsyben A, Hamm G, Stockis J, et al. ILC2-driven innate immune checkpoint mechanism antagonizes NK cell antimetastatic function in the lung. Nat Immunol. 2020;21(9):998–1009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331(6013):44–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Degli-Esposti MA, Smyth MJ. Close encounters of different kinds: dendritic cells and NK cells take centre stage. Nat Rev Immunol. 2005;5(2):112–24.

    Article  CAS  PubMed  Google Scholar 

  142. Ortaldo JR, Winkler-Pickett R, Wigginton J, Horner M, Bere EW, Mason AT, et al. Regulation of ITAM-positive receptors: role of IL-12 and IL-18. Blood. 2006;107(4):1468–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Walwyn-Brown K, Guldevall K, Saeed M, Pende D, Önfelt B, MacDonald AS, et al. Human NK cells lyse Th2-polarizing dendritic cells via NKp30 and DNAM-1. J Immunol. 2018;201(7):2028–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Awad A, Yassine H, Barrier M, Vorng H, Marquillies P, Tsicopoulos A, et al. Natural killer cells induce eosinophil activation and apoptosis. PLoS ONE. 2014;9(4):e94492.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Poirot A, Wacht G, Christine L, Saas P, Frossard N, Geny B et al. NK cells and lipoxin A4 promote resolution of eosinophilic inflammation after nasal allergen challenge. Authorea Preprints. 2021.

  146. Thorén FB, Riise RE, Ousbäck J, Della Chiesa M, Alsterholm M, Marcenaro E, et al. Human NK cells induce neutrophil apoptosis via an NKp46-and Fas-dependent mechanism. J Immunol. 2012;188(4):1668–74.

    Article  PubMed  Google Scholar 

  147. Jaeger BN, Donadieu J, Cognet C, Bernat C, Ordoñez-Rueda D, Barlogis V, et al. Neutrophil depletion impairs natural killer cell maturation, function, and homeostasis. J Exp Med. 2012;209(3):565–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Marquardt N, Kekäläinen E, Chen P, Kvedaraite E, Wilson JN, Ivarsson MA, et al. Human lung natural killer cells are predominantly comprised of highly differentiated hypofunctional CD69 – CD56dim cells. J Allergy Clin Immunol. 2017;139(4):1321–30. e4.

    Article  CAS  PubMed  Google Scholar 

  149. Bozward AG, Warricker F, Oo YH, Khakoo SI. Natural killer cells and regulatory T cells cross talk in hepatocellular carcinoma: exploring therapeutic options for the next decade. Front Immunol. 2021;12:643310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Geiger TL, Sun JC. Development and maturation of natural killer cells. Curr Opin Immunol. 2016;39:82–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Dogra P, Rancan C, Ma W, Toth M, Senda T, Carpenter DJ, et al. Tissue determinants of human NK cell development, function, and residence. Cell. 2020;180(4):749–63. e13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Paul S, Lal G. The molecular mechanism of natural killer cells function and its importance in cancer immunotherapy. Front Immunol. 2017;8:290728.

    Article  Google Scholar 

  153. Del Vecchio F, Martinez-Rodriguez V, Schukking M, Cocks A, Broseghini E, Fabbri M. Professional killers: the role of extracellular vesicles in the reciprocal interactions between natural killer, CD8 + cytotoxic T‐cells and tumour cells. J Extracell Vesicles. 2021;10(6):e12075.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Sun H, Liu L, Huang Q, Liu H, Huang M, Wang J, et al. Accumulation of tumor-infiltrating CD49a + NK cells correlates with poor prognosis for human hepatocellular carcinoma. Cancer Immunol Res. 2019;7(9):1535–46.

    Article  CAS  PubMed  Google Scholar 

  155. Mazinani M, Rahbarizadeh F. New cell sources for CAR-based immunotherapy. Biomark Res. 2023;11(1):49.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Abbas AK, Benoist C, Bluestone JA, Campbell DJ, Ghosh S, Hori S, et al. Regulatory T cells: recommendations to simplify the nomenclature. Nat Immunol. 2013;14(4):307–8.

    Article  CAS  PubMed  Google Scholar 

  157. Wu S-Y, Fu T, Jiang Y-Z, Shao Z-M. Natural killer cells in cancer biology and therapy. Mol Cancer. 2020;19:1–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Malhotra D, Burrack KS, Jenkins MK, Frosch AE. Antigen-specific CD4 + T cells exhibit distinct kinetic and phenotypic patterns during primary and secondary responses to infection. Front Immunol. 2020;11:553193.

    Article  Google Scholar 

  159. Sun JC, Lanier LL. NK cell development, homeostasis and function: parallels with CD8 + T cells. Nat Rev Immunol. 2011;11(10):645–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Fang F, Xie S, Chen M, Li Y, Yue J, Ma J, et al. Advances in NK cell production. Cell Mol Immunol. 2022;19(4):460–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Zhu H, Blum RH, Bernareggi D, Ask EH, Wu Z, Hoel HJ, et al. Metabolic reprograming via deletion of CISH in human iPSC-derived NK cells promotes in vivo persistence and enhances anti-tumor activity. Cell Stem Cell. 2020;27(2):224–37. e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Dixon KJ, Wu J, Walcheck B. Engineering anti-tumor monoclonal antibodies and fc receptors to enhance ADCC by human NK cells. Cancers. 2021;13(2):312.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. van de Komlósi ZI, Kovács N, Szűcs G, Sokolowska M, O’Mahony L, et al. Cellular and molecular mechanisms of allergic asthma. Mol Aspects Med. 2022;85:100995.

    Article  PubMed  Google Scholar 

  164. Frank K, Paust S. Dynamic natural killer cell and T cell responses to influenza infection. Front Cell Infect Microbiol. 2020;10:425.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295(5562):2097–100.

    Article  CAS  PubMed  Google Scholar 

  166. Lupo KB, Matosevic S. Natural killer cells as allogeneic effectors in adoptive cancer immunotherapy. Cancers. 2019;11(6):769.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Quatrini L, Della Chiesa M, Sivori S, Mingari MC, Pende D, Moretta L. Human NK cells, their receptors and function. Eur J Immunol. 2021;51(7):1566–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Wang X, Huang L, Wen X, Li D, Yang G, Zheng J. Altered NCR3 splice variants may result in deficient NK cell function in renal cell carcinoma patients. vivo. 2024;38(1):174–83.

    Article  CAS  Google Scholar 

  169. Moretta L, Moretta A. Unravelling natural killer cell function: triggering and inhibitory human NK receptors. EMBO J. 2004;23(2):255–9.

    Article  CAS  PubMed  Google Scholar 

  170. Pinheiro PF, Justino GC, Marques MM. NKp30-A prospective target for new cancer immunotherapy strategies. Br J Pharmacol. 2020;177(20):4563–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Deniz G, Akdis M. NK cell subsets and their role in allergy. Expert Opin Biol Ther. 2011;11(7):833–41.

    Article  CAS  PubMed  Google Scholar 

  172. Kinlein A, Janes ME, Kincer J, Almeida T, Matz H, Sui J, et al. Analysis of shark NCR3 family genes reveals primordial features of vertebrate NKp30. Immunogenetics. 2021;73(4):333–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Mantovani S, Oliviero B, Lombardi A, Varchetta S, Mele D, Sangiovanni A, et al. Deficient natural killer cell NKp30-mediated function and altered NCR3 splice variants in hepatocellular carcinoma. Hepatology. 2019;69(3):1165–79.

    Article  CAS  PubMed  Google Scholar 

  174. Delahaye NF, Rusakiewicz S, Martins I, Ménard C, Roux S, Lyonnet L, et al. Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat Med. 2011;17(6):700–7.

    Article  CAS  PubMed  Google Scholar 

  175. Medjouel Khlifi H, Guia S, Vivier E, Narni-Mancinelli E. Role of the ITAM-bearing receptors expressed by natural killer cells in cancer. Front Immunol. 2022;13:898745.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Lanier LL. Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol. 2008;9(5):495–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Duvall M, Chiha T, Reilly R, Ramdour S, Krishnamoorthy N, Cagnina R, BIOLOGY TO THE REAL WORLD IN ASTHMA AND ALLERGIC INFLAMMATION. Circulating natural killer cell immunophenotypes correlate with severe Asthma Clinical traits. A101 LINKING. American Thoracic Society; 2023. pp. A2554–A.

  178. Kim JH, Jang YJ. Role of natural killer cells in airway inflammation. Allergy Asthma Immunol Res. 2018;10(5):448.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Elhaik Goldman S, Moshkovits I, Shemesh A, Filiba A, Tsirulsky Y, Vronov E, et al. Natural killer receptor 1 dampens the development of allergic eosinophilic airway inflammation. PLoS ONE. 2016;11(8):e0160779.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Diwanji N, Getts D, Wang Y. In vivo programming of natural killer cells and T cells using mRNA delivered cytotoxic chimeric antigen receptors. Cancer Res. 2024;84(6Supplement):1319.

    Article  Google Scholar 

  181. Kasahara Y, Shin C, Kubo N, Mihara K, Iwabuchi H, Takachi T, et al. Development and characterisation of NKp44-based chimeric antigen receptors that confer T cells with NK cell‐like specificity. Clin Translational Immunol. 2020;9(7):e1147.

    Article  CAS  Google Scholar 

  182. Ng Y-Y, Tay JC, Li Z, Wang J, Zhu J, Wang S. T cells expressing NKG2D CAR with a DAP12 signaling domain stimulate lower cytokine production while effective in tumor eradication. Mol Ther. 2021;29(1):75–85.

    Article  CAS  PubMed  Google Scholar 

  183. Xiao L, Cen D, Gan H, Sun Y, Huang N, Xiong H, et al. Adoptive transfer of NKG2D CAR mRNA-engineered natural killer cells in colorectal cancer patients. Mol Ther. 2019;27(6):1114–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Pereira BI, De Maeyer RP, Covre LP, Nehar-Belaid D, Lanna A, Ward S, et al. Sestrins induce natural killer function in senescent-like CD8 + T cells. Nat Immunol. 2020;21(6):684–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL, et al. An activating immunoreceptor complex formed by NKG2D and DAP10. Science. 1999;285(5428):730–2.

    Article  CAS  PubMed  Google Scholar 

  186. JL U. KG2D-mediated signaling requires a DAP10-bound Grb2-Vavl intermediate and phosphatidylinositol-3-kinase in human natural killer ceils. Nat Immunol. 2006;7:524–32.

  187. Diefenbach A, Tomasello E, Lucas M, Jamieson AM, Hsia JK, Vivier E, et al. Selective associations with signaling proteins determine stimulatory versus costimulatory activity of NKG2D. Nat Immunol. 2002;3(12):1142–9.

    Article  CAS  PubMed  Google Scholar 

  188. Gilfillan S, Ho EL, Cella M, Yokoyama WM, Colonna M. NKG2D recruits two distinct adapters to trigger NK cell activation and costimulation. Nat Immunol. 2002;3(12):1150–5.

    Article  CAS  PubMed  Google Scholar 

  189. Nie F, Chen Y, Hu Y, Huang P, Shi X, Cai J et al. TREM1/DAP12 based novel multiple chain CAR-T cells targeting DLL3 show robust anti‐tumour efficacy for small cell lung cancer. Immunology. 2024.

  190. Yu T, Nie F-Q, Zhang Q, Yu S-K, Zhang M-L, Wang Q, et al. Effects of methionine deficiency on B7H3-DAP12-CAR-T cells in the treatment of lung squamous cell carcinoma. Cell Death Dis. 2024;15(1):12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Li S, Zhao R, Zheng D, Qin L, Cui Y, Li Y, et al. DAP10 integration in CAR-T cells enhances the killing of heterogeneous tumors by harnessing endogenous NKG2D. Mol Therapy-Oncolytics. 2022;26:15–26.

