International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Immune cellular therapy represents a large number of therapeutic modalities used in immune-oncology and that has been at the forefront of antitumor clinical strategies (1-3). Chimeric antigen receptor (CAR) T cell therapy has demonstrated consistent therapeutic advances for the treatment of patients, although there are challenges in the latter that need to be overcome (4,5). An illustration of clinical therapy employing CAR-engineered cells is adoptive T cell therapy (ACT), which is an example of passive immunotherapy that employs lymphocyte infusion to produce an anti-tumor response (6). CARs are expressed in genetically modified T cells using receptors with an external antigen-recognition domain derived from an antibody, a hinge domain, a transmembrane helix and an intracellular signaling component that activates and enhances T cell function upon target antigen binding. Due to their size and complexity, monoclonal antibodies have low cellular penetrance and poor tissue infiltration, making them ineffective for cancer immunotherapy and thought to cause drug resistance (7). By contrast, nanobodies are 10-times smaller than monoclonal antibodies and can permeate cell membranes and tissues. Due to having >80% sequence homology with human IgG heavy chains and high similarity to the human VH3 family, in addition to their small size, nanobodies have low immunogenicity and excellent biocompatibility. Moreover, humanizing camelid heavy chain antibodies (VHH) domains is easy due to their structural commonality (8).
Nanobodies are different from single-chain variable fragments (scFvs), as they are comprised of heavy chain antibody domains. While scFvs do not always fold efficiently and can be prone to aggregation (9,10), nanobodies are small, persistent, single-domain antibody fragments produced from camelids, exhibiting binding affinities comparable to conventional scFvs (11,12). Furthermore, the small size of nanobodies allows them to target different epitopes from those recognized by scFvs and they are typically less immunogenic than murine scFvs (13). In addition, unlike scFvs, nanobodies do not require the extra folding and assembly stages associated with V-region pairing. They can undergo cell surface display without the need for substantial linker optimization or other forms of structural reconfiguration. In addition, some reports found that scFvs are associated with tumor recurrence in some individuals due to their large size, high immunogenicity, weak affinity, easy aggregation, tonic signaling and low efficiency (14,15).
Therefore, it is essential to employ novel antibody types that improve the efficacy of CAR-T cell therapy. The structural engineering of CAR-T cells requires the genetic modification of receptors that may recognize malignant cells expressing tumor-associated molecules and which avoid working in a major histocompatibility complex (MHC)-dependent pattern, through antibody-antigen binding. For example, the CAR-T cell products that target CD19 primarily consist of Yescarta, Kymriah, Tecartus and Breyanzi, all of which have received approval from regulatory authorities. The US FDA (Food and Drug Administration) has approved CAR-T cells as a therapeutic intervention for various malignancies, including B-cell precursor acute lymphoblastic leukemia (B-ALL), follicular lymphoma, multiple myeloma (MM) and mantle cell lymphoma (MCL). While their application within clinical studies has yielded encouraging results, there is often a tendency to cause various degrees of cytokine release syndrome (CRS) (16), immune effector cell-associated neurotoxicity syndrome (ICANS) (17) and various forms of special B-cell deficiency, such as infection and low toxicity (18,19). The present review summarized the limitations and risks of nanobody CAR-T-cell therapy and potential solutions to these problems.
In the 1960s and 1970s, Jaques Miller discovered that the thymus was the origin of T cells, also referred to as T helper (TH) cells and that B lymphocytes (B cells) originated from the bone marrow (20,21). Two key interacting components primarily contribute to the exceptional cancer cell recognition capabilities of T cells, a compatible T cell receptor (TCR) associated with the cell membrane and a short peptide on the tumor cell that conveys tumor-associated antigens (TAAs). The MHC and human leukocyte antigen are both responsible for antigen presentation and helping the immune system recognize 'self' vs. 'non-self'. In the early 1980s, Steven Rosenberg isolated and described lymphokine-activated killer (LAK) cells from peripheral blood mononuclear cells (22).
In 1993, Klaus et al (23) observed that the TCR's α, β, γ and δ chains undergo rearrangement to form a clonal, non-specific TCR. Following this study, CD4-positive T helper cells, including regulatory T cells (Tregs) and CD8-positive cytotoxic T cells were identified (24). Subsequently, in 1989, Eshhar et al (25) proposed the existence of target-specific T lymphocytes using chimeric receptor molecules, opening up a new era of genetically engineered T cells. These targeted molecules consist of variable domains from antibodies and constant domains from TCRα or β, with a heavy variable (VH) domain from an anti-2,4,6-trinitrophenol (TNP) antibody and a light variable (VL) domain from an anti-αβ-TCR antibody switched out. Furthermore, Eshhar et al (25) redesigned an antibody-derived scFv into chimeric genes linked to the CD3ζ domain of the TCR. Fig. 1 shows that TCR has an antigen-binding domain, a transmembrane domain and a CD3ζ. CAR's binding domain is made up of a scFv, which contains the VL and heavy VH variable segments of a TAA-specific monoclonal antibody connected by a flexible linker. The scFv detects certain TAAs, which include proteins and glycoproteins (26). Fig. 2 shows how early CAR-T used monoclonal antibody protein-binding fragments to generate synthetic receptors, known as scFvs.
There have been five iterations of the CAR design, which have been designed mostly by altering the composition of the intracellular signaling environment of the cell, as illustrated in Fig. 3. In the genetically based production of CAR-T cells (Fig. 4), CAR-T cells are first multiplied in vitro before being administered into the peripheral circulation of the recipient patients.
The first generation of CARs was created by combining the scFv region of the anti-TNP antibody with a single ITAM region from CD3ξ or the Fc receptor γ-chain of the TCR. The first-generation CAR-T approach, also known as the 'T-body' technique, is used to genetically construct CAR-T cells (25). Despite the notable outcomes of first-generation CARs using in vitro and in vivo animal models or in the initial clinical trials, they demonstrated diminished cytotoxic potency and restricted persistence in vivo (27,28).
The second generation of CARs uses an extra-cytoplasmic signaling domain associated with the tumor necrosis factor receptor (TNFR) family, generated from co-stimulatory receptors such as CD28, CD134 (OX-40, TNFRSF4), 4-1BB (CD137, TNFRSF9) and ICOS (CD278) (29-31). These changes to the method improve T cell activation, cell proliferation in vitro, cytotoxic efficacy with increased persistence in vivo and anticancer activity (32). In addition, second-generation CAR-T cells possess the CD3ζ-activating domain, which includes co-stimulatory receptors like CD28, 4-1BB and CD137 (33,34).
The third-generation CAR features more co-stimulatory motifs on the cytoplasmic side compared with the second-generation CAR, which boosts T cell activation, proliferation and malignant cell cytotoxicity (32). The cytoplasmic domains of third-generation CAR-T cells often combine CD28 with 4-1BB/CD3ξ or CD134/CD3ξ and are now being assessed in clinical trials (35-39). However, the immunosuppressive tumor microenvironment (TME) hinders T lymphocyte persistence, penetration and function, thereby hindering CAR-T cell cytotoxicity against target tumor cells (40-42).
The molecular design of fourth-generation CARs was developed by integrating components into additional T cell activation signals. For example, T cells redirected for antigen-unrestricted cytokine-initiated killing (TRUCKs) are fourth-generation CAR-T cells that have the unique capacity to regulate cytotoxic activity in the absence of an antigen. These cells integrate the cytotoxic functions of CAR-T cells with the immunomodulatory potential to deliver cytokines, including IL-12, IL-7, IL-15, IL-21 and IL-23, as well as their combinations (43-49).
The fifth generation of CAR-T cells uses unique CAR designs to bind and activate the target antigen and induce cytokine signaling to achieve efficient antitumor effects with minimal off-target toxicity. Multiple CAR module designs should be improved to overcome challenges in their clinical application. For example, the iCasp9 'suicide gene' reduces the toxicity of CD19-targeted CAR-T cell therapy while maintaining its anti-tumor efficacy (50,51).
CARs typically comprise the antigen-binding fragment (Fab) of an antibody that targets surface antigens on tumor cells, together with an intracellular component that incorporates co-stimulatory signaling domains from CD3ζ along with supplementary co-stimulatory signals, including CD28 and 4-1BB. So far, the FDA has approved five CAR-T cell products for commercial use, four of which utilize scFv derived from the monoclonal antibody FMC63 and are designed to target CD19 (52). The VH and VL sections of these CARs are connected by a genetically encoded flexible linker, either (G4S)3 or (SG4)3, to produce an scFv for antigen recognition (53,54). The scFv exhibits superior tissue penetration compared with monoclonal antibodies (mAbs). The diminutive dimensions, enhanced affinity and prolonged specificity in recognizing target antigens make it a more suitable format for using antibodies in CAR-T applications and the modification of T cells (55). Nonetheless, the simple structure of the VH and VL domains in scFvs presents several constraints owing to the absence of an Fc region, which is present in a full immunoglobulin structure. In comparison to a full immunoglobulin, the scFv has similar instability and is prone to aggregation and misfolding. Therefore, scFv antigen binding stability and capacity are essential for CAR production and activity (56). Furthermore, the hydrophobic fragment of the scFv structure requires significant modification and further engineering to meet clinical safety requirements.
While the scFv can stably bind its target antigens, the molecular structure of scFvs leads to various unexpected challenges in practical applications. Fundamentally, a strong pairing linker is essential between the VH and VL domains. Products of CAR-T cells may stimulate host immune responses and, consequently, the arrangement of the linker and non-human protein structure can lead to an enhanced risk of immunogenicity, which, parallel to the generation of drug-resistant antibodies, may cause patients to develop mechanisms that enhance resistance to therapy (57). Should an antidrug reaction manifest in patients, the therapeutic efficacy of CAR-T will be reduced, ultimately leading to CAR-T cell death and treatment ineffectiveness (58). Humanized CAR-T cells improve survival, due to their decreased immunogenicity and are just as effective at cytotoxicity as murine CAR-T cells (59). The common occurrence of CAR-T cell loss during in vivo growth is attributed to the scFv format, which consists of natural dimeric variable sections linked by hydrophobic patches following their separation from constant domains (9). Prior research suggests that these hydrophobic structures are present on variable domains, increasing the propensity of recombinant scFvs for self-aggregation.
In addition, the antigen-independent self-aggregation of CAR-T cells promotes premature receptor clustering on the T cell surface, leading to tonic CD3ζ phosphorylation and subsequent cell exhaustion. The sustained activation-induced cell death resulting from this process leads to a progressive decline in functional CAR-T cell populations (60). Critically, this CAR-T cell depletion not only limits its therapeutic efficacy but also creates a permissive environment for immune escape, which is a key driver of tumor recurrence that has been observed in clinical CAR-T therapy. Moreover, some studies have shown that residual tumor cells that evade the initial CAR-T pressure often upregulate PD-L1 or downregulate the CAR-T target antigens, further exacerbating immune evasion (61-63).
On the other hand, a notable constraint is that the basic scFv structure restricts its further advancement in the use of complex CAR structures. The isolated Fv domain can only perform a function when the VH and VL domains are linked by a covalent bond, usually through a genetically programmed peptide, which leads to the formation of the scFv. scFvs were disregarded in prior research due to their instability, unfolding, domain shifting and aggregation (64). As such, simultaneously targeting two or more antigens may effectively reduce the likelihood of antigen escape variants in tumor cells. Dual targeting can be accomplished by employing two tandem antigen-binding regions to form bi-specific CARs. Two distinct tandem CARs (TanCAR) can precisely identify antigens or their epitopes. However, the production of TanCAR structures encounters significant obstacles because of the substantial inadequacy of scFv molecules, which depend on the potential right cross-pairing between VH and VL chains for both of the distinct scFvs (65). Furthermore, producing functional scFvs may be challenging because of the complexities involved in identifying the optimal linker (66). Alternatively, multiple CAR molecules can be expressed by several CAR-T cells. Triple CAR-T cells are derived from a tri-cistronic vector, as indicated by the relevant studies (67,68). This expanded alteration of T cells necessitates larger transgene inserts, which, in turn, hinders viral packaging and transduction effectiveness. In addition, multiple scFvs possess enlarged hydrophobic patches that encourage T cell surface receptor aggregation, which may result in overexpression of the CAR gene during dynamic domain swapping (69). Ultimately, scFv clustering or misfolding may result from the poor folding stability of the VH and VL domains, as well as the exposure of hydrophobic components in the framework to solvents (70). Furthermore, the clustering of CARs can result from interactions within the scFv framework and the binding affinity affects the target's tonic excessive signaling, resulting in T cell exhaustion in the early treatment stage (71).
