Next Article in Journal
The DEAD-Box Protein Rok1 Coordinates Ribosomal RNA Processing in Association with Rrp5 in Drosophila
Next Article in Special Issue
EpCAM- and EGFR-Specific Antibody Drug Conjugates for Triple-Negative Breast Cancer Treatment
Previous Article in Journal
SK119, a Novel Shikonin Derivative, Leads to Apoptosis in Melanoma Cell Lines and Exhibits Synergistic Effects with Vemurafenib and Cobimetinib
Previous Article in Special Issue
Reply to Sheval, E.V. Comment on “Piña et al. Ten Approaches That Improve Immunostaining: A Review of the Latest Advances for the Optimization of Immunofluorescence. Int. J. Mol. Sci. 2022, 23, 1426”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy

1
Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea
2
R&D Division, GC Biopharma, Yongin 16924, Korea
3
Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Korea
4
Antibody Research Institute, Kookmin University, Seoul 02707, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(10), 5686; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23105686
Submission received: 20 April 2022 / Revised: 16 May 2022 / Accepted: 17 May 2022 / Published: 19 May 2022

Abstract

:
Cancer is the second leading cause of death worldwide after cardiovascular diseases. Harnessing the power of immune cells is a promising strategy to improve the antitumor effect of cancer immunotherapy. Recent progress in recombinant DNA technology and antibody engineering has ushered in a new era of bispecific antibody (bsAb)-based immune-cell engagers (ICEs), including T- and natural-killer-cell engagers. Since the first approval of blinatumomab by the United States Food and Drug Administration (US FDA), various bsAb-based ICEs have been developed for the effective treatment of patients with cancer. Simultaneously, several potential therapeutic targets of bsAb-based ICEs have been identified in various cancers. Therefore, this review focused on not only highlighting the action mechanism, design and structure, and status of bsAb-based ICEs in clinical development and their approval by the US FDA for human malignancy treatment, but also on summarizing the currently known and emerging therapeutic targets in cancer. This review provides insights into practical considerations for developing next-generation ICEs.

1. Introduction

Cancer is one of the major leading causes of death worldwide. In 2020, nearly 19.3 million new cancer cases and 10.0 million cancer deaths were reported globally. More specifically, the most common cancers among new cases were breast cancer (11.7%), followed by lung (11.4%), colorectal (10.0%), prostate (7.3%), and stomach (5.6%) cancers [1]. The order of the mortality rate on the basis of cancer types was as follows: lung (18%), colorectal (9.4%), liver (8.3%), stomach (7.7%), and breast (6.9%) cancers [2]. By 2040, the number of new cancer cases is expected to be approximately 28.3 million, which is nearly >50% from that reported in 2020 [3]. Currently, various therapeutic regimens, such as surgical resection, chemotherapy, antibody therapy, radiotherapy, and combination therapy, have been used in clinical practice for the effective treatment of patients with cancers, depending on their health conditions and cancer status [4].
One of the most promising targeted therapies for cancer treatment is antibody therapy. It has a superior targeting ability for antigens that are expressed on cancer cells, which results in prominent antitumor activity and lower toxicity, compared with that of chemotherapeutic agents [5]. As of December 2021, 110 therapeutic antibodies, including monoclonal antibodies (mAbs), bispecific antibodies (bsAbs), and antibody–drug conjugates (ADCs), have been approved by the United States Food and Drug Administration (US FDA) and/or the European Medicines Agency (EMA). Among them, 46 antibodies are indicated for cancer treatment [6]. Generally, immunoglobulin (Ig) G-based mAb—the most widely used mAb form for antibody therapy—comprises two heavy and two light chains. The light chain has one variable (VL) and one constant (CL) domain, whereas the heavy chain has one variable (VH) and three constant (CH; CH1–CH3) domains [7,8]. Furthermore, the fragment antigen-binding (Fab) region of mAb plays a key role in cancer therapy, and specifically in modulating or blocking the signaling pathways that are involved in cancer development; on the other hand, the fragment crystallizable (Fc) region interacts with Fc receptors that are expressed on immune cells and participates in various effector functions, such as killing cancer cells via antigen-dependent T-cellular cytotoxicity (ADCC) [9,10].
BsAbs harness the specificities of two antibodies and combine them to simultaneously recognize two independent epitopes or antigens [11]. More specifically, these antibodies are designed and manufactured to contain two target-binding units in one antibody-based molecule, whereby each unit independently recognizes its unique epitope, through quadromas, chemical conjugation, or genetic recombination [12]. Compared with mAbs in cancer therapy, bsAbs have several potential benefits, such as the improvement in therapeutic efficacy, the enhancement of tumor-cell selectivity, and the reduction in tumor-cell resistance [13]. In 2009, catumaxomab—a rat–mouse hybrid bsAb for the CD3 epithelial cell adhesion molecule (EpCAM)—was first approved by the EMA for the treatment of malignant ascites in patients with EpCAM-positive cancer [14]. However, it was voluntarily withdrawn from the US market in 2013, and from the European Union (EU) market in 2017, because of commercial reasons [15]. Since the US FDA approval of blinatumomab—a CD19 × CD3 mouse bispecific T-cell engager (BiTE) antibody—for the treatment of acute lymphoblastic leukemia (ALL) in 2014, much attention has been paid to the development of bsAb-based immune-cell engagers (ICEs) that redirect immune effector cells against cancer cells and promote antitumor activities [16,17]. Furthermore, compared with adoptive immune-cell therapy, which requires an expensive and complicated manufacturing process, ICE has become a feasible therapeutic cancer-therapy approach. Currently, increasing numbers of bsAb-based ICEs are extensively evaluated in clinical trials worldwide [18,19].
Herein, we focus on highlighting the action mechanisms, structures, roles, and relevance of the currently known and potential therapeutic targets of bsAb-based ICEs in cancers. In addition, this review covers the current developmental status of bsAb-based ICEs approved by the US FDA and/or the EMA, or in clinical development for cancer therapy. This review provides insights into practical considerations for the development of the next-generation bsAb-based ICEs.