    Article  CAS  Google Scholar 

  192. Wilton KM, Overlee BL, Billadeau DD. NKG2D–DAP10 signaling recruits EVL to the cytotoxic synapse to generate F-actin and promote NK cell cytotoxicity. J Cell Sci. 2020;133(5):jcs230508.

    Article  CAS  Google Scholar 

  193. Peng Y, Zhang W, Chen Y, Zhang L, Shen H, Wang Z, et al. Engineering c-Met-CAR NK-92 cells as a promising therapeutic candidate for lung adenocarcinoma. Pharmacol Res. 2023;188:106656.

    Article  CAS  PubMed  Google Scholar 

  194. Jackson D, Walum J, Banerjee P, Lewis BW, Prakash Y, Sathish V, et al. Th1 cytokines synergize to change gene expression and promote corticosteroid insensitivity in pediatric airway smooth muscle. Respir Res. 2022;23(1):126.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Branchett WJ, Stölting H, Oliver RA, Walker SA, Puttur F, Gregory LG, et al. AT cell–myeloid IL-10 axis regulates pathogenic IFN-γ–dependent immunity in a mouse model of type 2–low asthma. J Allergy Clin Immunol. 2020;145(2):666–78. e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Schülke S. Induction of interleukin-10 producing dendritic cells as a tool to suppress allergen-specific T helper 2 responses. Front Immunol. 2018;9:347534.

    Article  Google Scholar 

  197. Berti FCB, Pereira APL, Cebinelli GCM, Trugilo KP, de Oliveira KB. The role of interleukin 10 in human papilloma virus infection and progression to cervical carcinoma. Cytokine Growth Factor Rev. 2017;34:1–13.

    Article  CAS  PubMed  Google Scholar 

  198. Mohseni YR, Saleem A, Tung SL, Dudreuilh C, Lang C, Peng Q, et al. Chimeric antigen receptor-modified human regulatory T cells that constitutively express IL‐10 maintain their phenotype and are potently suppressive. Eur J Immunol. 2021;51(10):2522–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Porro C, Cianciulli A, Panaro MA. The regulatory role of IL-10 in neurodegenerative diseases. Biomolecules. 2020;10(7):1017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Cheng SB, Sharma S. Interleukin-10: a pleiotropic regulator in pregnancy. Am J Reprod Immunol. 2015;73(6):487–500.

    Article  CAS  PubMed  Google Scholar 

  201. Hendriks RW. Interleukin-10 multitasking in allergic airway inflammation. Cell Mol Immunol. 2023;20(12):1530–2.

    Article  CAS  PubMed  Google Scholar 

  202. Branchett WJ, Lloyd CM. Regulatory cytokine function in the respiratory tract. Mucosal Immunol. 2019;12(3):589–600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Campbell JD, Buckland KF, McMillan SJ, Kearley J, Oldfield WL, Stern LJ, et al. Peptide immunotherapy in allergic asthma generates IL-10–dependent immunological tolerance associated with linked epitope suppression. J Exp Med. 2009;206(7):1535–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Gupta A, Dimeloe S, Richards DF, Chambers ES, Black C, Urry Z, et al. Defective IL-10 expression and in vitro steroid-induced IL-17A in paediatric severe therapy-resistant asthma. Thorax. 2014;69(6):508–15.

    Article  PubMed  Google Scholar 

  205. da Silva C, Peres Leal M, Brochetti RA, Braga T, Vitoretti LB, Saraiva Camara NO et al. Low level laser therapy reduces the development of lung inflammation induced by formaldehyde exposure. PloS one. 2015;10(11):e0142816.

  206. de Brito AA, Gonçalves Santos T, da Palma RK, Ligeiro, de Oliveira AP. Photobiomodulation therapy restores IL-10 secretion in a murine model of chronic asthma: relevance to the population of CD4 + CD25 + Foxp3 + cells in lung. Frontiers in Immunology. 2022;12:789426.

  207. Golebski K, Layhadi JA, Sahiner U, Steveling-Klein EH, Lenormand MM, Li RC, et al. Induction of IL-10-producing type 2 innate lymphoid cells by allergen immunotherapy is associated with clinical response. Immunity. 2021;54(2):291–307. e7.

    Article  CAS  PubMed  Google Scholar 

  208. Barnes PJ. Endogenous inhibitory mechanisms in asthma. Am J Respir Crit Care Med. 2000;161(supplement2):S176–81.

    Article  CAS  PubMed  Google Scholar 

  209. Zhao Y, Chen J, Andreatta M, Feng B, Xie Y-Q, Wenes M et al. IL-10-expressing CAR T cells resist dysfunction and mediate durable clearance of solid tumors and metastases. Nat Biotechnol. 2024:1–12.

  210. Chen N, Xu Y, Mou J, Rao Q, Xing H, Tian Z, et al. Targeting of IL-10R on acute myeloid leukemia blasts with chimeric antigen receptor-expressing T cells. Blood Cancer J. 2021;11(8):144.

    Article  PubMed  PubMed Central  Google Scholar 

  211. Campbell KS, Purdy AK. Structure/function of human killer cell immunoglobulin-like receptors: lessons from polymorphisms, evolution, crystal structures and mutations. Immunology. 2011;132(3):315–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Debska-Zielkowska J, Moszkowska G, Zielinski M. KIR Receptors as key regulators of NK cells activity in Health and Disease. Cells. 2021;10(7):1777. Published 2021 Jul 14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Mahaweni NM, Ehlers FA, Bos GM, Wieten L. Tuning natural killer cell anti-multiple myeloma reactivity by targeting inhibitory signaling via KIR and NKG2A. Front Immunol. 2018;9:427229.

    Article  Google Scholar 

  214. Mohammad Taheri M, Javan F, Poudineh M, Athari SS. CAR-NKT cells in asthma: Use of NKT as a promising cell for CAR therapy. Clinical Reviews in Allergy & Immunology. https://doi.org/10.1007/s12016-024-08998-0.

  215. Rajagopalan S, Fu J, Long EO. Cutting edge: induction of IFN-γ production but not cytotoxicity by the killer cell Ig-like receptor KIR2DL4 (CD158d) in resting NK cells. J Immunol. 2001;167(4):1877–81.

    Article  CAS  PubMed  Google Scholar 

  216. Goodridge JP, Lathbury LJ, Steiner NK, Shulse CN, Pullikotil P, Seidah NG, et al. Three common alleles of KIR2DL4 (CD158d) encode constitutively expressed, inducible and secreted receptors in NK cells. Eur J Immunol. 2007;37(1):199–211.

    Article  CAS  PubMed  Google Scholar 

  217. Yu Y-R, Tian X-H, Wang Y, Feng M-F. Rapid production of human KIR2DL4 extracellular domain and verification of its interaction with HLA-G. Biochem (Moscow). 2006;71:S60–4.

    Article  CAS  Google Scholar 

  218. Nicolae D, Cox NJ, Lester LA, Schneider D, Tan Z, Billstrand C, et al. Fine mapping and positional candidate studies identify HLA-G as an asthma susceptibility gene on chromosome 6p21. Am J Hum Genet. 2005;76(2):349–57.

    Article  CAS  PubMed  Google Scholar 

  219. LeMaoult J, Le Discorde M, Rouas-Freiss N, Moreau P, Menier C, McCluskey J, et al. Biology and functions of human leukocyte antigen‐G in health and sickness. Tissue Antigens. 2003;62(4):273–84.

    Article  CAS  PubMed  Google Scholar 

  220. White SR, Loisel DA, McConville JF, Stern R, Tu Y, Marroquin BA, et al. Levels of soluble human leukocyte antigen-G are increased in asthmatic airways. Eur Respir J. 2010;35(4):925–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Szabo SJ, Sullivan BM, Peng SL, Glimcher LH. Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol. 2003;21(1):713–58.

    Article  CAS  PubMed  Google Scholar 

  222. Vacca P, Cantoni C, Vitale M, Prato C, Canegallo F, Fenoglio D, et al. Crosstalk between decidual NK and CD14 + myelomonocytic cells results in induction of Tregs and immunosuppression. Proc Natl Acad Sci. 2010;107(26):11918–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Faure M, Long EO. KIR2DL4 (CD158d), an NK cell-activating receptor with inhibitory potential. J Immunol. 2002;168(12):6208–14.

    Article  CAS  PubMed  Google Scholar 

  224. Kikuchi-Maki A, Yusa S-i, Catina TL, Campbell KS. KIR2DL4 is an IL-2-regulated NK cell receptor that exhibits limited expression in humans but triggers strong IFN-γ production. J Immunol. 2003;171(7):3415–25.

    Article  CAS  PubMed  Google Scholar 

  225. Rajagopalan S, Bryceson YT, Kuppusamy SP, Geraghty DE, Meer Avd, Joosten I, et al. Activation of NK cells by an endocytosed receptor for soluble HLA-G. PLoS Biol. 2006;4(1):e9.

    Article  PubMed  Google Scholar 

  226. Kikuchi-Maki A, Catina TL, Campbell KS. Cutting edge: KIR2DL4 transduces signals into human NK cells through association with the fc receptor γ protein. J Immunol. 2005;174(7):3859–63.

    Article  CAS  PubMed  Google Scholar 

  227. Shahjahan Miah S, Hughes TL, Campbell KS. KIR2DL4 differentially signals downstream functions in human NK cells through distinct structural modules. J Immunol. 2008;180(5):2922–32.

    Article  CAS  Google Scholar 

  228. Rajagopalan S. Endosomal signaling and a novel pathway defined by the natural killer receptor KIR2DL4 (CD158d). Traffic. 2010;11(11):1381–90.

    Article  CAS  PubMed  Google Scholar 

  229. Lanier LL, Corliss BC, Wu J, Leong C, Phillips JH. Immunoreceptor DAP12 bearing a tyrosine-based activation motif is involved in activating NK cells. Nature. 1998;391(6668):703–7.

    Article  CAS  PubMed  Google Scholar 

  230. Blunt MD, Khakoo SI. Activating killer cell immunoglobulin-like receptors: detection, function and therapeutic use. Int J Immunogenet. 2020;47(1):1–12.

    Article  CAS  PubMed  Google Scholar 

  231. Zhang M, Lam K-P, Xu S. Natural killer cell engagers (NKCEs): a new frontier in cancer immunotherapy. Front Immunol. 2023;14:1207276.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Naiyer MM, Cassidy SA, Magri A, Cowton V, Chen K, Mansour S, et al. KIR2DS2 recognizes conserved peptides derived from viral helicases in the context of HLA-C. Sci Immunol. 2017;2(15):eaal5296.

    Article  PubMed  Google Scholar 

  233. Rettman P, Blunt MD, Fulton RJ, Vallejo AF, Bastidas-Legarda LY, España-Serrano L et al. Peptide: MHC-based DNA vaccination strategy to activate natural killer cells by targeting killer cell immunoglobulin-like receptors. J Immunother Cancer. 2021;9(5).

  234. Wu WC, Shiu C, Tong TK, Leung SO, Hui CW. Suppression of NK Cell Activation by JAK3 Inhibition: Implication in the Treatment of Autoimmune Diseases. Journal of Immunology Research. 2023;2023.

  235. Creelan BC, Antonia SJ. The NKG2A immune checkpoint—a new direction in cancer immunotherapy. Nat Reviews Clin Oncol. 2019;16(5):277–8.

    Article  CAS  Google Scholar 

  236. Yang K, Zhao Y, Sun G, Zhang X, Cao J, Shao M, et al. Clinical application and prospect of immune checkpoint inhibitors for CAR-NK cell in tumor immunotherapy. Front Immunol. 2023;13:1081546.

    Article  PubMed  PubMed Central  Google Scholar 

  237. Zhang Y, Jin K, Dai Y, Hu N, Zhou T, Yang Z, et al. The change of Siglec-9 expression in peripheral blood NK cells of SFTS patients can affect the function of NK cells. Immunol Lett. 2023;263:97–104.