Recently, antigen-binding alternatives to scFvs have emerged, including nanobodies. The integration of nanobodies in CAR-T constructions may mitigate the previously noted limitations, which include antigen escape, off-tumor toxicity and immunogenicity. Hamers-Casterman et al (72) identified heavy-chain-only antibodies (HcAbs) are the source of the nanobody, or VHH, in 1993. Subsequently, it was also described by Camelidae (camel, llama, alpaca) and Sharks (73,74). A nanobody is a small antibody fragment, between ~12 and 15 kDa, which constitutes nearly half the size of a scFv (~25 kDa). These nanobodies are constructed with a pair of heavy chains lacking the constant heavy 1 (CH1) domain, while also expressing normal VH-only antibodies, including the CH2 and CH3 domains. Consequently, these HcAbs primarily rely on the variable domain, utilizing the DNA encoded within the VHH for sequencing. The nanobody functions by attaching and interacting with its target antigen without the VL or constant domains, utilizing the remaining complementarity-determining region 3, which is analogous to IgG. According to these characteristics, nanobodies can reach concealed and cryptic epitopes that are inaccessible to larger mAbs (75). Conversely, there exists a degree of comparison between nanobodies and traditional monoclonal antibodies. Nanobodies exhibit superior performance in four key aspects: Binding capacity, stability, penetrability and specificity. Notably, nanobodies exhibit robust structural integrity even when subjected to thermal stress (>60°C), mechanical pressure (up to 500 MPa), acidic environments (pH<3) and protease-rich milieu (76).
Furthermore, besides the biochemical and functional properties, high binding affinity and adequate immunogenicity and stability of nanobodies, the variable domain VHH is cloned with minimal complexity and expressed easily while retaining its affinity capabilities (77). The sequence of the human VH gene family III, together with VH size, exhibits significant similarity, resulting in extensive applicability in patients. The immunogenicity of VHH is less than that of non-human antibodies due to the absence of the Fc domain and its optimal length. Furthermore, nanobodies necessitate little adjustment to their immunogenicity, in particular due to their short half-life, hence facilitating the humanization process (78). In addition, humanized or camelid VHH domains, or single motifs of human antibodies, can be engineered to exhibit binding diversity and selectivity akin to full VH and VL dual-domain antibodies, enhancing their use in CAR designs for tumor targeting (79).
Meanwhile, antibody variable domains exhibit significant variability due to their inherent dynamic stability and solubility, compared with conventional antibodies that include both VH and VL chains, while the VHH lacks light chains and CH1 domains. scFvs exhibit reciprocal interactions among variable domains and constant domains, in addition to their exposure to hydrophobic regions, due to their derivation from monoclonal antibodies, which imposes constraints on future engineering endeavors (80). The advantageous characteristics of the VHH, including diminutive size, exceptional stability, robust antigen-binding affinity, solubility and natural origin, render it appropriate for diverse CAR-T applications. There is a growing exploration of nanobody-based CAR constructs (81,82).
The rising demand for candidate CARs to address therapeutic requirements has shifted the focus to nanobodies, which have been recognized for their notable properties and benefits in CAR-T treatments. Table I illustrates the structure of a synthesized nanobody.
CD38 is a membrane protein characterized by a single transmembrane region and a 256-amino acid catalytic carboxyl motif. The membrane protein CD38 is part of an evolutionarily conserved enzyme family and is expressed in several organs, particularly in mature immune cells. CD38 is notably appealing as a therapeutic target since multiple myeloma and other malignant tumors have exceptionally high levels of CD38 expression (83). CD38-specific conventional mAbs, including daratumumab, isatuximab and MOR202, were recently introduced in clinical settings to treat multiple myeloma (84,85). A set of recombinant C-terminal CD38-specific nanobodies was extracted from vaccinated llamas and which recognized CD38 at three distinct epitopes (86). The target domain comprises a nanobody against CD38, the CD8α transmembrane domain, the 4-1BB co-stimulatory domain and the CD3ζ-activating domain, to produce CD38 CAR-T cells, which efficiently lysed CD38-positive cells in vitro. Additionally, mice with non-obese diabetes or severe combined immunodeficiency that received subcutaneous injections of RPMI 8226 cells were found to benefit from the ability of the CD38 CAR-T cells to efficiently inhibit tumor growth (87). In addition, a separate study focused on targeting three distinct, non-overlapping alternative CD38 epitopes, resulting in the creation of humanized WF211, MU1067 and JK36 nanobodies (88). Using CRISPR/Cas9 technology, these nanobodies were transduced into NK-92 cells, which is an engineered human NK cell line that stably co-expresses GFP and CD16. The resulting CD38-directed CAR-NK cells demonstrated specific, effective and powerful cytotoxicity, as evidenced by their effects on multiple myeloma and Burkitt lymphoma cells taken from eight patients (89).
The checkpoint inhibitor protein programmed death-ligand 1 (PD-L1) is highly expressed in a number of cancers and immune cells, serving as a vital and complex regulator in the therapeutic application of CAR-T treatment (90). As such, B3 and A12 VHHs have been produced by immunizing an alpaca with the purified ectodomain of mouse PD-L1 and both exhibited binding to PD-L1 with overlapping epitopes (91). In addition, the second-generation CAR utilizing binder A12 was found to kill PD-L1-positive tumor cells in a dose-dependent way. Further to this, an anti-PD-L1 nanobody was extracted from a partially synthetic naïve shark VNAR phage library (92). Following the incorporation of the B2 VNAR fragment as a cross-reactive binder for PD-L1 and PD-1, B2-based CAR-T cells were found to effectively suppress tumor development in breast and liver cancer models.
The polytopic transmembrane protein CD20 has multiple membrane-spanning domains, which present substantial technical difficulties owing to its intricate structural organization. The extracellular domains of CD20 exhibit suboptimal folding attributed to the brevity of the hinge region. In pursuit of a long hinge CD20, researchers used an expression DNA plasmid that encodes the complete CD20 for the purposes of immunizing a llama. Subsequently, anti-CD20 nanoCARs demonstrated strong CD20 expression and effectively killed CD20+ B-cell lymphomas, including the RL and Raji cell lines. This confirms the potent cytotoxicity of anti-CD20 nanoCARs against non-Hodgkin lymphoma targets. Subsequently, a xenograft mouse model was created through the subcutaneous injection of RL cells. The engrafted mice then received intravenous injections of CD20 nanoCAR-T cells. In this model, the administration of CAR-T cells resulted in the complete eradication of subcutaneous RL tumors in under 20 days, in comparison to the PBS-injected control mice, greatly extending the lifespan of the mice (93). Moreover, Liu et al (94) aimed to develop bispecific single-domain antibodies against CD20 and CD3 in order to demonstrate their strong antitumor effect in killing B-cell lymphoma cells in vitro.
CD19 is a transmembrane glycoprotein antigen associated with the superfamily of immunoglobulins which represents a compelling therapeutic target. It is expressed by both healthy and malignant B cells, in addition to dendritic cells. In a research context, the isolation of the CD19 antigen occurred after the immunization of a camel. Following this, the vaccinated PBMCs were employed to establish a nanobody gene phage display system, facilitating the selection of specific nanobodies targeting CD19 on human B cells through phage display technology. The nanobody binder for CD19 exhibited a binding affinity characterized by a value ranging from 15 to 33 nM. One particular study identified CD19 and CD20 nanobodies from a naturally occurring nanobody-expressing phage display library, subsequently constructed CD19-, CD20- and bispecific nanobody CAR-T cells (95). These nanobody CAR T cells demonstrated a remarkable capacity to selectively identify and eliminate tumor cell lines expressing CD19 and CD20, such as Raji or Daudi cells (96). Furthermore, the lethal effects of nanobody CAR T cells have been verified using primary patient-derived (PD) malignant cells from ALL (PD-ALL), which exhibited CD19 expression and ~20% CD20 expression. The PD-ALL cells then underwent incubation with nanobody CAR-T cells for five days, leading to the lysis of the PD-ALL cells and the subsequent secretion of IL-2 (96). A diverse array of CD19 nanobodies has been developed, exhibiting significant therapeutic effectiveness in the field of CD19-expressing B cell tumors (33,97).
B-cell maturation antigen (BCMA) is a member of the tumor necrosis factor family of proteins, which is mainly produced by both healthy and cancerous plasma cells, in addition to certain mature B cells (98). Two distinct types of nanobodies, namely monovalent anti-BCMA nanobodies and bivalent nanobodies that recognize BCMA, are in the process of development and have exhibited significant therapeutic response rates in instances of recurrence or refractory multiple myeloma. One particular study demonstrated the successful engineering of a monovalent anti-BCMA nanobody, which integrates the human CD8α signal peptide and a BCMA nanobody, along with the 4-1BB and CD3ζ intracellular domains (99). This alternatively engineered anti-BCMA CAR was developed with a bispecific BM38 CAR, integrating anti-BCMA and anti-CD38 nanobodies in a bi-epitopic configuration (100). This has been approved to treat B-cell malignancies with T lymphocytes that carry CARs specific to CD19 and BCMA. In order to ensure stable expression of BCMA, Wei et al (101) targeted G protein-coupled receptor C5 family member D (GPRC5D) to form anti-BCMA/GPRC5D bispecific CAR-T cells and injected these CAR-T into relapsed/refractory multiple myeloma (R/R MM) patients with extramedullary disease who participated in a clinical trial of treatment for 3 months. Although the patient experienced four CAR-T cell expansions and developed Grade 3 CRS, the symptoms were well-controlled and the treatment was generally safe (101). The objectives and associated nanobody-based CARs underscore the prospective applications of these CARs, as demonstrated by cellular tests and mouse models (Table I).
Angiogenesis and tumor metastasis rely on VEGFs and their receptors (VEGFRs). Numerous VEGF- and VEGFR-targeted CAR-T cells have been engineered to suppress tumor proliferation, with the former functioning through the latter. VEGFR2 is well recognized to bind to fetal liver kinase-1 in mice and the orthologous kinase-insert domain receptor in humans. VEGFR2 is highly expressed in malignant endothelial cells and represents a promising particular target for anti-angiogenesis therapy (102). Various anti-angiogenic therapeutics and nanomedicines targeting VEGF or VEGFR have received market approval, yet certain undesirable effects persist in T cell antitumor treatments. For instance, VEGF possesses immunosuppressive properties, which include hindering Tregs and M2-like tumor-associated macrophage populations within the TME while augmenting T lymphocyte responses, dendritic cell maturation and facilitating effective T cell infiltration. The regulation of cellular immunity promotes angiogenesis and tissue healing in solid tumors (103,104). Additionally, there are encouraging outcomes from studies on VEGFR2 CAR-T cell therapies, which indicate that a uniform standardized dosage of VEGFR2 CAR-engineered T cells, administered alongside IL-2, can markedly suppress the proliferation of five distinct tumor types in experimental murine models, by targeting solid tumor vasculature (105). Furthermore, another relevant clinical investigation assessed the medicinal application of anti-VEGFR2 CAR-T cells in patients with solid tumors, although the findings on their responses were not entirely cohesive (106). In this study, not all participants exhibited favorable results, thus emphasizing the necessity for meticulously structured clinical trials using anti-VEGFR2 CAR-T cell therapy to reduce toxicities and explore the intricate mechanisms of CAR-T therapy within various tumor types (107).
VEGFR2-specific nanobodies have been extracted from the commercial humanized camelid single-domain antibody library HuSdLTM (Creative Biolabs), for example, the nanobody NTV1 selectively binds to VEGFR-2 D3. This recombinant nanobody was shown to be ~57 kDa, as assessed by surface plasmon resonance. NTV1 is an anti-VEGFR2 D3 candidate targeting human breast cancer cells. It was shown to almost completely reduce the development of capillary-like structures in human umbilical vein endothelial cells by blocking VEGFR2 activity (108). Based on this evidence, NTV1 provides an innovative strategy using nanobody-based therapeutic applications. In addition, CAR-modified T cells showed >50% surface expression of VEGFR2 in the context of breast cancers (109) and they produce IL-2 and IFN-γ, thereby having a damaging effect on solid tumors that express VEGFR2 (110).