2. Action Mechanism of bsAb-Based ICEs in Cancer

BsAb-based ICEs play a key role in cancer immunotherapy, and specifically in recruiting and engaging immune effector cells that are proximal to tumor-associated antigens (TAA) that are expressed on cancer cells and that allow the formation of immune synapses and a specialized cell–cell junction between the immune and cancer cells [20,21]. Ultimately, these immune synapses promote the elimination of target cancer cells [22]. These bsAb-based ICEs are currently classified into T- and natural killer (NK)-cell engagers (Figure 1).

2.1. T-Cell Engagers

T-cell engagers are engineered bsAbs that redirect and activate T-cells to induce the robust elimination of poorly immunogenic tumors [23]. Most T-cell engagers comprise two linked antigen-binding variable fragments (Fvs) that specifically target TAAs and the CD3 unit of the T-cell-receptor (TCR) complex, which thereby engages T-cells to form an immune synapse on the surface of tumor cells [24,25]. Generally, the TCR complex on T-cells directly interacts with antigens that are presented on the major-histocompatibility-complex (MHC) molecules of target T-cells, and this interaction plays a pivotal role in T-cell activation and target T-cell killing [26]. However, most tumor cells are known to exhibit the loss or depletion of MHC expression, which hampers the antitumor effects of activated T-cells [27]. The unique feature of T-cell engagers is the redirection of T-cells against TAAs on tumor cells, as well as the direct activation of T-cells without TCR/MHC interaction [28]. Simultaneously, it links T-cells with tumor cells to form immune synapses, wherein the activated T-cells release perforins to form pores on the surface of cancer cells, and granzymes to proteolyze cellular proteins. Eventually, these immune responses lead to the cell death (also known as apoptosis) of tumors [22,29,30]. Moreover, T-cell activation by an anti-CD3 arm of a T-cell engager induces cytokine secretion and concomitant T-cell proliferation that sustain durable antitumor immune responses [31]. Currently, 47 bsAb-based T-cell engagers have been studied in clinical trials (Table 1).