    Article  CAS  PubMed  Google Scholar 

  238. Yang Y, Han Q, Zhang C, Xiao M, Zhang J. Hepatitis B virus antigens impair NK cell function. Int Immunopharmacol. 2016;38:291–7.

    Article  CAS  PubMed  Google Scholar 

  239. Letafati A, Ardekani OS, Naderisemiromi M, Norouzi M, Shafiei M, Nik S, et al. Unraveling the dynamic mechanisms of natural killer cells in viral infections: insights and implications. Virol J. 2024;21(1):18.

    Article  PubMed  PubMed Central  Google Scholar 

  240. Wang X, Xiong H, Ning Z. Implications of NKG2A in immunity and immune-mediated diseases. Front Immunol. 2022;13:960852.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Antonioli L, Fornai M, Pellegrini C, Blandizzi C. NKG2A and COVID-19: another brick in the wall. Cell Mol Immunol. 2020;17(6):672–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. McWilliams EM, Mele JM, Cheney C, Timmerman EA, Fiazuddin F, Strattan EJ, et al. Therapeutic CD94/NKG2A blockade improves natural killer cell dysfunction in chronic lymphocytic leukemia. Oncoimmunology. 2016;5(10):e1226720.

    Article  PubMed  PubMed Central  Google Scholar 

  243. Kaulfuss M, Mietz J, Fabri A, Vom Berg J, Münz C, Chijioke O. The NK cell checkpoint NKG2A maintains expansion capacity of human NK cells. Sci Rep. 2023;13(1):10555.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Grote S, Ureña-Bailén G, Chan KC-H, Baden C, Mezger M, Handgretinger R, et al. In vitro evaluation of CD276-CAR NK-92 functionality, migration and invasion potential in the presence of immune inhibitory factors of the tumor microenvironment. Cells. 2021;10(5):1020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Ureña-Bailén G, Dobrowolski J-M, Hou Y, Dirlam A, Roig-Merino A, Schleicher S, et al. Preclinical evaluation of CRISPR-edited CAR-NK-92 cells for off-the-shelf treatment of AML and B-ALL. Int J Mol Sci. 2022;23(21):12828.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Albinger N, Bexte T, Buchinger L, Wendel P, Al-Ajami A, Gessner A, et al. CRISPR/Cas9 gene editing of immune checkpoint receptor NKG2A improves the efficacy of primary CD33-CAR-NK cells against AML. Blood. 2022;140(Supplement 1):4558–9.

    Article  Google Scholar 

  247. Encinas J, García-Ortiz A, Maroto-Martín E, Castellano E, Oliva R, Fernández RA, et al. S256: HLA-E/NKG2A CHECKPOINT DRIVES MULTIPLE MYELOMA RESISTANCE TO CAR-NK THERAPY. HemaSphere. 2023;7(S3):e16745e9.

    Article  PubMed Central  Google Scholar 

  248. Panek M, Stawiski K, Kaszkowiak M, Kuna P. Cytokine TGFβ gene polymorphism in asthma: TGF-related SNP analysis enhances the prediction of disease diagnosis (a case-control study with multivariable data-mining model development). Front Immunol. 2022;13:746360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Yu J, Wei M, Becknell B, Trotta R, Liu S, Boyd Z, et al. Pro-and antiinflammatory cytokine signaling: reciprocal antagonism regulates interferon-gamma production by human natural killer cells. Immunity. 2006;24(5):575–90.

    Article  CAS  PubMed  Google Scholar 

  250. Laouar Y, Sutterwala FS, Gorelik L, Flavell RA. Transforming growth factor-β controls T helper type 1 cell development through regulation of natural killer cell interferon-γ. Nat Immunol. 2005;6(6):600–7.

    Article  CAS  PubMed  Google Scholar 

  251. Saito A, Horie M, Nagase T. TGF-β signaling in lung health and disease. Int J Mol Sci. 2018;19(8):2460.

    Article  PubMed  PubMed Central  Google Scholar 

  252. Bergantini L, d’Alessandro M, Pianigiani T, Cekorja B, Bargagli E, Cameli P. Benralizumab affects NK cell maturation and proliferation in severe asthmatic patients. Clin Immunol. 2023;253:109680.

    Article  CAS  PubMed  Google Scholar 

  253. Batlle E, Massagué J. Transforming growth factor-β signaling in immunity and cancer. Immunity. 2019;50(4):924–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, et al. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010;363(13):1211–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Krammer S, Yang Z, Mitländer H, Grund JC, Trump S, Mittler S, et al. Rhinovirus suppresses TGF-β-GARP presentation by Peripheral NK cells. Cells. 2022;12(1):129.

    Article  PubMed  PubMed Central  Google Scholar 

  256. Koćwin M, Jonakowski M, Majos A, Szemraj J, Kuna P, Panek M. Evaluation of TGF-β isoforms based on selected clinical parameters in asthmatic patients. 2023.

  257. Tang J, Liu J, Zhang X. The role of osthole on TGF-β-induced lung epithelium apoptosis injury and epithelial-mesenchymal transition-mediated airway remodeling in pediatric asthma. Journal of Healthcare Engineering. 2022;2022.

  258. Peng H, Sun F, Jiang Y, Guo Z, Liu X, Zuo A, et al. Semaphorin 7a aggravates TGF-β1-induced airway EMT through the FAK/ERK1/2 signaling pathway in asthma. Front Immunol. 2023;14:1167605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Gutiérrez-Vera C, García-Betancourt R, Palacios PA, Müller M, Montero DA, Verdugo C, et al. Natural killer T cells in allergic asthma: implications for the development of novel immunotherapeutical strategies. Front Immunol. 2024;15:1364774.

    Article  PubMed  PubMed Central  Google Scholar 

  260. Min KY, Koo J, Noh G, Lee D, Jo MG, Lee JE, et al. CD1dhiPD-L1hiCD27 + Regulatory Natural Killer Subset suppresses atopic dermatitis. Front Immunol. 2022;12:752888.

    Article  PubMed  PubMed Central  Google Scholar 

  261. Borkar NA, Combs CK, Sathish V. Sex steroids effects on asthma: a network perspective of immune and airway cells. Cells. 2022;11(14):2238.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Ochayon DE, Waggoner SN. The effect of unconventional cytokine combinations on NK-cell responses to viral infection. Front Immunol. 2021;12:645850.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Witkowski M, Tizian C, Ferreira-Gomes M, Niemeyer D, Jones TC, Heinrich F, et al. Untimely TGFβ responses in COVID-19 limit antiviral functions of NK cells. Nature. 2021;600(7888):295–301.

    Article  CAS  PubMed  Google Scholar 

  264. Daher M, Basar R, Shaim H, Gokdemir E, Uprety N, Kontoyiannis A, et al. The TGF-β/SMAD signaling pathway as a mediator of NK cell dysfunction and immune evasion in myelodysplastic syndrome. Blood. 2017;130:53.

    Google Scholar 

  265. Shaim H, Shanley M, Basar R, Daher M, Gumin J, Zamler DB et al. Targeting the αv integrin/TGF-β axis improves natural killer cell function against glioblastoma stem cells. J Clin Investig. 2021;131(14).

  266. Yvon ES, Burga R, Powell A, Cruz CR, Fernandes R, Barese C, et al. Cord blood natural killer cells expressing a dominant negative TGF-β receptor: implications for adoptive immunotherapy for glioblastoma. Cytotherapy. 2017;19(3):408–18.

    Article  CAS  PubMed  Google Scholar 

  267. Chaudhry K, Geiger A, Dowlati E, Lang H, Sohai DK, Hwang EI, et al. Co-transducing B7H3 CAR-NK cells with the DNR preserves their cytolytic function against GBM in the presence of exogenous TGF-β. Mol Therapy-Methods Clin Dev. 2022;27:415–30.

    Article  CAS  Google Scholar 

  268. Kloss CC, Lee J, Zhang A, Chen F, Melenhorst JJ, Lacey SF, et al. Dominant-negative TGF-β receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication. Mol Ther. 2018;26(7):1855–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Chmielewski M, Abken H. TRUCKS, the fourth-generation CAR T cells: current developments and clinical translation. Adv Cell Gene Therapy. 2020;3(3):e84.

    Article  CAS  Google Scholar 

  270. Narayan V, Barber-Rotenberg JS, Jung I-Y, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Kobayashi Y, Chu HH, Kanda A, Yun Y, Shimono M, Nguyen LM, et al. CCL4 functions as a biomarker of type 2 airway inflammation. Biomedicines. 2022;10(8):1779.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Yang Y-L, Li X-F, Song B, Wu S, Wu Y-Y, Huang C, et al. The role of CCL3 in the pathogenesis of rheumatoid arthritis. Rheumatol Therapy. 2023;10(4):793–808.

    Article  Google Scholar 

  273. Yu W, Ye T, Ding J, Huang Y, Peng Y, Xia Q, et al. miR-4456/CCL3/CCR5 pathway in the pathogenesis of tight junction impairment in chronic obstructive pulmonary disease. Front Pharmacol. 2021;12:551839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Camargo JF, Quinones MP, Mummidi S, Srinivas S, Gaitan AA, Begum K, et al. CCR5 expression levels influence NFAT translocation, IL-2 production, and subsequent signaling events during T lymphocyte activation. J Immunol. 2009;182(1):171–82.

    Article  CAS  PubMed  Google Scholar 

  275. Cheng NL, Chen X, Kim J, Shi AH, Nguyen C, Wersto R, et al. MicroRNA-125b modulates inflammatory chemokine CCL4 expression in immune cells and its reduction causes CCL4 increase with age. Aging Cell. 2015;14(2):200–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Maghazachi AA. Role of chemokines in the biology of natural killer cells. Chemokine Syst Experimental Clin Hematol. 2010:37–58.

  277. Kobayashi Y, Kanda A, Yun Y, Bui DV, Suzuki K, Sawada S, et al. Reduced local response to corticosteroids in eosinophilic chronic rhinosinusitis with asthma. Biomolecules. 2020;10(2):326.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Hong J-H, Lee Y-C. Anti-inflammatory effects of cicadidae periostracum extract and oleic acid through inhibiting inflammatory chemokines using PCR arrays in LPS-induced lung inflammation in vitro. Life. 2022;12(6):857.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Capelli A, Di Stefano A, Gnemmi I, Balbo P, Cerutti C, Balbi B, et al. Increased MCP-1 and MIP-1beta in bronchoalveolar lavage fluid of chronic bronchitics. Eur Respir J. 1999;14(1):160–5.

    Article  CAS  PubMed  Google Scholar 

  280. Rabe KF, Nair P, Brusselle G, Maspero JF, Castro M, Sher L, et al. Efficacy and safety of dupilumab in glucocorticoid-dependent severe asthma. N Engl J Med. 2018;378(26):2475–85.

    Article  CAS  PubMed  Google Scholar 

  281. Berghi NO, Dumitru M, Vrinceanu D, Ciuluvica RC, Simioniuc-Petrescu A, Caragheorgheopol R, et al. Relationship between chemokines and T lymphocytes in the context of respiratory allergies. Experimental Therapeutic Med. 2020;20(3):2352–60.

    CAS  Google Scholar 

  282. Marques RE, Guabiraba R, Russo RC, Teixeira MM. Targeting CCL5 in inflammation. Expert Opin Ther Targets. 2013;17(12):1439–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  283. Alturaiki WH. Evaluation of CC chemokine ligand 5 (CCL5) chemokine, interleukin 5 (IL-5) cytokine, and eosinophil counts as potential biomarkers in Saudi patients with chronic asthma during sandstorms. Cureus. 2020;12(4).