The tumor-associated glycoprotein-72 (TAG-72) antigen is a mucin-like transmembrane complex typically regarded as a biomarker indicative of poor prognosis in adenocarcinomas and is also a recognized target for selection. HER2 is a member of the human epidermal growth factor receptor family 1-4, which is highly expressed in individuals with breast or gastric cancer, thereby serving as an optimal target for antigen-specific guided CAR-T cells in these diseases. TAG-72 and HER2 nanobodies have been integrated into the second and third-generation CAR-Ts, respectively (111,112). A collection of 13 individual domains extracted from an immunized camel has been shown to produce TAG-specific CARs that activate against tumor cells expressing TAG-72 (LS-174T cells). The anti-TAG-72 CAR was engineered using a mixture of 13 VHHs that effectively activate in reaction to TAG-72-positive malignant cells, specifically LS-174T or MCF7 cells. These anti-TAG-72 CAR-T cells were shown to release IL-2 and also lyse cancerous cells. Following this, five oligoclonal HER2 VHHs were chosen from an immunological camel library to be incorporated into either the CD28-CD3ξ or CD28-OX40-CD3ξ signaling domains. As such, future studies on canine bladder cancer should take into account a research prediction analysis of the TAG72 and HER2 (113).
An attractive target for CAR-T therapy is epidermal growth factor receptor (EGFR), which is overexpressed in a number of solid tumors but expressed at lower levels in healthy tissues (114). McComb et al (115) used several camelid EGFR nanobodies as targets for CARs with high on-target activity against human EGFR in vitro and in vivo. As an approach, they gradually reduced the human CD8 hinge, a spacer region in several CAR constructs, to reduce their potent antigenic sensitivity. They proved that hinge-truncation can effectively reduce CAR antigenic sensitivity for epitopes near the target cell membrane. This approach could improve CAR selectivity for tumor-associated antigens and enable more complicated multi-antigen targeting CAR therapies. Hosking et al (116) used a human-derived HER2 targeting CAR with multiplex editing to overcome barriers and maximize efficacy in solid tumors. They used HER2-targeting CAR-T cells with multiplex editing to overcome barriers and maximize efficacy in solid tumors. By designing IL-7R-fusion, TGF-β-IL-18R and CXCR2, HER2-based CAR-Ts were able to persist and migrate to tumors.
Mucin A (MUC1) is a protein that can cross the cell membrane and is involved in immune system modulation, signal transduction and protecting mucous membranes. MUC1 can be selectively targeted by CAR-T cells without off-tumor effects, owing to its hypoglycosylated properties. In fact, the initial form of CAR-designed T cells incorporated a nanobody that uses anti-MUC1 VHH as the target attachment region (117). This restructured chimeric receptor design was integrated with human IgG3 or IgG3-Fc as the spacer, along with CD28 and CD3ζ for T cell activation purposes. Subsequently, CAR-transduced Jurkat cells had augmented proliferation after their co-cultivation with MUC1-positive MCF7 cancerous cells. In addition, these Jurkat cells demonstrated enhanced capabilities in IL-2 secretion and tumor cell lysis, despite the use of distinct hinges (IgG3-Fc and IgG3-Fc-hinge) within the CAR construct. However, this study demonstrated that using the FcγRII hinge in the CAR resulted in a distinct reduction in CAR production and IL-2 production (117). A separate study developed two anti-MUC1 CAR-T cell lines. The construct used SM3-CAR comprises an SM3 scFv that targets the MUC1 antigen and facilitates IL-2 secretion. This alternative CAR-T cell line, featuring the modified SM3 scFv (pSM3-CAR), displayed enhanced binding affinity to MUC1 and exhibited no IL-2 expression (118). Furthermore, to treat cholangiocarcinoma, researchers produced anti-MUC1-CAR T cells with PD-1-CD28 switch receptors that target MUC1 and activate the inhibitory signal of the PD-1/PD-L1 interaction (119).
With a molecular mass of 67 kDa, the PhiC31 integrase belongs to the resolvase/invertase family. The PhiC31 integrase system, derived from the phage of the Streptomyces bacteria, has the potential to enhance CAR expression efficiency (120). One study (121) employed the incorporation of donor plasmid (pCMV-γc-attB) and helper plasmid (pCAGGS-phiC31) DNA into targeted sites within mammalian genomes, leading to sustained and stable transgenic expression over time. The manifestation of anti-MUC1 on the surface of Jurkat cells was assessed using flow cytometry from 1-30 days post-transfection. In the initial phases, there was minimal distinction upon utilizing the PhiC31 integrase system. In the later stages, the expression of the anti-MUC1 CAR exceeded 50%. On day 30, the expression of the CAR was barely discernible in the control group. This suggested that the PhiC31 integrase system achieved optimal transfection efficiency and transgene stability. Rath et al (121) demonstrated strong expression of transgenes in stem cells and utilized recombinase-mediated cassette exchange at safe harbor loci. As such, the choice of recombinase enzyme is an important factor in ensuring the optimal efficiency and accuracy of the integration events.
Glypicans, including glypican-3 (GPC3), heparan sulfate proteoglycan (a biomarker for hepatocellular carcinoma) and glypican-2 (GPC2) (a related proteoglycan associated with neuroblastoma), are crucial in developmental morphogenesis and serve as modulators of the Wnt signaling pathway (122). For example, a llama VH single-domain antibody library was developed by screening llama-derived heavy chain antibody library against a target peptide antigen, followed by lead candidate production, purification and characterization by sdAb (123). The aforementioned operational procedures were conducted using recombinant antibody technology, yielding a phage-displayed library with specificities for target antigens separated through panning, including coating, blocking, binding, washing, elution and amplification (123). Using a comparable library, anti-GPC3 VH single-domain antibodies were identified through the phage display library and the leading nanobody demonstrated a specific binding capacity to a his-GPC3 antigen at a ratio of 1:3,000. In addition, Zhou et al (124) created bispecific CAR-T cells that target both FAP and GPC3 as a means of addressing tumor heterogeneity in HCC.
GPC2 represents an additional entity within the glypican family, characterized by a human single-domain demonstrating a high binding affinity (125). The CARs targeting GCP2 were obtained from eight different individual volunteers. In the study, the efficacy of GPC2-specific CAR-T cells regarding their cytotoxic capabilities was assessed. With a ratio of effectors to targets of 8:1, these cells exhibited lytic efficacy against IMR5 GPC2-positive neuroblastoma cells, with a mean of 56% (range 44-71). Moreover, GPC2 CAR-T cells have been demonstrated to have a significant capacity for inhibiting tumor growth in neuroblastoma models.
Fibronectin Extra Domain IIIB (EIIIB) is a splice variant of fibronectin that is highly expressed in tumor cells and tumor-associated neovasculature, with negligible expression in normal tissues (126,127). A number of aggressive tumors are susceptible to CAR-T cell targeting because they rely on the extracellular matrix (ECM) and angiogenesis for nutrient sustenance. It is therefore relevant that EIIIB antigens, which are rarely found in healthy adult tissues, are prevalent in the tumor ECM and newly created blood vessels (128). NJB2 is a camelid-derived VHH antibody developed to specifically target the EIIIB isoform, which is a tumor-restricted epitope of fibronectin that results from alternative splicing in malignant tissues. As such, the use of the nanobody NJB2 was used to produce B2 CAR-T cells and facilitate the transduction of B2 CARs in order to demonstrate their specificity and cytotoxicity in vitro. Following this, B16 melanoma tumor-positive mice were administered with B2 CAR-T cells, then demonstrating mitigated tumor progression and an increase in the percentage of surviving mice (129). However, further assessment of the efficacy of these CAR-T cells in an MC38 colon cancer model found a decreased expression level of EIIIB, relative to B16 tumors. Markedly, NJB2 was found to act efficiently with fewer adverse effects in solid tumor animal models that lack innate adaptive immunity and when targeting the tumor microenvironment and treating (130).
Prostate-specific membrane antigen (PSMA), a type II transmembrane glycoprotein with folate hydrolase activity, is a well-known cell surface target in the treatment of prostate cancer. PSMA belongs to the Thy-1/Ly-6 family, which is markedly associated with poor-prognosis malignancies and has been investigated as both a biomarker for the activity of the disease and a particular therapeutic target that has gathered considerable interest in drug research (131). The binding capability of nanobodies to target antigens is categorized based on their affinities, whereby dissociation constants (KD) ≤10 nM indicate high affinity, 10-100 nM represent moderate affinity and ≥100 nM is classified as low affinity. Another nanobody, designated JVZ-007, was isolated by immunizing a llama using four patient prostate carcinoma cell lines, demonstrating a KD of ~27.4 nM (132). First-generation CARs with tumor cells expressing PSMA were found to promote the production of IL2 and INF-γ while increasing the levels of CD69 (133). Subsequently, the second-generation NBPII-CAR, derived from an anti-PSMA nanobody, was expressed in transfected Jurkat cells and the cells were activated by targeting PSMA on tumor cells. In addition, NBPII-CAR was engineered with a complex comprising ζ activating components, the CD28 transmembrane domain and a nanobody linked by a spacer and it was found to enhance IL-2 cytokine synthesis, CD25 expression and cellular proliferation (134). This novel anti-PSMA CAR may provide enhanced opportunities for PSMA-targeted immunotherapy for the treatment of patients with prostate cancer (135).
The utilization of scFvs in CAR-T cell design may inadvertently lead to excessive tonic signaling independent of their target antigens, ultimately culminating in the premature exhaustion of T cells, even though they are capable of promoting receptor clustering on T cell surfaces and inducing strong signaling activation (136). By contrast, as illustrated in Fig. 5, nanobodies emerge as a compelling and advantageous candidate for the construction of optimized and multi-generation CARs, offering a potential solution to mitigate these issues with tonic signaling, while enhancing the durability and efficacy of CAR-T cell therapies overall. To improve the ability of CAR-T cells to proliferate and fewer unanticipated adverse effects, researchers have endeavored to implement a caspase-8-induced suicide gene system switch by incorporating relevant protein molecules within in vitro assays (136).
Bispecific NanoCARs face the challenge of aggregation associated with two VHH-based CARs. To address the challenge posed by tumor cell antigen escape variants resulting from the absence of target epitopes, bispecific CAR-T cells have been posited to circumvent this obstacle. These more sophisticated CAR-T cells integrate the VHH antigen-binding domains while omitting light chains, potentially reducing the probability of domain interchange during the concurrent expression of numerous nanobodies (137). For example, nanobodies that selectively target CD20 and HER2 can be combined to create a bispecific CAR-T cell, demonstrating comparable efficacy in eliminating tumor cells that express CD20, HER2, or both antigens. This design presents an opportunity to avert antigen escape and circumvent tumor resistance, despite the absence of functional tests in vitro and clinical data (138). In one study (139), a bispecific CAR-T was developed using CD30 and CD5 nanobodies derived from an alpaca nanobody phage library. The authors presented potential bispecific CAR-T cell therapy approaches for B-cell lymphoma treatment, effectively avoiding antigen escape. As such, the authors illustrated that nanobody-derived bispecific CAR-T cells exhibit a marked enhancement in tumor-fighting efficacy when compared with both single-target and bispecific scFv-derived CAR-T cells.
Furthermore, TandemCAR encodes numerous scFvs, leading to possible cross-pairing across the various VL and VH chains of the scFvs. Nanobodies have demonstrated benefits and practicality over scFvs in bispecific CAR-T applications. For example, LCAR-B38M CAR-T cells, equipped with two VHHs, specifically target distinct epitopes located in the extracellular region of BCMA, as manufactured by Nanjing Legend Biotech Corporation. The LCAR-B38M CAR-T demonstrated efficacy in Phase II/III clinical trials (140).