2.2. NK-Cell Engagers

NK cells are effector lymphocytes of the innate immune system that play protective roles against both infectious pathogens and cancers [32]. NK cells are divided into several subpopulations on the basis of the relative expressions of the adhesion molecule CD56 and the activating receptor CD16 (FcγRIIIa) [33]. The CD56dim CD16bright NK cells represent at least 90% of all peripheral blood NK cells [34]. Currently, CD16 is the most important NK-cell target for the NK-cell engagers. CD16-targeting NK-cell engagers lead to both NK-cell activation and tumor-cell-specific cytotoxicity [35]. More specifically, NK-cell engagers recruit NK cells toward target tumor cells by binding a tumor-specific antigen with one arm of the engagers, and by bridging CD16 onto NK cells with the other [36]. Then, the NK-cell engagers trigger the death of target cancer cells by not only releasing cytotoxic granules that contain granzymes and perforin, but also by secreting chemokines and cytokines, such as those regulated on activation, normal T-cell expressed and secreted, and interferon-gamma [32]. AFM13 is a bsAb-based NK-cell engager that specifically targets CD16 and CD30; it is currently under clinical development for use in the treatment of hematological malignancy (Table 1).

3. Role of Known and Emerging Targets of ICEs

3.1. Single-Pass ICE Targets in Solid Cancers

3.1.1. Delta-Like Ligand 3

Delta-like ligand 3 (DLL3) is a 65-kDa type I transmembrane protein and a Notch receptor ligand. It plays an important role in the regulation of Notch signaling [37]. In small-cell lung cancer (SCLC), DLL3 has been reported as a key factor in the promotion of the tumor growth, migration, and invasion of SCLC cells. Several lines of evidence support this notion [37,38]. Upregulated DLL3 expression was verified to promote tumor growth in a mouse xenograft model that was implanted with DLL3-overexpressing SBC-5 human SCLC cells. Additionally, DLL3 knockdown reduces SCLC-cell migration and invasion, whereas its overexpression in the cells increases these activities [38]. This protein is highly upregulated and aberrantly expressed in SCLC and other neuroendocrine malignancies, but not in nonmalignant T-cells [37,39]. Currently, DLL3 is considered an attractive novel potential therapeutic target in neuroendocrine tumors (NETs), including SCLC. A preclinical study on robalpituzumab tesirin—an ADC that targets DLL3—showed a dose-dependent reduction in the tumor size with a complete response (CR) in B6129SF1/J mice that were implanted with DLL3-positive KP1 SCLC cells, which led to the absence of measurable tumors for >80 days after treatment [40].

3.1.2. Epidermal Growth Factor Receptor

Epidermal growth factor receptor (EGFR) is a 170-kDa receptor tyrosine kinase that belongs to the ErbB family and that comprises two major functional domains—the extracellular and cytoplasmic domains—and a tyrosine kinase domain that is linked by a single transmembrane region [41,42]. EGF binding to the receptor induces the dimerization of the receptor; triggers the autophosphorylation of cytoplasmic tyrosine residues; and eventually participates in the regulation of cell proliferation, migration, and adhesion [41,43,44,45]. EGFR is overexpressed in various cancers, such as colorectal cancer (CRC), lung cancer, breast cancer, glioblastoma, and head and neck squamous cell carcinoma. EGFR overexpression in CRC has been closely associated with tumor progression and poor prognosis [46,47,48,49,50]. Currently, EGFR is one of the most well-known therapeutic targets in various cancers. Phase II clinical studies on cetuximab—a human/mouse chimeric mAb that targets EGFR in advanced CRC—have demonstrated that the use of cetuximab as monotherapy exerts anticancer effects with approximately 10% partial response (PR) and 33% stable disease (SD) [51].

3.1.3. EpCAM

EpCAM is a 40-kDa type I transmembrane glycoprotein that plays a key role in the regulation of cell adhesion, proliferation, and differentiation [52,53,54]. EpCAM is overexpressed in various cancers, such as ovarian cancer, CRC, breast cancer, lung cancer, and pancreatic cancer [55,56,57,58]. Its protease-cleaved intracellular domain associates with β-catenin to form a nuclear protein complex that is translocated to the nucleus, activates the transcription of genes that are involved in cancer-cell proliferation, and results in tumorigenesis [52]. Several studies have suggested EpCAM as a potential target for antibody therapy against cancers. For instance, a phase II clinical study on adecatumumab (MT201)—a fully human mAb that targets EpCAM in metastatic breast cancer—reports that, of 112 patients treated with adecatumumab, 2 showed a PR and 10 had SD, according to the response evaluation criteria in solid tumors (RECIST) [59].