  284. Lukacs NW, Strieter RM, Warmington K, Lincoln P, Chensue SW, Kunkel SL. Differential recruitment of leukocyte populations and alteration of airway hyperreactivity by CC family chemokines in allergic airway inflammation. Journal of immunology (Baltimore, Md: 1950). 1997;158(9):4398 – 404.

  285. Allard B, Levardon H. Asthmatic bronchial smooth muscle increases CCL5-dependent monocyte migration in response to rhinovirus-infected epithelium. Front Immunol. 2020;10:496326.

    Article  Google Scholar 

  286. Adams GP, Weiner LM. Monoclonal antibody therapy of cancer. Nat Biotechnol. 2005;23(9):1147–57.

    Article  CAS  PubMed  Google Scholar 

  287. Möller E. Contact-induced cytotoxicity by lymphoid cells containing foreign isoantigens. Science. 1965;147(3660):873–9.

    Article  Google Scholar 

  288. He Y, Tian Z. NK cell education via nonclassical MHC and non-MHC ligands. Cell Mol Immunol. 2017;14(4):321–30.

    Article  CAS  PubMed  Google Scholar 

  289. Flaherty DK, Martin JM, Storms WW, Kriz RJ, Surfus JE, Reed C. Antibody-dependent cellular cytotoxicity in asthmatics. J Allergy Clin Immunol. 1977;59(1):48–53.

    Article  CAS  PubMed  Google Scholar 

  290. Nimmerjahn F, Ravetch JV. Fcγ receptors as regulators of immune responses. Nat Rev Immunol. 2008;8(1):34–47.

    Article  CAS  PubMed  Google Scholar 

  291. de Saint Basile G, Ménasché G, Fischer A. Molecular mechanisms of biogenesis and exocytosis of cytotoxic granules. Nat Rev Immunol. 2010;10(8):568–79.

    Article  PubMed  Google Scholar 

  292. Eischen C, Leibson P. Role for NK-cell-associated Fas ligand in cell-mediated cytotoxicity and apoptosis. Res Immunol. 1997;148(3):164–9.

    Article  CAS  PubMed  Google Scholar 

  293. Battella S, Cox MC, Santoni A, Palmieri G. Natural killer (NK) cells and anti-tumor therapeutic mAb: unexplored interactions. J Leucocyte Biology. 2016;99(1):87–96.

    Article  CAS  Google Scholar 

  294. Inc SB, Potentiated. CD16 activated ADCC CAR-NK cells: News-Medical; 2024 [ https://www.news-medical.net/Potentiated-CD16-activated-ADCC-CAR-NK-cells

  295. Lepretre F, Gras D, Chanez P, Duez C. Natural killer cells in the lung: potential role in asthma and virus-induced exacerbation? Eur Respir Rev. 2023;32(169).

  296. Franklin M, Connolly E, Hussell T. Recruited and tissue-resident natural killer cells in the lung during infection and cancer. Front Immunol. 2022;13:887503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  297. Duvall MG, Fuhlbrigge ME, Reilly RB, Walker KH, Kılıç A, Levy BD. Human NK cell cytoskeletal dynamics and cytotoxicity are regulated by LIM kinase. J Immunol. 2020;205(3):801–10.

    Article  CAS  PubMed  Google Scholar 

  298. Kim M-H, Bae C-S, Bok S-H, Choi H-S, Ahn T, Cho S-S, et al. Drug Development from Natural products based on the pathogenic mechanism of Asthma. Int J Mol Sci. 2023;24(15):12469.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. Mathias CB. Natural killer cells in the development of asthma. Curr Allergy Asthma Rep. 2015;15:1–8.

    Article  CAS  Google Scholar 

  300. Shamji MH, Palmer E, Layhadi JA, Moraes TJ, Eiwegger T. Biological treatment in allergic disease. Wiley Online Library; 2021.

  301. Pelaia C, Vatrella A, Bruni A, Terracciano R, Pelaia G. Benralizumab in the treatment of severe asthma: design, development and potential place in therapy. Drug design, development and therapy. 2018:619 – 28.

  302. Dagher R, Kumar V, Copenhaver AM, Gallagher S, Ghaedi M, Boyd J et al. Novel mechanisms of action contributing to benralizumab’s potent anti-eosinophilic activity. Eur Respir J. 2022;59(3).

  303. Volpe E, Sambucci M, Battistini L, Borsellino G. Fas–Fas ligand: checkpoint of t cell functions in multiple sclerosis. Front Immunol. 2016;7:382.

    Article  PubMed  PubMed Central  Google Scholar 

  304. Mondal T, Gaur H, Wamba BE, Michalak AG, Stout C, Watson MR et al. Characterizing the regulatory Fas (CD95) epitope critical for agonist antibody targeting and CAR-T bystander function in ovarian cancer. Cell Death Differ. 2023:1–24.

  305. Zamai L, Ahmad M, Bennett IM, Azzoni L, Alnemri ES, Perussia B. Natural killer (NK) cell–mediated cytotoxicity: differential use of TRAIL and Fas ligand by immature and mature primary human NK cells. J Exp Med. 1998;188(12):2375–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  306. Screpanti V, Wallin RP, Grandien A, Ljunggren H-G. Impact of FASL-induced apoptosis in the elimination of tumor cells by NK cells. Mol Immunol. 2005;42(4):495–9.

    Article  CAS  PubMed  Google Scholar 

  307. Hatami Z, Hashemi ZS, Eftekhary M, Amiri A, Karpisheh V, Nasrollahi K, et al. Natural killer cell-derived exosomes for cancer immunotherapy: innovative therapeutics art. Cancer Cell Int. 2023;23(1):157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  308. Liu L, Zhou L, Wang L-L, Zheng P-D, Zhang F-Q, Mao Z-Y et al. Programmed cell death in asthma: apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis. J Inflamm Res. 2023:2727–54.

  309. Athari SS. Targeting cell signaling in allergic asthma. Signal Transduct Target Therapy. 2019;4(1):45.

    Article  Google Scholar 

  310. Sharma SK, Almeida FA, Kierstein S, Hortobagyi L, Lin T, Larkin A, et al. Systemic FasL neutralization increases eosinophilic inflammation in a mouse model of asthma. Allergy. 2012;67(3):328–35.

    Article  CAS  PubMed  Google Scholar 

  311. Rivera A, Ro G, Van Epps HL, Simpson T, Leiner I, Sant’Angelo DB, et al. Innate immune activation and CD4 + T cell priming during respiratory fungal infection. Immunity. 2006;25(4):665–75.

    Article  CAS  PubMed  Google Scholar 

  312. Ray A, Kolls JK. Neutrophilic inflammation in asthma and association with disease severity. Trends Immunol. 2017;38(12):942–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  313. Culley FJ. Natural killer cells in infection and inflammation of the lung. Immunology. 2009;128(2):151–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  314. Yi F, Cohen T, Zumbo P, Eltilib R, Arkin H, Gormally M, et al. Persistence but not antitumor efficacy of CAR-engineered lymphocytes is governed by a FAS/FAS ligand auto-regulatory circuit. Cancer Res. 2024;84(6Supplement):40.

    Article  Google Scholar 

  315. Al Subeh ZY, Poschel DB, Redd PS, Klement JD, Merting AD, Yang D, et al. Lipid nanoparticle delivery of fas plasmid restores fas expression to suppress melanoma growth in vivo. ACS Nano. 2022;16(8):12695–710.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  316. Garris CS, Arlauckas SP, Kohler RH, Trefny MP, Garren S, Piot C, et al. Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity. 2018;49(6):1148–61. e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  317. Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell. 2000;100(6):655–69.

    Article  CAS  PubMed  Google Scholar 

  318. Kato M. New insights into IFN-γ in rheumatoid arthritis: role in the era of JAK inhibitors. Immunological Med. 2020;43(2):72–8.

    Article  Google Scholar 

  319. Alspach E, Lussier DM, Schreiber RD. Interferon γ and its important roles in promoting and inhibiting spontaneous and therapeutic cancer immunity. Cold Spring Harb Perspect Biol. 2019;11(3):a028480.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  320. Song M, Ping Y, Zhang K, Yang L, Li F, Zhang C, et al. Low-dose IFNγ induces tumor cell stemness in tumor microenvironment of non–small cell lung cancer. Cancer Res. 2019;79(14):3737–48.

    Article  CAS  PubMed  Google Scholar 

  321. Gao Y, Yang J, Cai Y, Fu S, Zhang N, Fu X, et al. IFN-γ‐mediated inhibition of lung cancer correlates with PD‐L1 expression and is regulated by PI3K‐AKT signaling. Int J Cancer. 2018;143(4):931–43.

    Article  CAS  PubMed  Google Scholar 

  322. Frey A, Lunding LP, Ehlers JC, Weckmann M, Zissler UM, Wegmann M. More than just a barrier: the immune functions of the airway epithelium in asthma pathogenesis. Front Immunol. 2020;11:527401.

    Article  Google Scholar 

  323. Yan B, Chen F, Xu L, Wang Y, Wang X. Interleukin-28B dampens airway inflammation through up-regulation of natural killer cell-derived IFN-γ. Sci Rep. 2017;7(1):3556.

    Article  PubMed  PubMed Central  Google Scholar 

  324. Ji T, Li H. T-helper cells and their cytokines in pathogenesis and treatment of asthma. Front Immunol. 2023;14:1149203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  325. Victor JR, Lezmi G, Leite-de-Moraes M. New insights into asthma inflammation: focus on iNKT, MAIT, and γδT cells. Clin Rev Allergy Immunol. 2020;59:371–81.

    Article  CAS  PubMed  Google Scholar 

  326. Amniai L, Ple C, Barrier M, De Nadai P, Marquillies P, Vorng H, et al. Natural killer cells from allergic donors are defective in their response to CCL18 chemokine. Int J Mol Sci. 2021;22(8):3879.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  327. Renzi PM, Turgeon JP, Marcotte JE, Drblik SP, Berube D, Gagnon MF, et al. Reduced interferon-γ production in infants with bronchiolitis and asthma. Am J Respir Crit Care Med. 1999;159(5):1417–22.

    Article  CAS  PubMed  Google Scholar 

  328. Renzi PM, Turgeon JP, Yang JP, Drblik SP, Marcotte JE, Pedneault L, et al. Cellular immunity is activated and a TH-2 response is associated with early wheezing in infants after bronchiolitis. J Pediatr. 1997;130(4):584–93.

    Article  CAS  PubMed  Google Scholar 

  329. Do-Umehara HC, Chen C, Zhang Q, Schleimer RP, Budinger GS, Liu J. Suppression of allergic asthma by loss of function of Miz1-mediated Th1 skewing. Am J Respir Cell Mol Biol. 2022;67(3):346–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  330. Jazaeri S, Goldsmith AM, Jarman CR, Lee J, Hershenson MB, Lewis TC. Nasal interferon responses to community rhinovirus infections are similar in controls and children with asthma. Ann Allergy Asthma Immunol. 2021;126(6):690–5. e1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  331. Harrer DC, Schenkel C, Bezler V, Kaljanac M, Hartley J, Barden M, et al. CAR triggered release of type-1 interferon limits CAR T-cell activities by an artificial negative autocrine loop. Cells. 2022;11(23):3839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  332. Chmielewski M, Abken H. CAR T cells releasing IL-18 convert to T-Bethigh FoxO1low effectors that exhibit augmented activity against advanced solid tumors. Cell Rep. 2017;21(11):3205–19.

    Article  CAS  PubMed  Google Scholar 

  333. Kumric M, Urlic H, Bilalic A, Rezic-Muzinic N, Mastelic A, Markotic A, et al. Dynamic of circulating DNAM-1 + monocytes and NK Cells in patients with STEMI following primary percutaneous coronary intervention. J Cardiovasc Dev Disease. 2022;9(11):395.

    Article  CAS  Google Scholar 

  334. Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T, et al. DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity. 1996;4(6):573–81.