The NanoCAR technique, using VHH-based CARs, produces CAR-T cells equipped with two VHHs that can concurrently activate two tumor-eradicating pathways (138). Most publications concerning the NanoCAR-T format are confined to preclinical research due to the aggregation issue associated with scFv-based CARs, which is known to result in potential immunogenicity and unpredictability, therefore, investigating the a number of different forms of antibody release warrants greater focus (141). In one study (142), the engineered CAR-T cell structure was designed with a sophisticated dual-functional mechanism. This included an integrated release mechanism for anti-CD47 VHHs and an anti-PD-L1 inhibitory pathway-targeting mechanism. To evaluate the functional efficacy of this innovative CAR-T cell design, a killing assay was conducted using PD-L1-positive B16F10 cells. The results demonstrated that there were no statistically significant differences in either the killing activity or IFNγ production between CAR-T cells armed solely with the anti-PD-L1 mechanism and those co-expressing the anti-PD-L1 mechanism alongside the secreted anti-CD47 VHHs. B16F10 was injected into C57BL/6 PD-L1 knockout mice to assess its tumor-fighting efficacy on day one of the experiment, followed by therapy with varying quantities of anti-PD-L1 CAR-T cells on days 2, 6 and 12 (143). The cells that secrete anti-PD-L1 and target CD47 showed superior efficacy in preventing tumor growth and extending the life span of the mice, in contrast to anti-PD-L1 alone. Furthermore, the authors engineered EIIIB-target CAR-T cells that expressed anti-CD47 VHH to assess the in vivo production of anti-CD47 in a tumor model using C57BL/6 WT mice, which exhibited prolonged life compared with unmodified anti-EIIIB CAR-T cells. The results of this research confirmed that anti-CD47-expressing anti-PD-L1 CAR-T cells have enhanced effectiveness in combating cancer and can extend life expectancy via epitope spreading (143). Furthermore, two types of nanoCAR-T cells have undergone phase I clinical trials, BCMA-specific NanoCAR (NCT03664661) and CD19- or CD20-bispecific NanoCAR (NCT03881761).
By addressing the problem of cytokine release and precisely regulating cell activity to mitigate on-target and off-target undesirable effects, UniCAR, which is a switchable universal modular platform, has been designed to improve the safety and effectiveness of CARs (144). UniCAR consists of two components, UniCAR-T cells and an interchangeable tumor-specific targeting module. This UniCAR structure comprises an extracellular interaction portion originating from the antibody structure, a membrane region and an intracellular activation region featuring CD28 co-stimulatory and CD3ζ signaling components (145). In contrast to traditional CAR-T cells, the extracellular binding domain of the UniCAR fails to recognize a specific TAA on the target cells, but rather selectively binds to a shorter peptide, E5B6. At its C-terminal, this construct serves as a crucial connection between the α-EGFR-EGFR target module on tumor cells and has been demonstrated to guide UniCAR T lymphocytes to EGFR-expressing malignant cells both in vitro and in vivo (146). The authors of this study created a bivalent anti-EGFR antibody by fusing two α-EGFR nanobody domains using an E5B6 tag, which activates tumor cells when the UniCAR T cells are equipped with it (147). The E5B6 tag was incorporated into a lentiviral construct, which was then transduced into human primary T lymphocytes. In tumor-bearing murine models, the activation or deactivation of the UniCAR system occurs upon the fusion of the tumor-specific nanobody with the E5B6 tag, contingent upon the disruption of α-EGFR-EGFR (147). Whether UniCAR T cells can eradicate target-positive cells depends on the level of expression of EGFR by tumor cells and the binding ability of a bivalent EGFR-specific tumor marker.
Nanobody-based CAR-T cells have proven to be viable and promising for the advancement of immunotherapy. One monovalent anti-BCMA nanobody-based CAR-T cell, designated PRG1801, comprises CD8α, 4-1BB and CD3ζ and has been developed by Lu Han et al to target BCMA (99). At present, idecabtagene vicleucel (ide-cel or bb2121) and ciltacabtagene autoleucel (cilta-cel) are anti-BCMA CAR-T cell treatments developed by Memorial Sloan Kettering and Janssen (148,149), both of which obtained authorization for their clinical use from the US FDA in 2021 and 2022, respectively. Ide-cel consists of a murine anti-BCMA scFv split, which includes a 4-1BB co-stimulatory region, a CD3ζ signaling region, a CD8α hinge and a membrane region. The therapeutic effects of cilta-cel are analogous to those of ide-cel, which is known for its two OBAMA-binding scFv regions, transmembrane component, CD3ζ signaling region and 4-1BB co-stimulatory region (150). PRG1801 consists of a single anti-BCMA nanobody in addition to 4-1BB and CD3ζ co-stimulatory domains, whereas an alternative anti-BCMA CAR-T construct (Nanjing Legend Biotech) utilizes two distinct nanobodies for dual-epitope BCMA targeting (140,148,151,152). Certain clinical therapies utilize dual-targeted CAR-T cells aimed at BCMA for comparable clinical treatment efficacy, incorporating the recognition of single antigen scFv, bi-epitopic tandem VHHs, bispecific VHHs and single humanized VHH. Table II presents a comparison of the clinical data of four distinct anti-BCMA CAR-T cells.
Table IISummary table showing clinical trials and adverse effects linked to four anti-BCMA CAR T cell therapies. |
According to the results presented in Table II, two anti-BCMA CAR-T cells, including ide-cel and cilia-cel, have obtained US FDA approval for the therapy of R/R MM, while PRG-1801 and orva-cel are under investigation. The clinical perfusion dosage for ide-cel and orva-cel ranged from 50 to 800×106 cells, the aim for cilia-cel was 0.75×106 CAR-positive T cells per kg, LCAR-B38M ranged from 0.5 to 0.75×106 cells per kg and PRG1801 ranged from 2.5 to 10×106 cells per kg. The trial results for ide-cel reported an objective response rate (ORR) of 85%, a complete response (CR) rate of 45.5% and a partial response (PR) rate of 48.5%. The orva-cel results indicated an ORR of 91%, a stringent CR (sCR) rate of 39% and a PR rate of 27%. As the CAR structure continues to be optimized, two VHHs have been incorporated into it more recently: For example, cilta-cel, which demonstrated an ORR of 97%, a sCR rate of 67% and a PR rate of 48%. According to the published clinical outcomes of LCAR-B38M, the number of clinical trials rose from 57 to 97, with ORRs between 87.8 and 97.7%, CRS rates from 68 to 82.5% and PR rates from 3.1 to 5%. According to the data for PRG1801, the sCR was 55.9% and the overall response rate was 88.2%.
Three primary adverse effects have been reported across these trials as follows: CRS, hematologic toxicity and neurotoxicity, with modest distinctions between them. CRS and neurotoxicity are the most prevalent side effects linked to CAR-T cell treatments (153,154) and neurotoxicity frequently occurs along with CRS, partly due to excessive cytokine release (155). The American Society for Transplantation and Cellular Therapy (ASTCT) Consensus Guidelines classify CRS into five distinct grades based on clinical severity: Grade 1 corresponds to mild symptoms, Grade 2 indicates moderate symptoms, Grade 3 signifies severe symptoms, Grade 4 represents life-threatening symptoms and Grade 5 denotes mortality (156). The incidence of CRS for ide-cel is 76%, with 6% classified as Grade 3 CRS, whereas neurotoxicity affects 42% of individuals and 3% experience Grade 4 CRS. Orva-cel-treated patients showed no significant difference and the frequency of ≥Grade 3 CRS was 2% lower than that of ide-cel, while the frequency of neurotoxicity ≥Grade 3 was 4%. Patients treated with cilta-cel exhibited more pronounced CRS, affecting 92 out of 97 patients, with 4% experiencing Grade 3 or 4 CRS and 1% Grade 5 CRS. The overall incidence of neurotoxicity was 21%, with 9% classified as Grade 3 or above. In three separate clinical trials with LCAR-B38M, the incidence of CRS ranged from 90-91.9%, with ≥Grade 3 CRS occurring in 7-9.5% of cases (151,152,157,158). Their neurotoxicity was modest, with an overall incidence ranging from 1-1.8%, which is considerably lower than the occurrence likelihood of other CAR-T therapies. The CRS of PRG1801 was seen in 29 individuals, with a total incidence of 85.3% and a Grade 3 CRS rate of 2.9%, with no instances of neurotoxicity reported. Together, nanobody-derived therapies using CAR-T are currently being employed in clinical settings and the safety data have bolstered trust in their potential. Updated discoveries may facilitate the comprehensive development of CAR designs that incorporate the unique and advantageous characteristics of nanobodies.
Despite the success of nanobody-based CAR-T cell clinical therapy, reports indicate that CAR-T cells utilizing camel/mouse-derived antigen recognition structures can elicit neutralizing antibodies and induce hypersensitivity responses due to potential immunogenicity, potentially compromising the anti-tumor therapeutic efficacy of CAR-T cells (159,160). To address the risks of immunogenicity, nanobodies can easily be modified by humanizing the animal antibody-derived antigen-recognition domains, which may be a more reasonable option. However, in creating the native or humanized VHH-based CD19-redirected CAR-T cells, using fully human monoclonal antibody-derived antigen-recognition domains with a specific affinity range may not always be feasible (161,162). However, these humanized CAR-T cells exhibited comparable antitumor effects against tumor cells and produced comparable quantities of IFN-γ, IL-2 and TNF-α after co-cultivation.
Rapid blood clearance allows the use of short-lived isotopes in nanobody-based positron emission tomography imaging probes, decreasing patient radiation exposure (163). Conversely, mAb radiolabeling requires long-lived isotopes (164). However, for therapeutic applications, the rapid clearance rate of nanobodies from the circulation constitutes a significant limitation, requiring strategies to prolong their duration of action. One approach to mitigate this involves conjugating these single-domain antibodies into bivalent and trivalent forms (165). Furthermore, researchers have investigated the conjugation of specific VHHs with additional VHHs that target various antigens and epitopes, improving the remarkable circulatory half-life of these constructs (165). This advance is expected to facilitate more effective therapeutic applications. While this particular study highlights that nanobodies exhibit low immunogenicity owing to their small size, the humanization of selected VHHs remains a widely adopted strategy to mitigate the potential risks of immunogenicity. This humanization process typically involves fusing the chosen VHH with the Fc region of human IgGs (166). In addition, creating immune libraries with specialized nanobodies enables more efficient expression and purification operations. Under this approach, protein purification using PEG techniques can be used to purify the modified nanobodies, which may also be produced on a large scale, making them commercially viable. Furthermore, their enhanced penetration into solid tumors, owing to their small size and functionalization due to their low-fouling PEG coating, lowers transitory cellular interactions while increasing plasma circulation length (167,168). This can also be achieved via the slow release of nanobody, which is a high-affinity anti-human PD-L1 specific nanomaterial (also known as K2), within a peptide hydrogel, leading to enhanced accumulation of nanobodies in solid tumors to prolong their persistence (169,170).
The use of T cells in immunotherapy for neoplasms has achieved significant results in recent decades. Research has progressed from lacking an identifiable target to possessing a confirmed target and receiving clinical approval. The present review summarized a number of therapeutic targets that have become targetable by antibody-based CARs, which demonstrate anti-tumor effects analogous to those of scFvs. The diminutive size of nanobodies presents several significant advantages, including enhanced tissue penetration, the capability to cross the blood-brain barrier and access to epitopes that are otherwise difficult to reach (163). However, the nanobody structure requires considerable manufacturing complexity and exhibits the typical characteristics of reduced molecular weight (11,15). Furthermore, the small size of nanobodies results in a reduced circulating half-life and elevated renal clearance rates, which pose challenges for their therapeutic efficacy (78,165). Various innovative strategies have been developed to address these issues and improve the viability of nanobody-based therapies, such as fusing two nanobodies to a human IgG Fc fragment and utilizing PEG techniques to purify nanobodies (165,169,170).
In conclusion, as biotechnology advances, unique and useful nanobody-based CARs are being developed and have shown encouraging results, persuading researchers to investigate their therapeutic potential. Markedly, CAR-T cells produced with just one nanobody or bivalent nanobodies have demonstrated potential clinical efficacy. The viability of creating and producing bi-specific CARs in clinical trials has been demonstrated by LCAR-B38M bivalent nanobody CAR-T cells, paving the way for further advances in personalized cancer immunotherapy and potentially offering new hope for patients with previously challenging-to-treat malignancies.