3.1.4. Glycoprotein A33

Glycoprotein A33 (GPA33)—also known as cell surface A33 antigen—is a 43-kDa cell surface differentiation glycoprotein that belongs to the type I transmembrane protein family [60,61,62]. It is associated with cell–cell adhesion [60]. GPA33 is highly overexpressed in >95% of human CRCs but is not detected in any other tissues [62]. Several studies have indicated GPA33 as a potential target in immunotherapy against CRC. For instance, in vivo studies on KRN330—a human mAb that targets GPA33—have demonstrated its dose-dependent antitumor activities in mouse and rat xenograft models implanted with LS174T human CRC cells [63,64].

3.1.5. Human EGFR 2

Human EGFR 2 (HER2) is a member of the ErbB family and is a 185-kDa single-pass transmembrane receptor. To the best of our knowledge, direct ligands for HER2 have not been identified yet. HER2 activation is achieved through homo- or heterodimerization with HER2 or other ErbB-family receptor members, including EGFR and HER3 [65,66,67]. It is overexpressed in various cancers, such as breast, gastric/gastroesophageal, and colon cancers [68,69,70]. HER2 is closely associated with cancer-cell proliferation and invasion, as well as with tumor growth [71,72]. Particularly in breast cancers, HER2 is overexpressed in 15–30% of the total patients with breast cancers [65]. Substantial evidence has shown that HER2 is an important predictive biomarker in HER2-targeted therapies, and a well-known therapeutic target in breast cancers. Trastuzumab is the first anti-HER2 humanized mAb that targets HER2 in breast cancer. In a phase III clinical study, patients with breast cancer treated with trastuzumab combined with chemotherapy had a longer survival (median survival, 25.1 vs. 20.3 months) and prolonged disease progression (median, 7.4 vs. 4.6 months) than those treated with chemotherapy alone [73].

3.1.6. Mucin 16

Mucin 16 (MUC16)—also known as human carbohydrate antigen 125 (CA-125)—is a heavily glycosylated 300–500-kDa type I transmembrane protein [74,75,76]. It is a biomarker for ovarian cancer. MUC16 is overexpressed in ovarian cancer and contributes to ovarian-cancer progression and metastasis [76,77]. Increased MUC16 expression is associated with poor prognosis in patients with ovarian cancer [78]. Some studies have reported that MUC16 is a potential target for antibody therapy against ovarian cancers. Oregovomab is a mouse mAb that targets MUC16 in advanced ovarian cancer. In a phase II clinical study, 145 patients with stage III/IV ovarian cancer were randomized to receive oregovomab (n = 73) or placebo (n = 72). The time to recurrence was prolonged in the oregovomab group (24.0 months) compared with that in the placebo group (10.8 months) [79].

3.1.7. Mucin 17

Mucin 17 (MUC17) is a 452-kDa type I membrane-associated mucin that is expressed on the apical surface of gastrointestinal epithelial cells [80,81,82]. As a key component of the mucosal layer, MUC17 has been suggested to play a crucial role in the restoration and protection of epithelial cells [80]. Recent studies have demonstrated that aberrant MUC17 overexpression is correlated with the malignant potential of gastric and pancreatic cancers [81,83]. Particularly in gastric-cancer tissues, MUC17 is overexpressed in approximately 50% of the gastric-cancer cases. Thus, MUC17 is a compelling target in gastric cancer because of its prevalent expression on tumor cells compared with its low, relatively restricted expression in normal tissues [84].

3.1.8. Prostate-Specific Membrane Antigen

Prostate-specific membrane antigen (PSMA) is a 100-kDa type II membrane protein, is exclusively overexpressed in prostate cancer, and acts as a glutamate-preferring carboxypeptidase. Its expression is associated with tumor invasiveness [85,86]. PSMA is not only a well-known biomarker but is also a potential therapeutic target in prostate cancer. Its expression is 100–1000-fold higher in prostate-cancer tissue than in normal tissue, and it is present on the cell surface without being released into the circulation [87,88,89,90]. Furthermore, J591 was recently developed as the first humanized mAb that targets the extracellular domain of PSMA in prostate cancer. A phase I/II study was conducted to evaluate the safety and efficacy of J591 in patients with metastatic castration-resistant prostate cancer (mCRPC). Of the 23 patients with measurable disease, 14 (60.8%) had SD and 6 (26.1%) had progressive disease, according to the RECIST [91].