    Article  CAS  PubMed  Google Scholar 

  335. Cifaldi L, Melaiu O, Giovannoni R, Benvenuto M, Focaccetti C, Nardozi D, et al. DNAM-1 chimeric receptor-engineered NK cells: a new frontier for CAR-NK cell-based immunotherapy. Front Immunol. 2023;14:1197053.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  336. Hermans D, van Beers L, Broux B. Nectin family ligands trigger immune effector functions in health and autoimmunity. Biology. 2023;12(3):452.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  337. Perrone C, Bozzano F, Dal Bello MG, Del Zotto G, Antonini F, Munari E, et al. CD34 + DNAM-1brightCXCR4 + haemopoietic precursors circulate after chemotherapy, seed lung tissue and generate functional innate-like T cells and NK cells. Front Immunol. 2024;15:1332781.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  338. Tong L, Kremer V, Neo SY, Liu Y, Chen Y, Wagner AK, et al. Renal cell carcinoma escapes NK cell-mediated immune surveillance through the downregulation of DNAM‐1. Cancer Commun. 2023;43(7):855.

    Article  Google Scholar 

  339. Paolini R, Molfetta R. Dysregulation of DNAM-1-Mediated NK Cell anti-cancer responses in the Tumor Microenvironment. Cancers. 2023;15(18):4616.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  340. Murata R, Kinoshita S, Matsuda K, Kawaguchi A, Shibuya A, Shibuya K. G307S DNAM-1 mutation exacerbates autoimmune encephalomyelitis via enhancing CD4 + T cell activation. J Immunol. 2022;209(12):2304–12.

    Article  CAS  Google Scholar 

  341. Wang N, Liang S, Jin J, Fang L, Ma Q, Wang X, et al. CD226 attenuates Treg suppressive capacity via CTLA-4 and TIGIT during EAE. Immunol Res. 2019;67:486–96.

    Article  CAS  PubMed  Google Scholar 

  342. Chiang EY, de Almeida PE, de Almeida Nagata DE, Bowles KH, Du X, Chitre AS, et al. CD96 functions as a co-stimulatory receptor to enhance CD8 + T cell activation and effector responses. Eur J Immunol. 2020;50(6):891–902.

    Article  CAS  PubMed  Google Scholar 

  343. Zhang C, Hu Y, Xiao W, Tian Z. Chimeric antigen receptor-and natural killer cell receptor-engineered innate killer cells in cancer immunotherapy. Cell Mol Immunol. 2021;18(9):2083–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  344. Melaiu O, Lucarini V, Cifaldi L, Fruci D. Influence of the tumor microenvironment on NK cell function in solid tumors. Front Immunol. 2019;10:3038.

    Article  CAS  PubMed  Google Scholar 

  345. Oberschmidt O, Kloess S, Koehl U. Redirected primary human chimeric antigen receptor natural killer cells as an off-the-shelf immunotherapy for improvement in cancer treatment. Front Immunol. 2017;8:262112.

    Article  Google Scholar 

  346. Huang Y, Zeng J, Liu T, Xu Q, Song X, Zeng J. DNAM1 and 2B4 costimulatory domains enhance the cytotoxicity of anti-GPC3 chimeric antigen receptor-modified natural killer cells against hepatocellular cancer cells in vitro. Cancer Manage Res. 2020:3247–55.

  347. Abdullah M, Zahoor S, Ali A, Babar ME. Association of single nucleotide polymorphism in the upstream region of tumor necrosis factor alpha gene with asthma. Age (year). 2021;32(1250):4959–1582.

    Google Scholar 

  348. Jang D-i, Lee A-H, Shin H-Y, Song H-R, Park J-H, Kang T-B, et al. The role of tumor necrosis factor alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. Int J Mol Sci. 2021;22(5):2719.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  349. Chen J, Liu X, Zeng Z, Li J, Luo Y, Sun W, et al. Immunomodulation of NK cells by ionizing radiation. Front Oncol. 2020;10:874.

    Article  PubMed  PubMed Central  Google Scholar 

  350. Medler J, Wajant H. Tumor necrosis factor receptor-2 (TNFR2): an overview of an emerging drug target. Expert Opin Ther Targets. 2019;23(4):295–307.

    Article  CAS  PubMed  Google Scholar 

  351. Gough P, Myles IA. Tumor necrosis factor receptors: pleiotropic signaling complexes and their differential effects. Front Immunol. 2020;11:585880.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  352. Jiang Y, Yu M, Hu X, Han L, Yang K, Ba H, et al. STAT1 mediates transmembrane TNF-alpha-induced formation of death-inducing signaling complex and apoptotic signaling via TNFR1. Cell Death Differ. 2017;24(4):660–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  353. Fischer R, Kontermann RE, Pfizenmaier K. Selective targeting of TNF receptors as a novel therapeutic approach. Front cell Dev Biology. 2020;8:401.

    Article  Google Scholar 

  354. Kucka K, Wajant H. Receptor oligomerization and its relevance for signaling by receptors of the tumor necrosis factor receptor superfamily. Front cell Dev Biology. 2021;8:615141.

    Article  Google Scholar 

  355. Niessen NM, Gibson PG, Baines KJ, Barker D, Yang IA, Upham JW, et al. Sputum TNF markers are increased in neutrophilic and severe asthma and are reduced by azithromycin treatment. Allergy. 2021;76(7):2090–101.

    Article  CAS  PubMed  Google Scholar 

  356. Choi JP, Kim YS, Kim O, Kim YM, Jeon S, Roh TY, et al. TNF-alpha is a key mediator in the development of T h2 cell response to inhaled allergens induced by a viral PAMP double‐stranded RNA. Allergy. 2012;67(9):1138–48.

    Article  CAS  PubMed  Google Scholar 

  357. Sieck GC, Dogan M, Young-Soo H, Osorio Valencia S, Delmotte P. Mechanisms underlying TNFα‐induced enhancement of force generation in airway smooth muscle. Physiological Rep. 2019;7(17):e14220.

    Article  CAS  Google Scholar 

  358. Habib N, Pasha MA, Tang DD. Current understanding of asthma pathogenesis and biomarkers. Cells. 2022;11(17):2764.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  359. Hynes GM, Hinks TS. The role of interleukin-17 in asthma: a protective response? ERJ open Res. 2020;6(2).

  360. Huang G-H, Zeng X-L, Cheng Y-X, Zhu S-F, Luo W, Wen Q, et al. Adenoviral delivery of recombinant soluble human tumor necrosis factor receptor 1 partially normalized mouse model of asthma. J Investig Med. 2015;63(5):765–72.

    Article  CAS  PubMed  Google Scholar 

  361. Maier O, Fischer R, Agresti C, Pfizenmaier K. TNF receptor 2 protects oligodendrocyte progenitor cells against oxidative stress. Biochem Biophys Res Commun. 2013;440(2):336–41.

    Article  CAS  PubMed  Google Scholar 

  362. Peng J, Li X-M, Zhang G-R, Cheng Y, Chen X, Gu W, et al. TNF-TNFR2 signaling inhibits Th2 and Th17 polarization and alleviates allergic airway inflammation. Int Arch Allergy Immunol. 2019;178(3):281–90.

    Article  CAS  PubMed  Google Scholar 

  363. So T, Ishii N. The TNF–TNFR family of co-signal molecules. Co-signal molecules in T cell activation: Immune Regulation in Health and Disease. 2019:53–84.

  364. Ye L-L, Wei X-S, Zhang M, Niu Y-R, Zhou Q. The significance of tumor necrosis factor receptor type II in CD8 + regulatory T cells and CD8 + effector T cells. Front Immunol. 2018;9:343592.

    Article  Google Scholar 

  365. Morrissey JH, Fakhrai H, Edgington TS. Molecular cloning of the cDNA for tissue factor, the cellular receptor for the initiation of the coagulation protease cascade. Cell. 1987;50(1):129–35.

    Article  CAS  PubMed  Google Scholar 

  366. Konigsberg WH, Nemerson Y. Molecular cloning of the cDNA for human tissue factor. Cell. 1988;52(5):639–40.

    Article  CAS  PubMed  Google Scholar 

  367. Alashkar Alhamwe B, Potaczek DP, Miethe S, Alhamdan F, Hintz L, Magomedov A, et al. Extracellular vesicles and asthma—more than just a co-existence. Int J Mol Sci. 2021;22(9):4984.

    Article  PubMed  PubMed Central  Google Scholar 

  368. Zhao J, Jiang T, Li P, Dai L, Shi G, Jing X, et al. Tissue factor promotes airway pathological features through epithelial-mesenchymal transition of bronchial epithelial cells in mice with house dust mite-induced asthma. Int Immunopharmacol. 2021;97:107690.

    Article  CAS  PubMed  Google Scholar 

  369. de Boer JD, Majoor CJ, van’t Veer C, van der Bel EH. Asthma and coagulation. Blood. J Am Soc Hematol. 2012;119(14):3236–44.

    Google Scholar 

  370. Vadasz Z, Toubi E, editors. Hemostasis in allergy. Seminars in thrombosis and Hemostasis. Thieme Medical; 2018.

  371. Shi G, Zhao JW, Sun XX, Ma JF, Wang P, He FC, et al. TIPE2 is negatively correlated with tissue factor and thrombospondin-1 expression in patients with bronchial asthma. Experimental Therapeutic Med. 2018;15(4):3449–54.

    CAS  Google Scholar 

  372. Hu Z. Tissue factor as a new target for CAR-NK cell immunotherapy of triple-negative breast cancer. Sci Rep. 2020;10(1):2815.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  373. Lambrecht BN, Hammad H. The airway epithelium in asthma. Nat Med. 2012;18(5):684–92.

    Article  CAS  PubMed  Google Scholar 

  374. Jia Z, Bao K, Wei P, Yu X, Zhang Y, Wang X, et al. EGFR activation-induced decreases in claudin1 promote MUC5AC expression and exacerbate asthma in mice. Mucosal Immunol. 2021;14(1):125–34.

    Article  CAS  PubMed  Google Scholar 

  375. Peng W, Xia Q, Zhang Y, Cao D, Zheng X. VEGF and EGFR signaling pathways are involved in the baicalein attenuation of OVA-induced airway inflammation and airway remodeling in mice. Respir Res. 2024;25(1):10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  376. Chen J, Zeng F, Forrester SJ, Eguchi S, Zhang M-Z, Harris RC. Expression and function of the epidermal growth factor receptor in physiology and disease. Physiol Rev. 2016;96(3):1025–69.

    Article  CAS  PubMed  Google Scholar 

  377. Inoue H, Akimoto K, Homma T, Tanaka A, Sagara H. Airway epithelial dysfunction in asthma: relevant to epidermal growth factor receptors and airway epithelial cells. J Clin Med. 2020;9(11):3698.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  378. Wang C-H, Huang C-D, Lin H-C, Huang T-T, Lee K-Y, Lo Y-L, et al. Increased activation of fibrocytes in patients with chronic obstructive asthma through an epidermal growth factor receptor–dependent pathway. J Allergy Clin Immunol. 2012;129(5):1367–76.

    Article  CAS  PubMed  Google Scholar 

  379. Pan HH, Hsiao YP, Chen PJ, Kang YT, Chao YH, Sheu JN, et al. Epithelial growth factor receptor tyrosine kinase inhibitors alleviate house dust mite allergen Der p2-induced IL‐6 and IL‐8. Environ Toxicol. 2019;34(4):476–85.

    Article  CAS  PubMed  Google Scholar 

  380. Weng C-M, Lee M-J, Chao W, Lin Y-R, Chou C-J, Chen M-C, et al. Airway epithelium IgE-FcεRI cross-link induces epithelial barrier disruption in severe T2-high asthma. Mucosal Immunol. 2023;16(5):685–98.