Not applicable.
HL was primarily responsible for the writing, review and revision of this manuscript. XL, XZ, XH and SD compiled the references and developed the tables and figures. HF and YK developed the conceptual framework and design for the study. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
The present study received funding from the Basic Scientific Research Ability Enhancement Program for Young and Middle-aged Teachers of Guangxi (grant no. 2025KY1806) and the Self-funded Research Project of Guangxi Zhuang Autonomous Region Health Commission (grant no. Z-A20241190).
|
Xin Yu J, Hubbard-Lucey VM and Tang J: Immuno-oncology drug development goes global. Nat Rev Drug Discov. 18:899–900. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sanmamed MF and Chen L: A paradigm shift in cancer immunotherapy: From enhancement to normalization. Cell. 175:313–326. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Xin Yu J, Hubbard-Lucey VM and Tang J: The global pipeline of cell therapies for cancer. Nat Rev Drug Discov. 18:821–822. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yagi Y, Kanemasa Y, Sasaki Y, Goto S, Yamamura Y, Masuda Y, Fujita K, Ishimine K, Hayashi Y, Mino M, et al: Early failure is still a poor prognostic factor in patients with relapsed or refractory large B-cell lymphoma in the era of CAR T-cell therapy. J Clin Exp Hematop. 64:107–118. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Shumilov E, Scholz JK, Seib M, Mazzeo P, Wurm-Kuczera R, Vucinic V, Holtick U, Boyadzhiev H, Melchardt T, Hölscher AS, et al: Outcomes of bispecific antibody therapy after CAR T-cell failure in relapsed/refractory large B-cell lymphoma. Blood Adv. 9:3955–3966. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Manfredi F, Cianciotti BC, Potenza A, Tassi E, Noviello M, Biondi A, Ciceri F, Bonini C and Ruggiero E: TCR redirected T cells for cancer treatment: Achievements, hurdles, and goals. Front Immunol. 11:16892020. View Article : Google Scholar : PubMed/NCBI | |
|
Drewniak-Świtalska M, Fortuna P and Krzystek-Korpacka M: Negative immune checkpoint inhibitors. Pharmaceutics. 17:7132025. View Article : Google Scholar : | |
|
Safarzadeh Kozani P, Naseri A, Mirarefin SMJ, Salem F, Nikbakht M, Evazi Bakhshi S and Safarzadeh Kozani P: Nanobody-based CAR-T cells for cancer immunotherapy. Biomark Res. 10:242022. View Article : Google Scholar : PubMed/NCBI | |
|
Sun W, Xie J, Lin H, Mi S, Li Z, Hua F and Hu Z: A combined strategy improves the solubility of aggregation-prone single-chain variable fragment antibodies. Protein Expr Purif. 83:21–29. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
de Aguiar RB, da Silva TA, Costa BA, Machado MFM, Yamada RY, Braggion C, Perez KR, Mori MAS, Oliveira V and de Moraes JZ: Generation and functional characterization of a single-chain variable fragment (scFv) of the anti-FGF2 3F12E7 monoclonal antibody. Sci Rep. 11:14322021. View Article : Google Scholar : PubMed/NCBI | |
|
Koch-Nolte F: Nanobody-based heavy chain antibodies and chimeric antibodies. Immunol Rev. 328:466–472. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ingram JR, Schmidt FI and Ploegh HL: Exploiting nanobodies' singular traits. Annu Rev Immunol. 36:695–715. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
De Meyer T, Muyldermans S and Depicker A: Nanobody-based products as research and diagnostic tools. Trends Biotechnol. 32:263–270. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Natarajan V, Gopalakrishnan R, Matta H, Choi S, Gong S, Jeronimo A, Keerthipati PS, Morales A, Venkatesh H and Chaudhary PM: A fast and sensitive luciferase-based assay for antibody engineering and design of chimeric antigen receptors. Sci Rep. 10:23182020. View Article : Google Scholar : PubMed/NCBI | |
|
Guo S and Xi X: Nanobody-enhanced chimeric antigen receptor T-cell therapy: Overcoming barriers in solid tumors with VHH and VNAR-based constructs. Biomark Res. 13:412025. View Article : Google Scholar : PubMed/NCBI | |
|
Yoo JW: Management of adverse events in young adults and children with acute B-cell lymphoblastic leukemia receiving anti-CD19 chimeric antigen receptor (CAR) T-cell therapy. Blood Res. 58(S1): S20–S28. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Qi Y, Liu H, Li X, Shi Y, Mu J, Li J, Wang Y and Deng Q: Blinatumomab as salvage therapy in patients with relapsed/refractory B-ALL who have failed/progressed after anti-CD19-CAR T therapy. Ann Med. 55:22308882023. View Article : Google Scholar : PubMed/NCBI | |
|
Bouchkouj N, Kasamon YL, de Claro RA, George B, Lin X, Lee S, Blumenthal GM, Bryan W, McKee AE and Pazdur R: FDA approval summary: Axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma. Clin Cancer Res. 25:1702–1708. 2019. View Article : Google Scholar | |
|
Fischer JW and Bhattarai N: CAR-T cell therapy: Mechanism, management, and mitigation of inflammatory toxicities. Front Immunol. 12:6930162021. View Article : Google Scholar : PubMed/NCBI | |
|
Miller JF: Immunological function of the thymus. Lancet. 2:748–749. 1961. View Article : Google Scholar : PubMed/NCBI | |
|
Miller JF: Analysis of the thymus influence in leukaemogenesis. Nature. 191:248–249. 1961. View Article : Google Scholar : PubMed/NCBI | |
|
Taniguchi T, Matsui H, Fujita T, Takaoka C, Kashima N, Yoshimoto R and Hamuro J: Structure and expression of a cloned cDNA for human interleukin-2. Nature. 302:305–310. 1983. View Article : Google Scholar : PubMed/NCBI | |
|
Klaus SJ, Pinchuk LM, Ochs HD, Law CL, Fanslow WC, Armitage RJ and Clark EA: Costimulation through CD28 enhances T cell-dependent B cell activation via CD40-CD40L interaction. J Immunol. 152:5643–5652. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Mousset CM, Hobo W, Woestenenk R, Preijers F, Dolstra H and van der Waart AB: Comprehensive phenotyping of T cells using flow cytometry. Cytometry A. 95:647–654. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Eshhar Z, Waks T, Gross G and Schindler DG: Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci USA. 90:720–724. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Han D, Xu Z, Zhuang Y, Ye Z and Qian Q: Current progress in CAR-T cell therapy for hematological malignancies. J Cancer. 12:326–334. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hong M, Clubb JD and Chen YY: Engineering CAR-T cells for next-generation cancer therapy. Cancer Cell. 38:473–488. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Eshhar Z, Waks T and Gross G: The emergence of T-bodies/CAR T cells. Cancer J. 20:123–126. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Finney HM, Akbar AN and Lawson AD: Activation of resting human primary T cells with chimeric receptors: Costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain. J Immunol. 172:104–113. 2004. View Article : Google Scholar | |
|
Imai C, Mihara K, Andreansky M, Nicholson IC, Pui CH, Geiger TL and Campana D: Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 18:676–684. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
van der Stegen SJ, Hamieh M and Sadelain M: The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov. 14:499–509. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, Samanta M, Lakhal M, Gloss B, Danet-Desnoyers G, et al: Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 17:1453–1464. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Park JH, Rivière I, Gonen M, Wang X, Sénéchal B, Curran KJ, Sauter C, Wang Y, Santomasso B, Mead E, et al: Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 378:449–459. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Porter DL, Levine BL, Kalos M, Bagg A and June CH: Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 365:725–733. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong XS, Matsushita M, Plotkin J, Riviere I and Sadelain M: Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther. 18:413–420. 2010. View Article : Google Scholar | |
|
Pulè MA, Straathof KC, Dotti G, Heslop HE, Rooney CM and Brenner MK: A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol Ther. 12:933–941. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
George P, Dasyam N, Giunti G, Mester B, Bauer E, Andrews B, Perera T, Ostapowicz T, Frampton C, Li P, et al: Third-generation anti-CD19 chimeric antigen receptor T-cells incorporating a TLR2 domain for relapsed or refractory B-cell lymphoma: A phase I clinical trial protocol (ENABLE). BMJ Open. 10:e0346292020. View Article : Google Scholar : PubMed/NCBI | |
|
Schultz L: Chimeric antigen receptor T cell therapy for pediatric B-ALL: Narrowing the gap between early and long-term outcomes. Front Immunol. 11:19852020. View Article : Google Scholar : PubMed/NCBI | |
|
Ramos CA, Rouce R, Robertson CS, Reyna A, Narala N, Vyas G, Mehta B, Zhang H, Dakhova O, Carrum G, et al: In vivo fate and activity of second-versus third-generation CD19-Specific CAR-T cells in B cell non-hodgkin's lymphomas. Mol Ther. 26:2727–2737. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Pietrobon V, Todd LA, Goswami A, Stefanson O, Yang Z and Marincola F: Improving CAR T-cell persistence. Int J Mol Sci. 22:108282021. View Article : Google Scholar : PubMed/NCBI | |
|
Wagner J, Wickman E, DeRenzo C and Gottschalk S: CAR T cell therapy for solid tumors: Bright future or dark reality? Mol Ther. 28:2320–2339. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chin DS, Lim CSY, Nordin F, Arifin N and Jun TG: Antibody-dependent cell-mediated cytotoxicity through natural killer (NK) cells: Unlocking NK cells for future immunotherapy. Curr Pharm Biotechnol. 23:552–578. 2022. View Article : Google Scholar | |
|
Yasuda K, Nakanishi K and Tsutsui H: Interleukin-18 in health and disease. Int J Mol Sci. 20:6492019. View Article : Google Scholar : PubMed/NCBI | |
|
Ruixin S, Yifan L, Chuanlong W, Min Z, Hong L, Guoxiu D, Zhengyang L, Yansha S, Yiwei D, Jingwen S, et al: Expressing IL-15/IL-18 and CXCR2 improve infiltration and survival of EGFRvIII-targeting CAR-T cells in breast cancer. Biochem Pharmacol. 212:1155362023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M, Wen B, Anton OM, Yao Z, Dubois S, Ju W, Sato N, DiLillo DJ, Bamford RN, Ravetch JV and Waldmann TA: IL-15 enhanced antibody-dependent cellular cytotoxicity mediated by NK cells and macrophages. Proc Natl Acad Sci USA. 115:E10915–E10924. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Etxebeste-Mitxeltorena M, Del Rincón-Loza I and Martín-Antonio B: Tumor secretome to adoptive cellular immunotherapy: Reduce me before I make you my partner. Front Immunol. 12:7178502021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Y, Zhang M, Ramos CA, Durett A, Liu E, Dakhova O, Liu H, Creighton CJ, Gee AP, Heslop HE, et al: Closely related T-memory stem cells correlate with in vivo expansion of CAR. CD19-T cells and are preserved by IL-7 and IL-15. Blood. 123:3750–3759. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Lv X and Song Y: Short-term culture with IL-2 is beneficial for potent memory chimeric antigen receptor T cell production. Biochem Biophys Res Commun. 495:1833–1838. 2018. View Article : Google Scholar | |
|