3.2. Multi-Pass Transmembrane Proteins as ICE Targets in Solid Cancers

3.2.1. Claudin-18 Isoform 2

Claudin-18 isoform 2 (CLDN18.2) is a 23-kDa tetra-transmembrane protein. It plays an important role in the regulation of tight junction formation and cell adhesion [92,93,94]. It is known as a tumor-specific marker in gastric or gastroesophageal junction (GEJ) cancers because it is overexpressed exclusively in primary gastric malignancies, but not in any healthy tissues, except stomach mucosa [90,92,95]. Some studies have suggested that CLDN18.2 is a target for antibody therapy against cancer. Claudiximab (IMAB362) is a chimeric mAb that targets CLDN18.2 in gastric cancer. In a phase II study, patients with advanced/recurrent gastric and GEJ cancers who were treated with claudiximab combined with chemotherapy exhibited a significantly improved progression-free survival (PFS) (median, 7.9 vs. 4.8 months) and prolonged overall survival (OS) (median, 13.3 vs. 8.4 months) compared with those treated with chemotherapy alone [96].

3.2.2. Six-Transmembrane Epithelial Antigen of Prostate 1

Six-transmembrane epithelial antigen of prostate 1 (STEAP1) is a 39-kDa integral membrane protein that comprises six transmembrane helices [97,98]. In normal cells, STEAP1 plays a key role in the regulation of cell migration and proliferation, despite its low expression or absence in normal tissues [97,99]. It is highly overexpressed in various cancers, and particularly in prostate cancer, wherein it is involved in the regulation of various functions, such as cancer-cell invasion and proliferation, as well as tumorigenesis [98,99]. The knockdown of STEAP1 has been shown to inhibit T-cell growth in androgen-dependent prostate cancer [100]. Moreover, high STEAP1 expression is closely associated with poor outcomes in patients with prostate cancer [101]. These properties make STEAP1 a potential target for antibody therapy. DSTP3086S—an ADC that targets STEAP1—exhibited antitumor activity in a phase I clinical trial in patients with mCRPC. Of the 46 patients, 2 (4%) showed a PR and 24 (52%) had SD, according to the RECIST [102].

3.2.3. Somatostatin Receptor 2

Somatostatin receptor 2 (SSTR2) is a 41-kDa G protein-coupled receptor (GPCR), which is also known as a seven-transmembrane receptor. It is highly overexpressed in most NETs [103,104,105]. Among the NETs, and particularly in SCLC, the high expression of SSTR2 is closely associated with poor prognosis. Furthermore, the loss of SSTR2 reduced tumor growth in a mouse xenograft model implanted with H1048 human SCLC cells [104]. Several studies have suggested that SSTR2 is a therapeutic target in NETs. For instance, the antitumor efficacy of ADC that targets SSTR2 was evaluated in a mouse xenograft model implanted with BON-1 human NET cells, in which it reduced tumor growth [106].

3.3. Glycosylphosphatidylinositol-Anchored Proteins as ICE Targets in Solid Cancers

3.3.1. Carcinoembryonic Antigen

Carcinoembryonic antigen (CEA) is a 180–200-kDa member of the immunoglobulin supergene family. It plays a key role in the regulation of various cellular functions, such as cell interaction, cell adhesion, and immune response [107,108,109]. CEA is one of the most widely used tumor-marker proteins for various cancers, such as colorectal, gastric, and liver cancers [110,111,112]. It is highly overexpressed in 90% of the total CRC cases, and it is closely associated with poor prognosis in patients with CRC [113]. In CRCs, CEA is involved in cancer progression and metastasis, as well as drug resistance [110,114,115,116,117]. Furthermore, CEA appears to be a potential target for antibody therapy against CRC. A preclinical study on IMMU-130—an ADC that targets CEA—revealed that the ADC efficiently reduced tumor growth in a mouse xenograft model implanted with LS174T human CRC cells [118,119].