    Article  PubMed  Google Scholar 

  381. Andersson CK, Iwasaki J, Cook J, Robinson P, Nagakumar P, Mogren S, et al. Impaired airway epithelial cell wound-healing capacity is associated with airway remodelling following RSV infection in severe preschool wheeze. Allergy. 2020;75(12):3195–207.

    Article  CAS  PubMed  Google Scholar 

  382. Wypych TP, Pattaroni C, Perdijk O, Yap C, Trompette A, Anderson D, et al. Microbial metabolism of L-tyrosine protects against allergic airway inflammation. Nat Immunol. 2021;22(3):279–86.

    Article  CAS  PubMed  Google Scholar 

  383. Ma R, Lu T, Li Z, Teng K-Y, Mansour AG, Yu M, et al. An oncolytic virus expressing IL15/IL15Rα combined with off-the-shelf EGFR-CAR NK cells targets glioblastoma. Cancer Res. 2021;81(13):3635–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  384. Liu Y, Zhou Y, Huang KH, Fang X, Li Y, Wang F, et al. Targeting epidermal growth factor-overexpressing triple‐negative breast cancer by natural killer cells expressing a specific chimeric antigen receptor. Cell Prolif. 2020;53(8):e12858.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  385. Yang Y, Nilsson M, Patel S, Robichaux J, Poteete A, Yu X, et al. EGFR CAR-NK cells demonstrate potent activity against EGFR TKI resistant NSCLC. Cancer Res. 2022;82(12Supplement):548.

    Article  Google Scholar 

  386. Allen ES, Stroncek DF, Ren J, Eder AF, West KA, Fry TJ, et al. Autologous lymphapheresis for the production of chimeric antigen receptor T cells. Transfusion. 2017;57(5):1133–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  387. Skuljec J, Chmielewski M, Happle C, Habener A, Busse M, Abken H, et al. Chimeric antigen receptor-redirected regulatory T cells suppress experimental allergic airway inflammation, a model of asthma. Front Immunol. 2017;8:1125.

    Article  PubMed  PubMed Central  Google Scholar 

  388. Borgert R. Improving outcomes and mitigating costs associated with CAR T-cell therapy. Am J Managed Care. 2021;27.

  389. Palani HK, Arunachalam AK, Yasar M, Venkatraman A, Kulkarni U, Lionel SA, et al. Decentralized manufacturing of anti CD19 CAR-T cells using CliniMACS Prodigy®: real-world experience and cost analysis in India. Bone Marrow Transplant. 2023;58(2):160–7.

    Article  PubMed  Google Scholar 

  390. Kekre N, Hay KA, Webb JR, Mallick R, Nielsen JS, Quizi J, et al. CLIC-01: manufacture and distribution of non-cryopreserved CAR-T cells for patients with CD19 positive hematologic malignancies. Front Immunol. 2022;13:1074740.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  391. Ran T, Eichmüller SB, Schmidt P, Schlander M. Cost of decentralized CAR T-cell production in an academic nonprofit setting. Int J Cancer. 2020;147(12):3438–45.

    Article  CAS  PubMed  Google Scholar 

  392. Cutmore LC, Marshall JF. Current perspectives on the use of off the shelf CAR-T/NK cells for the treatment of cancer. Cancers. 2021;13(8):1926.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  393. Daher M, Rezvani K. Next generation natural killer cells for cancer immunotherapy: the promise of genetic engineering. Curr Opin Immunol. 2018;51:146–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  394. Sutlu T, Stellan B, Gilljam M, Quezada HC, Nahi H, Gahrton G, et al. Clinical-grade, large-scale, feeder-free expansion of highly active human natural killer cells for adoptive immunotherapy using an automated bioreactor. Cytotherapy. 2010;12(8):1044–55.

    Article  CAS  PubMed  Google Scholar 

  395. Wagner J, Pfannenstiel V, Waldmann A, Bergs JW, Brill B, Huenecke S, et al. A two-phase expansion protocol combining interleukin (IL)-15 and IL-21 improves natural killer cell proliferation and cytotoxicity against rhabdomyosarcoma. Front Immunol. 2017;8:676.

    Article  PubMed  PubMed Central  Google Scholar 

  396. Liu S, Galat V, Galat4 Y, Lee YKA, Wainwright D, Wu J. NK cell-based cancer immunotherapy: from basic biology to clinical development. J Hematol Oncol. 2021;14:1–17.

  397. Gurney M, Kundu S, O’Dwyer M. Feeder cells at the interface of natural killer cell activation, expansion and gene editing. Front Immunol. 2022;13:802906.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  398. Lapteva N, Durett AG, Sun J, Rollins LA, Huye LL, Fang J, et al. Large-scale ex vivo expansion and characterization of natural killer cells for clinical applications. Cytotherapy. 2012;14(9):1131–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  399. Fujisaki H, Kakuda H, Shimasaki N, Imai C, Ma J, Lockey T, et al. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res. 2009;69(9):4010–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  400. Johnson CD, Zale NE, Frary ED, Lomakin JA. Feeder-cell-free and serum-free expansion of natural killer cells using Cloudz microspheres, G-Rex6M, and human platelet lysate. Front Immunol. 2022;13:803380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  401. Luczo JM, Ronzulli SL, Tompkins SM. Influenza a virus hemagglutinin and other pathogen glycoprotein interactions with NK cell natural cytotoxicity receptors NKp46, NKp44, and NKp30. Viruses. 2021;13(2):156.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  402. Horton NC, Mathew PA. NKp44 and natural cytotoxicity receptors as damage-associated molecular pattern recognition receptors. Front Immunol. 2015;6:126219.

    Article  Google Scholar 

  403. Colamartino AB, Lemieux W, Bifsha P, Nicoletti S, Chakravarti N, Sanz J, et al. Efficient and robust NK-cell transduction with baboon envelope pseudotyped lentivector. Front Immunol. 2019;10:483230.

    Article  Google Scholar 

  404. Ojeda P. Biological engineering of natural killer cells for cellular therapy against cancer. Harvard University; 2020.

  405. Romee R, Foley B, Lenvik T, Wang Y, Zhang B, Ankarlo D, et al. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood. J Am Soc Hematol. 2013;121(18):3599–608.

    CAS  Google Scholar 

  406. Balke-Want H, Keerthi V, Cadinanos-Garai A, Fowler C, Gkitsas N, Brown A, et al. Non-viral chimeric antigen receptor (CAR) T cells going viral. Immuno-Oncology Technol. 2023;18:100375.

    Article  CAS  Google Scholar 

  407. El-Mayta R, Zhang Z, Hamilton AG, Mitchell MJ. Delivery technologies to engineer natural killer cells for cancer immunotherapy. Cancer Gene Ther. 2021;28(9):947–59.

    Article  CAS  PubMed  Google Scholar 

  408. Bozza M, De Roia A, Correia MP, Berger A, Tuch A, Schmidt A, et al. A nonviral, nonintegrating DNA nanovector platform for the safe, rapid, and persistent manufacture of recombinant T cells. Sci Adv. 2021;7(16):eabf1333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  409. Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster E, et al. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020;367(6481):eaba7365.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  410. Tang X, Yang Y, Zheng M, Yin T, Huang G, Lai Z, et al. Magnetic–acoustic sequentially actuated CAR T cell microrobots for precision navigation and in situ antitumor immunoactivation. Adv Mater. 2023;35(18):2211509.

    Article  CAS  Google Scholar 

  411. Irvine DJ, Maus MV, Mooney DJ, Wong WW. The future of engineered immune cell therapies. Science. 2022;378(6622):853–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  412. Drent E, Poels R, van de Ruiter R, Zweegman S, Yuan H, et al. Combined CD28 and 4-1BB costimulation potentiates affinity-tuned chimeric antigen receptor–engineered T cells. Clin Cancer Res. 2019;25(13):4014–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  413. Lowry LE, Zehring WA. Potentiation of natural killer cells for cancer immunotherapy: a review of literature. Front Immunol. 2017;8:274001.

    Article  Google Scholar 

  414. Shaffer BC, Le Luduec J-B, Forlenza C, Jakubowski AA, Perales M-A, Young JW, et al. Phase II study of haploidentical natural killer cell infusion for treatment of relapsed or persistent myeloid malignancies following allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2016;22(4):705–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  415. Nguyen R, Wu H, Pounds S, Inaba H, Ribeiro RC, Cullins D, et al. A phase II clinical trial of adoptive transfer of haploidentical natural killer cells for consolidation therapy of pediatric acute myeloid leukemia. J Immunother Cancer. 2019;7:1–7.

    Article  Google Scholar 

  416. Xu Y, Liu Q, Zhong M, Wang Z, Chen Z, Zhang Y, et al. 2B4 costimulatory domain enhancing cytotoxic ability of anti-CD5 chimeric antigen receptor engineered natural killer cells against T cell malignancies. J Hematol Oncol. 2019;12:1–13.

    Article  Google Scholar 

  417. Xu D-L, He Y-Q, Xiao B, Si Y, Shi J, Liu X-A, et al. A novel sushi-IL15-PD1 CAR-NK92 cell line with enhanced and PD-L1 targeted cytotoxicity against pancreatic cancer cells. Front Oncol. 2022;12:726985.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  418. Cichocki F, Goodridge JP, Bjordahl R, Mahmood S, Davis ZB, Gaidarova S, et al. Dual antigen–targeted off-the-shelf NK cells show durable response and prevent antigen escape in lymphoma and leukemia. Blood. J Am Soc Hematol. 2022;140(23):2451–62.

    CAS  Google Scholar 

  419. Foltz JA, Berrien-Elliott MM, Neal C, Foster M, McClain E, Schappe T, et al. Cytokine-induced memory-like (ML) NK cells persist for > 2 months following adoptive transfer into leukemia patients with a MHC-compatible hematopoietic cell transplant (HCT). Blood. 2019;134:1954.

    Article  Google Scholar 

  420. Gang M, Marin ND, Wong P, Neal CC, Marsala L, Foster M, et al. CAR-modified memory-like NK cells exhibit potent responses to NK-resistant lymphomas. Blood J Am Soc Hematol. 2020;136(20):2308–18.

    Google Scholar 

  421. Gupta A, Gill S. CAR-T cell persistence in the treatment of leukemia and lymphoma. Leuk Lymphoma. 2021;62(11):2587–99.

    Article  CAS  PubMed  Google Scholar 

  422. Zhang Z, Miao L, Ren Z, Tang F, Li Y. Gene-edited interleukin CAR-T cells therapy in the treatment of malignancies: present and future. Front Immunol. 2021;12:718686.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  423. Yeku OO, Brentjens RJ. Armored CAR T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy. Biochem Soc Trans. 2016;44(2):412–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  424. White LG, Goy HE, Rose AJ, McLellan AD. Controlling cell trafficking: addressing failures in CAR T and NK cell therapy of solid tumours. Cancers. 2022;14(4):978.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  425. Chmielewski M, Abken H. TRUCKs: the fourth generation of CARs. Expert Opin Biol Ther. 2015;15(8):1145–54.

    Article  CAS  PubMed  Google Scholar 

  426. Rudek LS, Zimmermann K, Galla M, Meyer J, Neyazi B, Altvater B, et al. Generation of an NFκB-driven alpharetroviral all-in-one vector construct as a potent tool for CAR NK cell therapy. Front Immunol. 2021;12:751138.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  427. Amini L, Silbert SK, Maude SL, Nastoupil LJ, Ramos CA, Brentjens RJ, et al. Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion. Nat Reviews Clin Oncol. 2022;19(5):342–55.

    Article  Google Scholar 

  428. Xie G, Dong H, Liang Y, Ham JD, Rizwan R, Chen J. CAR-NK cells: a promising cellular immunotherapy for cancer. EBioMedicine. 2020;59.

  429. Caldwell KJ, Gottschalk S, Talleur AC. Allogeneic CAR cell therapy—more than a pipe dream. Front Immunol. 2021;11:618427.