Santomasso B, Bachier C, Westin J, Rezvani K and Shpall EJ: The other side of CAR T-cell therapy: Cytokine release syndrome, neurologic toxicity, and financial Burden. Am Soc Clin Oncol Educ Book. 39:433–444. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Guercio M, Manni S, Boffa I, Caruso S, Di Cecca S, Sinibaldi M, Abbaszadeh Z, Camera A, Ciccone R, Polito VA, et al: Inclusion of the inducible caspase 9 suicide gene in CAR construct increases safety of CAR.CD19 T cell therapy in B-cell malignancies. Front Immunol. 12:7556392021. View Article : Google Scholar : PubMed/NCBI | |
|
Diaconu I, Ballard B, Zhang M, Chen Y, West J, Dotti G and Savoldo B: Inducible caspase-9 selectively modulates the toxicities of CD19-specific chimeric antigen receptor-modified T cells. Mol Ther. 25:580–592. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
No authors listed. FDA okays second CAR-T for Kite. Nat Biotechnol. 38:10122020. View Article : Google Scholar : PubMed/NCBI | |
|
Galli S, Melidis L, Flynn SM, Varshney D, Simeone A, Spiegel J, Madden SK, Tannahill D and Balasubramanian S: DNA G-quadruplex recognition in vitro and in live cells by a structure-specific nanobody. J Am Chem Soc. 144:23096–23103. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Majzner RG and Mackall CL: Clinical lessons learned from the first leg of the CAR T cell journey. Nat Med. 25:1341–1355. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Strohl WR and Naso M: Bispecific T-cell redirection versus chimeric antigen receptor (CAR)-T cells as approaches to kill cancer cells. Antibodies (Basel). 8:2019.PubMed/NCBI | |
|
Fujiwara K, Masutani M, Tachibana M and Okada N: Impact of scFv structure in chimeric antigen receptor on receptor expression efficiency and antigen recognition properties. Biochem Biophys Res Commun. 527:350–357. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Shah NN and Fry TJ: Mechanisms of resistance to CAR T cell therapy. Nat Rev Clin Oncol. 16:372–385. 2019.PubMed/NCBI | |
|
Gorovits B and Koren E: Immunogenicity of chimeric antigen receptor T-cell therapeutics. BioDrugs. 33:275–284. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wagner DL, Fritsche E, Pulsipher MA, Ahmed N, Hamieh M, Hegde M, Ruella M, Savoldo B, Shah NN, Turtle CJ, et al: Immunogenicity of CAR T cells in cancer therapy. Nat Rev Clin Oncol. 18:379–393. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, Ingaramo M, Smith JP, Walker AJ, Kohler ME, Venkateshwara VR, et al: 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 21:581–590. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Sotillo E, Barrett DM, Black KL, Bagashev A, Oldridge D, Wu G, Sussman R, Lanauze C, Ruella M, Gazzara MR, et al: Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 5:1282–1295. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Liu W, Zou H, Guo L, Zhou Z, Xie Y, Guo H, Wei G, Zhang K, Yin H, Wei S and Chi J: Coordinating oncogenesis and immune evasion: KPNA2, GOLM1, and TK1 as novel CAR T-cell targets in lung adenocarcinoma. Eur J Med Res. 30:7652025. View Article : Google Scholar : PubMed/NCBI | |
|
Yan K and Xiao Z: Enhancing the antitumor activity of CD19/BCMA CAR-T cells in vitro with a PD1IL7R chimeric switch receptor. Cell Immunol. 415-416:1050012025. View Article : Google Scholar : PubMed/NCBI | |
|
Miller BR, Demarest SJ, Lugovskoy A, Huang F, Wu X, Snyder WB, Croner LJ, Wang N, Amatucci A, Michaelson JS and Glaser SM: Stability engineering of scFvs for the development of bispecific and multivalent antibodies. Protein Eng Des Sel. 23:549–557. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Jensen MC and Riddell SR: Design and implementation of adoptive therapy with chimeric antigen receptor-modified T cells. Immunol Rev. 257:127–144. 2014. View Article : Google Scholar : | |
|
Zhang K, Geddie ML, Kohli N, Kornaga T, Kirpotin DB, Jiao Y, Rennard R, Drummond DC, Nielsen UB, Xu L and Lugovskoy AA: Comprehensive optimization of a single-chain variable domain antibody fragment as a targeting ligand for a cytotoxic nanoparticle. MAbs. 7:42–52. 2015. View Article : Google Scholar : | |
|
Broussau S, Lytvyn V, Simoneau M, Guilbault C, Leclerc M, Nazemi-Moghaddam N, Coulombe N, Elahi SM, McComb S and Gilbert R: Packaging cells for lentiviral vectors generated using the cumate and coumermycin gene induction systems and nanowell single-cell cloning. Mol Ther Methods Clin Dev. 29:40–57. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Okuma A: Generation of CAR-T cells by lentiviral transduction. Methods Mol Biol. 2312:3–14. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ajina A and Maher J: Strategies to address chimeric antigen receptor tonic signaling. Mol Cancer Ther. 17:1795–1815. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Gil D and Schrum AG: Strategies to stabilize compact folding and minimize aggregation of antibody-based fragments. Adv Biosci Biotechnol. 4:73–84. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Jayaraman J, Mellody MP, Hou AJ, Desai RP, Fung AW, Pham AHT, Chen YY and Zhao W: CAR-T design: Elements and their synergistic function. EBioMedicine. 58:1029312020. View Article : Google Scholar : PubMed/NCBI | |
|
Hamers-Casterman C, Atarhouch T, Muyldermans S, Robinson G, Hamers C, Songa EB, Bendahman N and Hamers R: Naturally occurring antibodies devoid of light chains. Nature. 363:446–448. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Muyldermans S: Nanobodies: Natural single-domain antibodies. Annu Rev Biochem. 82:775–797. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Greenberg AS, Avila D, Hughes M, Hughes A, McKinney EC and Flajnik MF: A new antigen receptor gene family that undergoes rearrangement and extensive somatic diversification in sharks. Nature. 374:168–173. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Misson Mindrebo L, Liu H, Ozorowski G, Tran Q, Woehl J, Khalek I, Smith JM, Barman S, Zhao F, Keating C, et al: Fully synthetic platform to rapidly generate tetravalent bispecific nanobody-based immunoglobulins. Proc Natl Acad Sci USA. 120:e22166121202023. View Article : Google Scholar : PubMed/NCBI | |
|
Kunz P, Ortale A, Mücke N, Zinner K and Hoheisel JD: Nanobody stability engineering by employing the ΔTm shift; a comparison with apparent rate constants of heat-induced aggregation. Protein Eng Des Sel. 32:241–249. 2019.PubMed/NCBI | |
|
Debie P, Lafont C, Defrise M, Hansen I, van Willigen DM, van Leeuwen FWB, Gijsbers R, D'Huyvetter M, Devoogdt N, Lahoutte T, et al: Size and affinity kinetics of nanobodies influence targeting and penetration of solid tumours. J Control Release. 317:34–42. 2020. View Article : Google Scholar | |
|
Vincke C, Loris R, Saerens D, Martinez-Rodriguez S, Muyldermans S and Conrath K: General strategy to humanize a camelid single-domain antibody and identification of a universal humanized nanobody scaffold. J Biol Chem. 284:3273–3284. 2009. View Article : Google Scholar | |
|
Rahbarizadeh F, Ahmadvand D and Moghimi SM: CAR T-cell bioengineering: Single variable domain of heavy chain antibody targeted CARs. Adv Drug Deliv Rev. 141:41–46. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Honegger A: Engineering Antibodies for Stability and Efficient Folding. Handbook of Experimental Pharmacology. Chernajovsky Y and Nissim A: 181. Springer; Berlin, Heidelberg: pp. 47–68. 2008, View Article : Google Scholar | |
|
Jovčevska I and Muyldermans S: The therapeutic potential of nanobodies. BioDrugs. 34:11–26. 2020. View Article : Google Scholar | |
|
Chanier T and Chames P: Nanobody engineering: Toward next generation immunotherapies and immunoimaging of cancer. Antibodies (Basel). 8:132019. View Article : Google Scholar : PubMed/NCBI | |
|
Malavasi F, Deaglio S, Funaro A, Ferrero E, Horenstein AL, Ortolan E, Vaisitti T and Aydin S: Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol Rev. 88:841–886. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Lonial S, Weiss BM, Usmani SZ, Singhal S, Chari A, Bahlis NJ, Belch A, Krishnan A, Vescio RA, Mateos MV, et al: Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): An open-label, randomised, phase 2 trial. Lancet. 387:1551–1560. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
van de Donk N, Richardson PG and Malavasi F: CD38 antibodies in multiple myeloma: Back to the future. Blood. 131:13–29. 2018. View Article : Google Scholar | |
|
Li T, Qi S, Unger M, Hou YN, Deng QW, Liu J, Lam CMC, Wang XW, Xin D, Zhang P, et al: Immuno-targeting the multi-functional CD38 using nanobody. Sci Rep. 6:270552016. View Article : Google Scholar | |
|
An N, Hou YN, Zhang QX, Li T, Zhang QL, Fang C, Chen H, Lee HC, Zhao YJ and Du X: Anti-multiple myeloma activity of nanobody-based anti-CD38 chimeric antigen receptor T cells. Mol Pharm. 15:4577–4588. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Schriewer L, Schütze K, Petry K, Hambach J, Fumey W, Koenigsdorf J, Baum N, Menzel S, Rissiek B, Riecken K, et al: Nanobody-based CD38-specific heavy chain antibodies induce killing of multiple myeloma and other hematological malignancies. Theranostics. 10:2645–2658. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Hambach J, Riecken K, Cichutek S, Schütze K, Albrecht B, Petry K, Röckendorf JL, Baum N, Kröger N, Hansen T, et al: Targeting CD38-expressing multiple myeloma and burkitt lymphoma cells in vitro with nanobody-based chimeric antigen receptors (Nb-CARs). Cells. 9:3212020. View Article : Google Scholar : PubMed/NCBI | |
|
Tseng YH, Ho HL, Lai CR, Luo YH, Tseng YC, Whang-Peng J, Lin YH, Chou TY and Chen YM: PD-L1 expression of tumor cells, macrophages, and immune cells in non-small cell lung cancer patients with malignant pleural effusion. J Thorac Oncol. 13:447–453. 2018. View Article : Google Scholar | |
|
Ingram JR, Dougan M, Rashidian M, Knoll M, Keliher EJ, Garrett S, Garforth S, Blomberg OS, Espinosa C, Bhan A, et al: PD-L1 is an activation-independent marker of brown adipocytes. Nat Commun. 8:6472017. View Article : Google Scholar : PubMed/NCBI | |
|
Li D, English H, Hong J, Liang T, Merlino G, Day CP and Ho M: A novel PD-L1-targeted shark V(NAR) single-domain-based CAR-T cell strategy for treating breast cancer and liver cancer. Mol Ther Oncolytics. 24:849–863. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
De Munter S, Van Parys A, Bral L, Ingels J, Goetgeluk G, Bonte S, Pille M, Billiet L, Weening K, Verhee A, et al: Rapid and effective generation of nanobody based CARs using PCR and gibson assembly. Int J Mol Sci. 21:8832020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Ao K, Bao F, Cheng Y, Hao Y, Zhang H, Fu S, Xu J and Wu Q: Development of a bispecific nanobody targeting CD20 on B-cell lymphoma cells and CD3 on T cells. Vaccines (Basel). 10:13352022. View Article : Google Scholar : PubMed/NCBI | |
|
Banihashemi SR, Hosseini AZ, Rahbarizadeh F and Ahmadvand D: Development of specific nanobodies (VHH) for CD19 immuno-targeting of human B-lymphocytes. Iran J Basic Med Sci. 21:455–464. 2018.PubMed/NCBI | |
|
Wang H, Wang L, Li Y, Li G, Zhang X, Jiang D, Zhang Y, Liu L, Chu Y and Xu G: Nanobody-armed T cells endow CAR-T cells with cytotoxicity against lymphoma cells. Cancer Cell Int. 21:4502021. View Article : Google Scholar : PubMed/NCBI | |
|
Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, et al: Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 378:439–448. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Carpenter RO, Evbuomwan MO, Pittaluga S, Rose JJ, Raffeld M, Yang S, Gress RE, Hakim FT and Kochenderfer JN: B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res. 19:2048–2060. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Han L, Zhou J, Zhou K, Zhu X, Zhao L, Fang B, Yin Q, Wei X, Zhou H, Li L, et al: Safety and efficacy of CAR-T cell targeting BCMA in patients with multiple myeloma coinfected with chronic hepatitis B virus. J Immunother Cancer. 8:e0009272020. View Article : Google Scholar : PubMed/NCBI | |
|
Mei H, Li C, Jiang H, Zhao X, Huang Z, Jin D, Guo T, Kou H, Liu L, Tang L, et al: A bispecific CAR-T cell therapy targeting BCMA and CD38 in relapsed or refractory multiple myeloma. J Hematol Oncol. 14:1612021. View Article : Google Scholar : PubMed/NCBI | |
|
Wei L, Xiao X, Jing X, Zheng Y, Sun X, Bai W, Li M, Luo M and Xiao Y: Case report: Summary of multiple CAR-T expansions in anti-BCMA/GPRC5D bispecific CAR-T cell therapy for multiple myeloma. Front Immunol. 16:16077782025. View Article : Google Scholar : PubMed/NCBI | |
|
Slaney CY, Kershaw MH and Darcy PK: Trafficking of T cells into tumors. Cancer Res. 74:7168–7174. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wu X, Giobbie-Hurder A, Liao X, Lawrence D, McDermott D, Zhou J, Rodig S and Hodi FS: VEGF neutralization plus CTLA-4 blockade alters soluble and cellular factors associated with enhancing lymphocyte infiltration and humoral recognition in melanoma. Cancer Immunol Res. 4:858–868. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Chen DS and Hurwitz H: Combinations of bevacizumab with cancer immunotherapy. Cancer J. 24:193–204. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Chinnasamy D, Yu Z, Theoret MR, Zhao Y, Shrimali RK, Morgan RA, Feldman SA, Restifo NP and Rosenberg SA: Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenic tumors in mice. J Clin Invest. 120:3953–3968. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Spratlin JL, Cohen RB, Eadens M, Gore L, Camidge DR, Diab S, Leong S, O'Bryant C, Chow LQ, Serkova NJ, et al: Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol. 28:780–787. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Fukumura D, Kloepper J, Amoozgar Z, Duda DG and Jain RK: Enhancing cancer immunotherapy using antiangiogenics: Opportunities and challenges. Nat Rev Clin Oncol. 15:325–340. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ma L, Gu K, Zhang CH, Chen XT, Jiang Y, Melcher K, Zhang J, Wang M and Xu HE: Generation and characterization of a human nanobody against VEGFR-2. Acta Pharmacol Sin. 37:857–864. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Rydén L, Jirström K, Bendahl PO, Fernö M, Nordenskjöld B, Stål O, Thorstenson S, Jönsson PE and Landberg G: Tumor-specific expression of vascular endothelial growth factor receptor 2 but not vascular endothelial growth factor or human epidermal growth factor receptor 2 is associated with impaired response to adjuvant tamoxifen in premenopausal breast cancer. J Clin Oncol. 23:4695–4704. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Hajari Taheri F, Hassani M, Sharifzadeh Z, Behdani M, Arashkia A and Abolhassani M: T cell engineered with a novel nanobody-based chimeric antigen receptor against VEGFR2 as a candidate for tumor immunotherapy. IUBMB Life. 71:1259–1267. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sharifzadeh Z, Rahbarizadeh F, Shokrgozar MA, Ahmadvand D, Mahboudi F, Jamnani FR and Moghimi SM: Genetically engineered T cells bearing chimeric nanoconstructed receptors harboring TAG-72-specific camelid single domain antibodies as targeting agents. Cancer Lett. 334:237–244. 2013. View Article : Google Scholar | |
|
Jamnani FR, Rahbarizadeh F, Shokrgozar MA, Mahboudi F, Ahmadvand D, Sharifzadeh Z, Parhamifar L and Moghimi SM: T cells expressing VHH-directed oligoclonal chimeric HER2 antigen receptors: towards tumor-directed oligoclonal T cell therapy. Biochim Biophys Acta. 1840:378–386. 2014. View Article : Google Scholar | |
|
Vitti Gambim V, Laufer-Amorim R, Fonseca Alves RH, Grieco V and Fonseca-Alves CE: A comparative meta-analysis and in silico analysis of differentially expressed genes and proteins in canine and human bladder cancer. Front Vet Sci. 7:5589782020. View Article : Google Scholar : PubMed/NCBI | |
|
Sharifi J, Khirehgesh MR, Safari F and Akbari B: EGFR and anti-EGFR nanobodies: Review and update. J Drug Target. 29:387–402. 2021. View Article : Google Scholar | |
|
McComb S, Nguyen T, Shepherd A, Henry KA, Bloemberg D, Marcil A, Maclean S, Zafer A, Gilbert R, Gadoury C, et al: Programmable attenuation of antigenic sensitivity for a nanobody-based EGFR chimeric antigen receptor through hinge domain truncation. Front Immunol. 13:8648682022. View Article : Google Scholar : PubMed/NCBI | |
|
Hosking MP, Shirinbak S, Omilusik K, Chandra S, Kaneko MK, Gentile A, Yamamoto S, Shrestha B, Grant J, Boyett M, et al: Preferential tumor targeting of HER2 by iPSC-derived CAR T cells engineered to overcome multiple barriers to solid tumor efficacy. Cell Stem Cell. 32:1087–1101.e4. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Bakhtiari SH, Rahbarizadeh F, Hasannia S, Ahmadvand D, Iri-Sofla FJ and Rasaee MJ: Anti-MUC1 nanobody can redirect T-body cytotoxic effector function. Hybridoma (Larchmt). 28:85–92. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
You F, Jiang L, Zhang B, Lu Q, Zhou Q, Liao X, Wu H, Du K, Zhu Y, Meng H, et al: Phase 1 clinical trial demonstrated that MUC1 positive metastatic seminal vesicle cancer can be effectively eradicated by modified Anti-MUC1 chimeric antigen receptor transduced T cells. Sci China Life Sci. 59:386–397. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Supimon K, Sangsuwannukul T, Sujjitjoon J, Chieochansin T, Junking M and Yenchitsomanus PT: Cytotoxic activity of anti-mucin 1 chimeric antigen receptor T cells expressing PD-1-CD28 switch receptor against cholangiocarcinoma cells. Cytotherapy. 25:148–161. 2023. View Article : Google Scholar | |
|
Ishikawa Y, Tanaka N, Murakami K, Uchiyama T, Kumaki S, Tsuchiya S, Kugoh H, Oshimura M, Calos MP and Sugamura K: Phage phiC31 integrase-mediated genomic integration of the common cytokine receptor gamma chain in human T-cell lines. J Gene Med. 8:646–653. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Rath P, Kramer P, Biggs D, Preece C, Hortin N, Diaz R, Perez-Alcantara M, Li X, Bolard A, Beer N, et al: Optimizing approaches for targeted integration of transgenic cassettes by integrase-mediated cassette exchange in mouse and human stem cells. Stem Cells. 43:sxae0922025. View Article : Google Scholar : PubMed/NCBI | |
|
Li M, Choo B, Wong ZM, Filmus J and Buick RN: Expression of OCI-5/glypican 3 during intestinal morphogenesis: Regulation by cell shape in intestinal epithelial cells. Exp Cell Res. 235:3–12. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Arbabi-Ghahroudi M, Tanha J and MacKenzie R: Isolation of monoclonal antibody fragments from phage display libraries. Methods Mol Biol. 502:341–364. 2009. View Article : Google Scholar | |
|
Zhou L, Li Y, Zheng D, Zheng Y, Cui Y, Qin L, Tang Z, Peng D, Wu Q, Long Y, et al: Bispecific CAR-T cells targeting FAP and GPC3 have the potential to treat hepatocellular carcinoma. Mol Ther Oncol. 32:2008172024. View Article : Google Scholar : PubMed/NCBI | |
|
Li N, Fu H, Hewitt SM, Dimitrov DS and Ho M: Therapeutically targeting glypican-2 via single-domain antibody-based chimeric antigen receptors and immunotoxins in neuroblastoma. Proc Natl Acad Sci USA. 114:E6623–E6631. 2017.PubMed/NCBI | |
|
Astrof S, Crowley D, George EL, Fukuda T, Sekiguchi K, Hanahan D and Hynes RO: Direct test of potential roles of EIIIA and EIIIB alternatively spliced segments of fibronectin in physiological and tumor angiogenesis. Mol Cell Biol. 24:8662–8670. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Efthymiou G, Saint A, Ruff M, Rekad Z, Ciais D and Van Obberghen-Schilling E: Shaping Up the tumor microenvironment with cellular fibronectin. Front Oncol. 10:6412020. View Article : Google Scholar : PubMed/NCBI | |
|
Ma W, Wang Y, Zhang R, Yang F, Zhang D, Huang M, Zhang L, Dorsey JF, Binder ZA, O'Rourke DM, et al: Targeting PAK4 to reprogram the vascular microenvironment and improve CAR-T immunotherapy for glioblastoma. Nat Cancer. 2:83–97. 2021. View Article : Google Scholar | |
|
Lutz EA, Jailkhani N, Momin N, Huang Y, Sheen A, Kang BH, Wittrup KD and Hynes RO: Intratumoral nanobody-IL-2 fusions that bind the tumor extracellular matrix suppress solid tumor growth in mice. PNAS Nexus. 1:pgac2442022. View Article : Google Scholar | |
|
Xie YJ, Dougan M, Jailkhani N, Ingram J, Fang T, Kummer L, Momin N, Pishesha N, Rickelt S, Hynes RO and Ploegh H: Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc Natl Acad Sci USA. 116:7624–7631. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Alon-Zchut G, Zalk R, Huynh TT, Zalutsky MR, Weizmann Y, Zarivach R and Papo N: Structural analysis of nanobody interactions with their prostate-specific membrane antigen binding epitopes. Int J Biol Macromol. 320(Pt 1): 1456932025. View Article : Google Scholar : PubMed/NCBI | |
|
Chatalic KL, Veldhoven-Zweistra J, Bolkestein M, Hoeben S, Koning GA, Boerman OC, de Jong M and van Weerden WM: A novel 111in-labeled anti-prostate-specific membrane antigen nanobody for targeted SPECT/CT imaging of prostate cancer. J Nucl Med. 56:1094–1099. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Georgiev T, Principi L, Galbiati A, Gilardoni E, Neri D and Cazzamalli S: Targeted interleukin-2 enhances the in vivo anti-cancer activity of Pluvicto™. Eur J Nucl Med Mol Imaging. 51:2332–2337. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Hassani M, Hajari Taheri F, Sharifzadeh Z, Arashkia A, Hadjati J, van Weerden WM, Abdoli S, Modarressi MH and Abolhassani M: Engineered jurkat cells for targeting prostate-specific membrane antigen on prostate cancer cells by nanobody-based chimeric antigen receptor. Iran Biomed J. 24:81–88. 2020. View Article : Google Scholar : | |
|
Ullah K, Addai Peprah F, Yu F and Shi H: The application of prostate specific membrane antigen in CART-cell therapy for treatment of prostate carcinoma (Review). Oncol Rep. 40:3136–3143. 2018.PubMed/NCBI | |
|
Khaleghi S, Rahbarizadeh F, Ahmadvand D, Rasaee MJ and Pognonec P: A caspase 8-based suicide switch induces apoptosis in nanobody-directed chimeric receptor expressing T cells. Int J Hematol. 95:434–444. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Wang Q, Li L, Gao F, Zhang Y and Zhu Y: The peptide-based bispecific CAR T cells target EGFR and tumor stroma for effective cancer therapy. Int J Pharm. 663:1245582024. View Article : Google Scholar : PubMed/NCBI | |
|
De Munter S, Ingels J, Goetgeluk G, Bonte S, Pille M, Weening K, Kerre T, Abken H and Vandekerckhove B: Nanobody based dual specific CARs. Int J Mol Sci. 19:4032018. View Article : Google Scholar : PubMed/NCBI | |
|