    Article  PubMed  PubMed Central  Google Scholar 

  430. Zhu X, Li Q, Zhu X. Mechanisms of CAR T cell exhaustion and current counteraction strategies. Front Cell Dev Biology. 2022;10:1034257.

    Article  Google Scholar 

  431. Arpinati L, Scherz-Shouval R. From gatekeepers to providers: regulation of immune functions by cancer-associated fibroblasts. Trends Cancer. 2023;9(5):421–43.

    Article  CAS  PubMed  Google Scholar 

  432. Ghassemi S, Nunez-Cruz S, O’Connor RS, Fraietta JA, Patel PR, Scholler J, et al. Reducing ex vivo culture improves the antileukemic activity of chimeric antigen receptor (CAR) T cells. Cancer Immunol Res. 2018;6(9):1100–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  433. Gattinoni L, Klebanoff CA, Palmer DC, Wrzesinski C, Kerstann K, Yu Z, et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8 + T cells. J Clin Investig. 2005;115(6):1616–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  434. Alizadeh D, Wong RA, Yang X, Wang D, Pecoraro JR, Kuo C-F, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  435. Liu L, Bi E, Ma X, Xiong W, Qian J, Ye L, et al. Enhanced CAR-T activity against established tumors by polarizing human T cells to secrete interleukin-9. Nat Commun. 2020;11(1):5902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  436. Ptáčková P, Musil J, Štach M, Lesný P, Němečková Š, Král V, et al. A new approach to CAR T-cell gene engineering and cultivation using piggyBac transposon in the presence of IL-4, IL-7 and IL-21. Cytotherapy. 2018;20(4):507–20.

    Article  PubMed  Google Scholar 

  437. Denman CJ, Senyukov VV, Somanchi SS, Phatarpekar PV, Kopp LM, Johnson JL, et al. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS ONE. 2012;7(1):e30264.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  438. Gong W, Xiao W, Hu M, Weng X, Qian L, Pan X, et al. Ex vivo expansion of natural killer cells with high cytotoxicity by K562 cells modified to co-express major histocompatibility complex class I chain‐related protein A, 4‐1BB ligand, and interleukin‐15. Tissue Antigens. 2010;76(6):467–75.

    Article  CAS  PubMed  Google Scholar 

  439. Naume B, Gately M, Espevik T. A comparative study of IL-12 (cytotoxic lymphocyte maturation factor)-, IL-2-, and IL-7-induced effects on immunomagnetically purified CD56 + NK cells. Journal of immunology (Baltimore, Md: 1950). 1992;148(8):2429-36.

  440. Klöß S, Oberschmidt O, Morgan M, Dahlke J, Arseniev L, Huppert V, et al. Optimization of human NK cell manufacturing: fully automated separation, improved ex vivo expansion using IL-21 with autologous feeder cells, and generation of anti-CD123-CAR-expressing effector cells. Hum Gene Ther. 2017;28(10):897–913.

    Article  PubMed  Google Scholar 

  441. Majzner RG, Mackall CL. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 2018;8(10):1219–26.

    Article  CAS  PubMed  Google Scholar 

  442. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  443. Dai H, Wu Z, Jia H, Tong C, Guo Y, Ti D, et al. Bispecific CAR-T cells targeting both CD19 and CD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia. J Hematol Oncol. 2020;13:1–10.

    Google Scholar 

  444. Furqan F, Shah NN. Bispecific CAR T-cells for B-cell malignancies. Expert Opin Biol Ther. 2022;22(8):1005–15.

    Article  CAS  PubMed  Google Scholar 

  445. Mei H, Li C, Jiang H, Zhao X, Huang Z, Jin D, et al. A bispecific CAR-T cell therapy targeting BCMA and CD38 in relapsed or refractory multiple myeloma. J Hematol Oncol. 2021;14:1–17.

    Article  Google Scholar 

  446. Yan L, Qu S, Shang J, Shi X, Kang L, Xu N, et al. Sequential CD19 and BCMA-specific CAR T‐cell treatment elicits sustained remission of relapsed and/or refractory myeloma. Cancer Med. 2021;10(2):563–74.

    Article  CAS  PubMed  Google Scholar 

  447. Meng Y, Deng B, Rong L, Li C, Song W, Ling Z, 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;11:640166.

    Article  PubMed  PubMed Central  Google Scholar 

  448. Wing A, Fajardo CA, Posey AD Jr, Shaw C, Da T, Young RM, et al. Improving CART-cell therapy of solid tumors with oncolytic virus–driven production of a bispecific T-cell engager. Cancer Immunol Res. 2018;6(5):605–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  449. Balakrishnan A, Rajan A, Salter AI, Kosasih PL, Wu Q, Voutsinas J, et al. Multispecific targeting with synthetic ankyrin repeat motif chimeric antigen receptors. Clin Cancer Res. 2019;25(24):7506–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  450. Kuhn NF, Purdon TJ, van Leeuwen DG, Lopez AV, Curran KJ, Daniyan AF, et al. CD40 ligand-modified chimeric antigen receptor T cells enhance antitumor function by eliciting an endogenous antitumor response. Cancer Cell. 2019;35(3):473–88. e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  451. Valeri A, García-Ortiz A, Castellano E, Cordoba L, Maroto-Martín E, Encinas J, et al. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front Immunol. 2022;13:953849.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  452. Klingemann H. Are natural killer cells superior CAR. Drivers? Oncoimmunology. 2014;3(4):e28147.

    Article  PubMed  Google Scholar 

  453. Oelsner S, Waldmann A, Billmeier A, Röder J, Lindner A, Ullrich E, et al. Genetically engineered CAR NK cells display selective cytotoxicity against FLT3-positive B‐ALL and inhibit in vivo leukemia growth. Int J Cancer. 2019;145(7):1935–45.

    Article  CAS  PubMed  Google Scholar 

  454. Li Y, Basar R, Wang G, Liu E, Moyes JS, Li L, et al. KIR-based inhibitory CARs overcome CAR-NK cell trogocytosis-mediated fratricide and tumor escape. Nat Med. 2022;28(10):2133–44.

    Article  PubMed  PubMed Central  Google Scholar 

  455. Ewen EM, Pahl JH, Miller M, Watzl C, Cerwenka A. KIR downregulation by IL-12/15/18 unleashes human NK cells from KIR/HLA‐I inhibition and enhances killing of tumor cells. Eur J Immunol. 2018;48(2):355–65.

    Article  CAS  PubMed  Google Scholar 

  456. Tian Y, Li Y, Shao Y, Zhang Y. Gene modification strategies for next-generation CAR T cells against solid cancers. J Hematol Oncol. 2020;13(1):54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  457. Ruppel KE, Fricke S, Köhl U, Schmiedel D. Taking lessons from CAR-T cells and going beyond: tailoring design and signaling for CAR-NK cells in cancer therapy. Front Immunol. 2022;13:822298.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  458. Wrona E, Borowiec M, Potemski P. CAR-NK cells in the treatment of solid tumors. Int J Mol Sci. 2021;22(11):5899.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  459. Schirrmacher V. Cancer vaccines and oncolytic viruses exert profoundly lower side effects in cancer patients than other systemic therapies: a comparative analysis. Biomedicines. 2020;8(3):61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  460. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  461. Park JH, Rivière I, Gonen M, Wang X, Sénéchal B, Curran KJ, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378(5):449–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  462. Lee DW, Santomasso BD, Locke FL, Ghobadi A, Turtle CJ, Brudno JN, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25(4):625–38.

    Article  CAS  PubMed  Google Scholar 

  463. Mahmoudjafari Z, Hawks KG, Hsieh AA, Plesca D, Gatwood KS, Culos KA. American Society for Blood and Marrow Transplantation Pharmacy Special Interest Group survey on chimeric antigen receptor T cell therapy administrative, logistic, and toxicity management practices in the United States. Biol Blood Marrow Transplant. 2019;25(1):26–33.

    Article  PubMed  Google Scholar 

  464. Kanannejad Z, Soleimanian S, Ghahramani Z, Sepahi N, Mohkam M, Alyasin S, et al. Immune checkpoint molecules in prevention and development of asthma. Front Immunol. 2023;14:1070779.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  465. Zhang P, Wang Y, Miao Q, Chen Y. The therapeutic potential of pd-1/pd-L1 pathway on immune-related diseases: based on the innate and adaptive immune components. Biomed Pharmacother. 2023;167:115569.

    Article  CAS  PubMed  Google Scholar 

  466. Akbari O, Stock P, Singh A, Lombardi V, Lee W, Freeman G, et al. PD-L1 and PD-L2 modulate airway inflammation and iNKT-cell-dependent airway hyperreactivity in opposing directions. Mucosal Immunol. 2010;3(1):81–91.

    Article  CAS  PubMed  Google Scholar 

  467. Liu Y, Hu P, Zheng Z, Zhong D, Xie W, Tang Z, et al. Photoresponsive Vaccine-Like CAR‐M system with high‐efficiency central immune regulation for inflammation‐related depression. Adv Mater. 2022;34(11):2108525.

    Article  CAS  Google Scholar 

  468. Zhou X, Cao H, Fang S-Y, Chow RD, Tang K, Majety M, et al. CTLA-4 tail fusion enhances CAR-T antitumor immunity. Nat Immunol. 2023;24(9):1499–510.

    Article  CAS  PubMed  Google Scholar 

  469. Bäckel N, Hort S, Kis T, Nettleton DF, Egan JR, Jacobs JJ, et al. Elaborating the potential of Artificial Intelligence in automated CAR-T cell manufacturing. Front Mol Med. 2023;3:1250508.

    Article  PubMed  PubMed Central  Google Scholar 

  470. Yang J, Chen Y, Jing Y, Green MR, Han L. Advancing CAR T cell therapy through the use of multidimensional omics data. Nat Reviews Clin Oncol. 2023;20(4):211–28.

    Article  CAS  Google Scholar 

  471. Rajpurkar P, Chen E, Banerjee O, Topol EJ. AI in health and medicine. Nat Med. 2022;28(1):31–8.

    Article  CAS  PubMed  Google Scholar 

  472. Kasap C, Izgutdina A, Patiño-Escobar B, Kang A, Chilakapati N, Akagi N et al. Targeting high-risk multiple myeloma genotypes with optimized anti-CD70 CAR-T cells. bioRxiv. 2024:2024.02. 24.581875.

  473. Yang J, Chen Y, Han L. A multi-omics perspective of CAR T cell therapy. Clin Translational Med. 2023;13(5):e1274.

    Article  CAS  Google Scholar 

  474. Daniels KG, Wang S, Simic MS, Bhargava HK, Capponi S, Tonai Y, et al. Decoding CAR T cell phenotype using combinatorial signaling motif libraries and machine learning. Science. 2022;378(6625):1194–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  475. Hort S, Herbst L, Bäckel N, Erkens F, Niessing B, Frye M, et al. Toward rapid, widely available autologous CAR-T cell therapy–artificial intelligence and automation enabling the smart manufacturing hospital. Front Med. 2022;9:913287.

    Article  Google Scholar 

  476. Hartmann FJ, Babdor J, Gherardini PF, Amir E-AD, Jones K, Sahaf B, et al. Comprehensive immune monitoring of clinical trials to advance human immunotherapy. Cell Rep. 2019;28(3):819–31. e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  477. Munoz J, Deshpande A, Rimsza L, Nowakowski GS, Kurzrock R. Navigating between Scylla and Charybdis: a roadmap to do better than pola-RCHP in DLBCL. Cancer Treat Rev. 2024:102691.

  478. Achar SR, Bourassa FX, Rademaker TJ, Lee A, Kondo T, Salazar-Cavazos E, et al. Universal Antigen encoding of T cell activation from high-dimensional cytokine dynamics. Science. 2022;376(6595):880–4.