Xia B, Lin K, Wang X, Chen F, Zhou M, Li Y, Lin Y, Qiao Y, Li R, Zhang W, et al: Nanobody-derived bispecific CAR-T cell therapy enhances the anti-tumor efficacy of T cell lymphoma treatment. Mol Ther Oncolytics. 30:86–102. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Xu J, Chen LJ, Yang SS, Sun Y, Wu W, Liu YF, Xu J, Zhuang Y, Zhang W, Weng XQ, et al: Exploratory trial of a biepitopic CAR T-targeting B cell maturation antigen in relapsed/refractory multiple myeloma. Proc Natl Acad Sci USA. 116:9543–9551. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Meeus F, Funeh CN, Awad RM, Zeven K, Autaers D, De Becker A, Van Riet I, Goyvaerts C, Tuyaerts S, Neyns B, et al: Preclinical evaluation of antigen-sensitive B7-H3-targeting nanobody-based CAR-T cells in glioblastoma cautions for on-target, off-tumor toxicity. J Immunother Cancer. 12:e0091102024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Arulraj T, Anbari S and Popel AS: Quantitative systems pharmacology modeling of macrophage-targeted therapy combined with PD-L1 inhibition in advanced NSCLC. Clin Transl Sci. 17:e138112024. View Article : Google Scholar : PubMed/NCBI | |
|
Xie YJ, Dougan M, Ingram JR, Pishesha N, Fang T, Momin N and Ploegh HL: Improved antitumor efficacy of chimeric antigen receptor T cells that secrete single-domain antibody fragments. Cancer Immunol Res. 8:518–529. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lebrec H, Maier CC, Maki K, Ponce R, Shenton J and Green S: Nonclinical safety assessment of engineered T cell therapies. Regul Toxicol Pharmacol. 127:1050642021. View Article : Google Scholar : PubMed/NCBI | |
|
Kegler A, Koristka S, Bergmann R, Berndt N, Arndt C, Feldmann A, Hoffmann A, Bornhäuser M, Schmitz M and Bachmann MP: T cells engrafted with a UniCAR 28/z outperform UniCAR BB/z-transduced T cells in the face of regulatory T cell-mediated immunosuppression. Oncoimmunology. 8:e16216762019. View Article : Google Scholar : PubMed/NCBI | |
|
Albert S, Arndt C, Feldmann A, Bergmann R, Bachmann D, Koristka S, Ludwig F, Ziller-Walter P, Kegler A, Gärtner S, et al: A novel nanobody-based target module for retargeting of T lymphocytes to EGFR-expressing cancer cells via the modular UniCAR platform. Oncoimmunology. 6:e12872462017. View Article : Google Scholar : PubMed/NCBI | |
|
Albert S, Arndt C, Koristka S, Berndt N, Bergmann R, Feldmann A, Schmitz M, Pietzsch J, Steinbach J and Bachmann M: From mono- to bivalent: Improving theranostic properties of target modules for redirection of UniCAR T cells against EGFR-expressing tumor cells in vitro and in vivo. Oncotarget. 9:25597–25616. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Berdeja JG, Madduri D, Usmani SZ, Jakubowiak A, Agha M, Cohen AD, Stewart AK, Hari P, Htut M, Lesokhin A, et al: Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): A phase 1b/2 open-label study. Lancet. 398:314–324. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Munshi NC, Anderson LD Jr, Shah N, Madduri D, Berdeja J, Lonial S, Raje N, Lin Y, Siegel D, Oriol A, et al: Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med. 384:705–716. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Raje N, Berdeja J, Lin Y, Siegel D, Jagannath S, Madduri D, Liedtke M, Rosenblatt J, Maus MV, Turka A, et al: Anti-BCMA CAR T-Cell Therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med. 380:1726–1737. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao WH, Liu J, Wang BY, Chen YX, Cao XM, Yang Y, Zhang YL, Wang FX, Zhang PY, Lei B, et al: A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma. J Hematol Oncol. 11:1412018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao WH, Wang BY, Chen LJ, Fu WJ, Xu J, Liu J, Jin SW, Chen YX, Cao XM, Yang Y, et al: Four-year follow-up of LCAR-B38M in relapsed or refractory multiple myeloma: A phase 1, single-arm, open-label, multicenter study in China (LEGEND-2). J Hematol Oncol. 15:862022. View Article : Google Scholar | |
|
Kamdar M, Solomon SR, Arnason J, Johnston PB, Glass B, Bachanova V, Ibrahimi S, Mielke S, Mutsaers P, Hernandez-Ilizaliturri F, et al: Lisocabtagene maraleucel versus standard of care with salvage chemotherapy followed by autologous stem cell transplantation as second-line treatment in patients with relapsed or refractory large B-cell lymphoma (TRANSFORM): Results from an interim analysis of an open-label, randomised, phase 3 trial. Lancet. 399:2294–2308. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Locke FL, Miklos DB, Jacobson CA, Perales MA, Kersten MJ, Oluwole OO, Ghobadi A, Rapoport AP, McGuirk J, Pagel JM, et al: Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma. N Engl J Med. 386:640–654. 2022. View Article : Google Scholar | |
|
Gazeau N, Liang EC, Wu QV, Voutsinas JM, Barba P, Iacoboni G, Kwon M, Ortega JLR, López-Corral L, Hernani R, et al: Anakinra for refractory cytokine release syndrome or immune effector cell-associated neurotoxicity syndrome after chimeric antigen receptor T cell therapy. Transplant Cell Ther. 29:430–437. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sborov DW, Pawlyn C, Ishida T, Huang JSY, Benjamin R, Iida S, Popat R, Kuroda J, Pianko MJ, Ramakrishnan A, et al: Evaluation of cytokine release syndrome (CRS) in patients with relapsed or refractory multiple myeloma (RRMM) receiving step-up priming doses and longer dosing intervals of elranatamab: MagnetisMM-9. J Clin Oncol. 42:75222024. View Article : Google Scholar | |
|
Martin T, Usmani SZ, Berdeja JG, Agha M, Cohen AD, Hari P, Avigan D, Deol A, Htut M, Lesokhin A, et al: Ciltacabtagene autoleucel, an anti-B-cell maturation antigen chimeric antigen receptor T-cell therapy, for relapsed/refractory multiple myeloma: CARTITUDE-1 2-year follow-up. J Clin Oncol. 41:1265–1274. 2023. View Article : Google Scholar | |
|
Han L, Gao Q, Zhou K, Zhou J, Yin Q, Fang B, Zhu XH, Zhang J, Bao C and Song Y: The clinical study of anti-BCMA CAR-T with single-domain antibody as antigen binding domain. J Clin Oncol. 39:80252021. View Article : Google Scholar | |
|
Wu A, Zhang T, Yu H, Cao Y, Zhang R, Shao R, Liu B, Chen L, Xu K, Chen W, et al: Mechanisms underlying resistance to CAR-T cell therapy and strategies for enhancement. Cytokine Growth Factor Rev. 83:66–76. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Kearl TJ, Furqan F and Shah NN: CAR T-cell therapy for B-cell lymphomas: Outcomes and resistance mechanisms. Cancer Metastasis Rev. 44:122024. View Article : Google Scholar : PubMed/NCBI | |
|
Safarzadeh Kozani P, Safarzadeh Kozani P and Rahbarizadeh F: Humanization of the antigen-recognition domain does not impinge on the antigen-binding, cytokine secretion, and antitumor reactivity of humanized nanobody-based CD19-redirected CAR-T cells. J Transl Med. 22:6792024. View Article : Google Scholar : PubMed/NCBI | |
|
Nasiri F, Safarzadeh Kozani P and Rahbarizadeh F: T-cells engineered with a novel VHH-based chimeric antigen receptor against CD19 exhibit comparable tumoricidal efficacy to their FMC63-based counterparts. Front Immunol. 14:10638382023. View Article : Google Scholar : PubMed/NCBI | |
|
Krol VE, Bansal A, Kethamreddy M, Ellinghuysen JR, Vail DJ, Lucien-Matteoni F, Dong H, Park SS and Pandey MK: Synthesis and in vitro evaluation of a scandium-44 radiolabeled nanobody as a PD-L1 PET imaging probe. Pharmaceutics. 17:7962025. View Article : Google Scholar : PubMed/NCBI | |
|
Bolzati C and Spolaore B: Enzymatic methods for the site-specific radiolabeling of targeting proteins. Molecules. 26:34922021. View Article : Google Scholar : PubMed/NCBI | |
|
Hanke L, Sheward DJ, Pankow A, Vidakovics LP, Karl V, Kim C, Urgard E, Smith NL, Astorga-Wells J, Ekström S, et al: Multivariate mining of an alpaca immune repertoire identifies potent cross-neutralizing SARS-CoV-2 nanobodies. Sci Adv. 8:eabm02202022. View Article : Google Scholar : PubMed/NCBI | |
|
Takeuchi T, Kawanishi M, Nakanishi M, Yamasaki H and Tanaka Y: Phase II/III results of a trial of anti-tumor necrosis factor multivalent NANOBODY compound ozoralizumab in patients with rheumatoid arthritis. Arthritis Rheumatol. 74:1776–1785. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yuen D, Feeney OM, Noi L, Shengule S, McLeod VM, Reitano P, Tsegay S, Hufton R, Houston ZH, Fletcher NL, et al: Nanobody-mediated cellular uptake maximizes the potency of polylysine dendrimers while preserving solid tumor penetration. ACS Nano. 19:6044–6057. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Olkowski CP, Basuli F, Fernandes B, Ghaemi B, Shi J, Zhang HH, Farber JM, Escorcia FE, Choyke PL and Jacobson O: Comparative kidney uptake of nanobody-based PET tracers labeled with various fluorine-18-labeled prosthetic groups. Mol Pharm. 22:533–543. 2025. View Article : Google Scholar | |
|
Chen Y, Xiong T, Peng Q, Du J, Sun W, Fan J and Peng X: Self-reporting photodynamic nanobody conjugate for precise and sustainable large-volume tumor treatment. Nat Commun. 15:69352024. View Article : Google Scholar : PubMed/NCBI | |
|
Heremans J, Maximilian Awad R, Bridoux J, Ertveldt T, Caveliers V, Madder A, Hoogenboom R, Devoogdt N, Ballet S, Hernot S, et al: Sustained release of a human PD-L1 single-domain antibody using peptide-based hydrogels. Eur J Pharm Biopharm. 196:1141832024. View Article : Google Scholar : PubMed/NCBI | |
|
Schubert ML, Schmitt A, Hückelhoven-Krauss A, Neuber B, Kunz A, Waldhoff P, Vonficht D, Yousefian S, Jopp-Saile L, Wang L, et al: Treatment of adult ALL patients with third-generation CD19-directed CAR T cells: Results of a pivotal trial. J Hematol Oncol. 16:792023. View Article : Google Scholar : PubMed/NCBI | |
|
Ren Q, Zu Y, Su H, Lu Q, Xiang B, Luo Y, Zhang J and Song Y: Single VHH-directed BCMA CAR-NK cells for multiple myeloma. Exp Hematol Oncol. 12:982023. View Article : Google Scholar : PubMed/NCBI | |
|
Sadeghi A, Behdani M, Muyldermans S, Habibi-Anbouhi M and Kazemi-Lomedasht F: Development of a mono-specific anti-VEGF bivalent nanobody with extended plasma half-life for treatment of pathologic neovascularization. Drug Test Anal. 12:92–100. 2020. View Article : Google Scholar | |
|
Budi HS, Ahmad FN, Achmad H, Ansari MJ, Mikhailova MV, Suksatan W, Chupradit S, Shomali N and Marofi F: Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor (CAR) for tumor immunotherapy; recent progress. Stem Cell Res Ther. 13:402022. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Zhong D, Luo H, Shi W, Xie S, Qiang H, Zhu L, Gao L, Liu J, Sun S, et al: Nanobody-based CAR T cells targeting intracellular tumor antigens. Biomed Pharmacother. 156:1139192022. View Article : Google Scholar : PubMed/NCBI | |
|
Heitzeneder S, Bosse KR, Zhu Z, Zhelev D, Majzner RG, Radosevich MT, Dhingra S, Sotillo E, Buongervino S, Pascual-Pasto G, et al: GPC2-CAR T cells tuned for low antigen density mediate potent activity against neuroblastoma without toxicity. Cancer Cell. 40:53–69.e9. 2022. View Article : Google Scholar : | |
|
Mailankody S, Jakubowiak AJ, Htut M, Costa LJ, Lee K, Ganguly S, Kaufman JL, Siegel DSD, Bensinger W, Cota M, et al: Orvacabtagene autoleucel (orva-cel), a B-cell maturation antigen (BCMA)-directed CAR T cell therapy for patients (pts) with relapsed/refractory multiple myeloma (RRMM): Update of the phase 1/2 EVOLVE study (NCT03430011). J Clin Oncol. 38:85042020. View Article : Google Scholar |