    Article  CAS  PubMed  Google Scholar 

  479. Capponi S, Daniels KG. Harnessing the power of artificial intelligence to advance cell therapy. Immunol Rev. 2023;320(1):147–65.

    Article  CAS  PubMed  Google Scholar 

  480. Bogatu A, Wysocka M, Wysocki O, Butterworth H, Pillai M, Allison J, et al. Meta-analysis informed machine learning: supporting cytokine storm detection during CAR-T cell therapy. J Biomed Inform. 2023;142:104367.

    Article  PubMed  Google Scholar 

  481. Song Y, Zhao J, Cai T, Stephens A, Su S-H, Sandford E, et al. Machine learning-based cytokine microarray digital immunoassay analysis. Biosens Bioelectron. 2021;180:113088.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  482. Wei Z, Xu J, Zhao C, Zhang M, Xu N, Kang L, et al. Prediction of severe CRS and determination of biomarkers in B cell-acute lymphoblastic leukemia treated with CAR-T cells. Front Immunol. 2023;14:1273507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  483. Zhang R, Han X, Lei Z, Jiang C, Gul I, Hu Q, et al. RCMNet: a deep learning model assists CAR-T therapy for leukemia. Comput Biol Med. 2022;150:106084.

    Article  CAS  PubMed  Google Scholar 

  484. Dagar G, Gupta A, Masoodi T, Nisar S, Merhi M, Hashem S, et al. Harnessing the potential of CAR-T cell therapy: progress, challenges, and future directions in hematological and solid tumor treatments. J Translational Med. 2023;21(1):449.

    Article  Google Scholar 

  485. Niazi SK. The Coming of Age of AI/ML in Drug Discovery, Development, Clinical Testing, and Manufacturing: The FDA Perspectives. Drug Design, Development and Therapy. 2023:2691 – 725.

  486. Rodriguez-Garcia A, Palazon A, Noguera-Ortega E, Powell DJ Jr, Guedan S. CAR-T cells hit the tumor microenvironment: strategies to overcome tumor escape. Front Immunol. 2020;11:1109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  487. 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.

    Article  CAS  PubMed  Google Scholar 

  488. Dragon AC, Zimmermann K, Nerreter T, Sandfort D, Lahrberg J, Klöß S et al. CAR-T cells and TRUCKs that recognize an EBNA-3 C-derived epitope presented on HLA-B* 35 control Epstein-Barr virus-associated lymphoproliferation. J Immunother Cancer. 2020;8(2).

  489. n den Eynde A, Gehrcken L, Verhezen T, Lau HW, Hermans C, Lambrechts H, et al. IL-15-secreting CAR natural killer cells directed toward the pan-cancer target CD70 eliminate both cancer cells and cancer-associated fibroblasts. J Hematol Oncol. 2024;17(1):1–19.

    Google Scholar 

  490. Ma X, Shou P, Smith C, Chen Y, Du H, Sun C, et al. Interleukin-23 engineering improves CAR T cell function in solid tumors. Nat Biotechnol. 2020;38(4):448–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  491. Brog RA, Ferry SL, Schiebout CT, Messier CM, Cook WJ, Abdullah L, et al. Superkine IL-2 and IL-33 armored CAR T cells reshape the tumor microenvironment and reduce growth of multiple solid tumors. Cancer Immunol Res. 2022;10(8):962–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  492. Włodarczyk M, Pyrzynska B. CAR-NK as a rapidly developed and efficient immunotherapeutic strategy against cancer. Cancers. 2022;15(1):117.

    Article  PubMed  PubMed Central  Google Scholar 

  493. Hawkins ER, D’Souza RR, Klampatsa A. Armored CAR T-cells: the next chapter in T-cell cancer immunotherapy. Biol Targets Ther. 2021:95–105.

  494. Safarzadeh Kozani P, Naseri A, Mirarefin SMJ, Salem F, Nikbakht M, Evazi Bakhshi S, et al. Nanobody-based CAR-T cells for cancer immunotherapy. Biomark Res. 2022;10(1):24.

    Article  PubMed  PubMed Central  Google Scholar 

  495. Lam N, Trinklein ND, Buelow B, Patterson GH, Ojha N, Kochenderfer JN. Anti-BCMA chimeric antigen receptors with fully human heavy-chain-only antigen recognition domains. Nat Commun. 2020;11(1):283.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  496. Wang W, Jiang J, Wu C. CAR-NK for tumor immunotherapy: clinical transformation and future prospects. Cancer Lett. 2020;472:175–80.

    Article  CAS  PubMed  Google Scholar 

  497. Wang X, Jasinski DL, Medina JL, Spencer DM, Foster AE, Bayle JH. Inducible MyD88/CD40 synergizes with IL-15 to enhance antitumor efficacy of CAR-NK cells. Blood Adv. 2020;4(9):1950–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  498. Srivastava S, Salter AI, Liggitt D, Yechan-Gunja S, Sarvothama M, Cooper K, et al. Logic-gated ROR1 chimeric antigen receptor expression rescues T cell-mediated toxicity to normal tissues and enables selective tumor targeting. Cancer Cell. 2019;35(3):489–503. e8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  499. Roybal KT, Rupp LJ, Morsut L, Walker WJ, McNally KA, Park JS, et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell. 2016;164(4):770–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  500. Webster B, Xiong Y, Hu P, Wu D, Alabanza L, Orentas RJ, et al. Self-driving armored CAR-T cells overcome a suppressive milieu and eradicate CD19 + raji lymphoma in preclinical models. Mol Ther. 2021;29(9):2691–706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  501. Kosti P, Opzoomer JW, Larios-Martinez KI, Henley-Smith R, Scudamore CL, Okesola M et al. Hypoxia-sensing CAR T cells provide safety and efficacy in treating solid tumors. Cell Rep Med. 2021;2(4).

  502. Raj D, Yang M-H, Rodgers D, Hampton EN, Begum J, Mustafa A, et al. Switchable CAR-T cells mediate remission in metastatic pancreatic ductal adenocarcinoma. Gut. 2019;68(6):1052–64.

    Article  CAS  PubMed  Google Scholar 

  503. Kuo Y-C, Kuo C-F, Jenkins K, Hung AF-H, Chang W-C, Park M et al. Antibody-based redirection of universal Fabrack-CAR T cells selectively kill antigen bearing tumor cells. J Immunother Cancer. 2022;10(6).

  504. Qi J, Tsuji K, Hymel D, Burke TR Jr, Hudecek M, Rader C, et al. Chemically programmable and switchable CAR-T therapy. Angew Chem Int Ed. 2020;59(29):12178–85.

    Article  CAS  Google Scholar 

  505. Cho JH, Collins JJ, Wong WW. Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell. 2018;173(6):1426–38. e11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  506. Walseng E, Köksal H, Sektioglu IM, Fåne A, Skorstad G, Kvalheim G, et al. A TCR-based chimeric antigen receptor. Sci Rep. 2017;7(1):10713.

    Article  PubMed  PubMed Central  Google Scholar 

  507. Jan M, Scarfò I, Larson RC, Walker A, Schmidts A, Guirguis AA, et al. Reversible ON-and OFF-switch chimeric antigen receptors controlled by lenalidomide. Sci Transl Med. 2021;13(575):eabb6295.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  508. Weber EW, Parker KR, Sotillo E, Lynn RC, Anbunathan H, Lattin J, et al. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science. 2021;372(6537):eaba1786.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  509. Labanieh L, Majzner RG, Klysz D, Sotillo E, Fisher CJ, Vilches-Moure JG, et al. Enhanced safety and efficacy of protease-regulated CAR-T cell receptors. Cell. 2022;185(10):1745–63. e22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  510. Li H-S, Wong NM, Tague E, Ngo JT, Khalil AS, Wong WW. High-performance multiplex drug-gated CAR circuits. Cancer Cell. 2022;40(11):1294–305. e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  511. Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol. 2013;31(1):71–5.

    Article  CAS  PubMed  Google Scholar 

  512. Sukumaran S, Watanabe N, Bajgain P, Raja K, Mohammed S, Fisher WE, et al. Enhancing the potency and specificity of engineered T cells for cancer treatment. Cancer Discov. 2018;8(8):972–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  513. Zhu I, Liu R, Garcia JM, Hyrenius-Wittsten A, Piraner DI, Alavi J, et al. Modular design of synthetic receptors for programmed gene regulation in cell therapies. Cell. 2022;185(8):1431–43. e16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  514. Fedorov VD, Themeli M, Sadelain M. PD-1–and CTLA-4–based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci Transl Med. 2013;5(215):ra215172–215172.

    Article  Google Scholar 

  515. Sandberg ML, Wang X, Martin AD, Nampe DP, Gabrelow GB, Li CZ, et al. A carcinoembryonic antigen-specific cell therapy selectively targets tumor cells with HLA loss of heterozygosity in vitro and in vivo. Sci Transl Med. 2022;14(634):eabm0306.

    Article  CAS  PubMed  Google Scholar 

  516. Tokatlian T, Asuelime GE, Mock J-Y, DiAndreth B, Sharma S, Warshaviak DT et al. Mesothelin-specific CAR-T cell therapy that incorporates an HLA-gated safety mechanism selectively kills tumor cells. J Immunother Cancer. 2022;10(1).

  517. Sweeney NP, Vink CA. The impact of lentiviral vector genome size and producer cell genomic to gag-pol mRNA ratios on packaging efficiency and titre. Mol Therapy Methods Clin Dev. 2021;21:574–84.

    Article  CAS  Google Scholar 

  518. Clubb JD, Gao TA, Chen YY. Synthetic biology in the engineering of CAR-T and CAR-NK cell therapies: facts and hopes. Clin Cancer Res. 2023;29(8):1390–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  519. Feng K-c, Guo Y-l, Liu Y, Dai H-r, Wang Y, Lv H-y, et al. Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J Hematol Oncol. 2017;10:1–11.

    Article  Google Scholar 

  520. Liu S, Deng B, Yin Z, Lin Y, An L, Liu D, et al. Combination of CD19 and CD22 CAR-T cell therapy in relapsed B‐cell acute lymphoblastic leukemia after allogeneic transplantation. Am J Hematol. 2021;96(6):671–9.

    Article  CAS  PubMed  Google Scholar 

  521. Shah NN, Johnson BD, Schneider D, Zhu F, Szabo A, Keever-Taylor CA, 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;26(10):1569–75.

    Article  CAS  PubMed  Google Scholar 

  522. Qu C, Zou R, Wang P, Zhu Q, Kang L, Ping N, et al. Decitabine-primed tandem CD19/CD22 CAR-T therapy in relapsed/refractory diffuse large B-cell lymphoma patients. Front Immunol. 2022;13:969660.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  523. 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.

    Article  CAS  PubMed  Google Scholar 

  524. Fousek K, Watanabe J, Joseph SK, George A, An X, Byrd TT, et al. CAR T-cells that target acute B-lineage leukemia irrespective of CD19 expression. Leukemia. 2021;35(1):75–89.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not Applicable.

Funding

No funding.

Author information

Authors and Affiliations

Authors

Contributions

M.M., F.J., and M.P. contributed to data gathering, preparing the primary manuscript, and designing the tables. M.M. and M.P. reviewed the last manuscript and edited the revisions and contributed to the scientific and grammatical revising the manuscript. F.J., M.M., and M.P. designed the figures. S.S.A. contributed to the final manuscript’s hypothesis, correspondence, and revising.

Corresponding author

Correspondence to Seyed Shamseddin Athari.

Ethics declarations

Ethics approval and consent to participate

Not Applicable.

Consent for publication

Not Applicable.

Conflict of interest

The authors declare no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it.The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mohammad Taheri, M., Javan, F., Poudineh, M. et al. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 22, 736 (2024). https://doi.org/10.1186/s12967-024-05534-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05534-8