3.3.2. Glypican 3

Glypican 3 (GPC3) is a 60-kDa glycosylphosphatidylinositol (GPI)-anchored membrane-bound heparin sulfate proteoglycan. It plays an important role in normal cell growth [120,121]. GPC3 is overexpressed in various cancers, such as hepatocellular carcinoma (HCC), lung squamous cell carcinoma, and ovarian clear cell carcinoma [122,123,124]. In particular, it is highly overexpressed in 70–81% of HCCs; its overexpression correlates with the poor prognosis of patients with HCC [121]. Several studies have suggested that GPC3 is a target for antibody therapy against HCCs. For instance, a preclinical study on GC33—a mAb that targets GPC3—demonstrated its prominent antitumor activity in a mouse xenograft mouse model implanted with SK-HEP-1 human HCCs. The administration of 1 mg/kg of GC33 significantly inhibited tumor growth, and that of 5 mg/kg resulted in tumor remission [125].

3.4. Sphingolipid as ICE Targets in Solid Cancers

GD2

GD2 is a 1.6-kDa glycosylated lipid molecule that belongs to the class of glycosphingolipids [126,127,128,129]. It plays a key role in the attachment of tumor cells to extracellular matrix proteins. It is overexpressed in various cancers, such as neuroblastoma, melanoma, and SCLC, but not in normal tissues [127,130,131,132]. Particularly in SCLC and neuroblastoma, GD2 overexpression is involved in cell proliferation [130]. GD2 has been suggested as a target for antibody therapy against cancer. A phase II clinical study on 3F8—a mouse mAb that targets GD2 in patients with neuroblastoma—revealed that, of 16 patients, 1 showed a CR, and 1 showed a mixed response [133].

3.5. Single Transmembrane Proteins as ICE Targets in Hematological Cancers

3.5.1. B-Cell Maturation Antigen

B-cell maturation antigen (BCMA)—a member of tumor necrosis factor receptor superfamily member 17 (TNFRSF17)—is a 20-kDa type III transmembrane protein [134]. BCMA binds to its ligands, such as proliferation-inducing ligand and B-cell-activating factor, which thus promotes the survival of B-cells [135,136]. It is overexpressed in malignant plasma cells, including multiple myeloma (MM) cells, and it plays a crucial role in the growth of MM [137,138]. A preclinical study that was conducted that used a mouse xenograft model implanted with RPMI 8226 human MM cells has shown that BCMA overexpression promotes tumor growth [137]. Furthermore, high BCMA expression is associated with poor prognosis in patients with MM [139]. Substantial evidence has shown that BCMA is a target for antibody therapy against MM. Preclinical studies on belantamab mafodotin—a US FDA-approved ADC that targets BCMA—showed that it efficiently inhibited tumor growth and prolonged survival in a mouse xenograft model implanted with H929 human MM cells [140,141].

3.5.2. CD19

CD19 is a 95-kDa type I transmembrane protein [142]. It is a coreceptor of B-cell antigen receptor (BCR), and it plays a role in regulating B-cell growth [143,144]. It is overexpressed in most B-cell malignancies, such as ALL, non-Hodgkin lymphoma (NHL), and chronic lymphocytic leukemia (CLL). The overexpression of CD19 promotes the proliferation and survival of these B-cell malignancies [145,146]. Previous studies have suggested that CD19 is an attractive target for antibody therapy against B-cell malignancies. In a phase IIa clinical study on XmAb5574 (MOR00208)—a humanized mAb that targets CD19—of the total patients with relapsed and/or refractory (R/R) NHL who received XmAb5574 monotherapy, 8% showed a CR [147].

3.5.3. CD22

CD22 is a 140-kDa type I transmembrane protein and an inhibitory coreceptor of the BCR that regulates the overstimulation of B-cells [148,149]. CD22 is overexpressed in various B-cell lymphomas, such as CLL, ALL, and NHL, but it is expressed at low levels on immature B-cells and plasma cells [150,151]. Owing to the restricted expression on the B-cell and the inhibitory function of CD22, CD22 has been indicated as a therapeutic target in B-cell lymphoma [152]. Epratuzumab—a humanized mAb that targets CD22—has been reported to be a CD22 agonistic antibody that leads to B-cell inhibition [153]. In a phase II clinical trial, patients with R/R indolent or aggressive NHL were enrolled to receive epratuzumab combined with rituximab, which is a US FDA-approved anti-CD20 mAb. Of the 16 patients with indolent NHL, 9 showed a CR and an unconfirmed CR, and 1 showed a PR. Furthermore, of the six patients with aggressive NHL, three showed a CR, and one showed a PR [154,155].

3.5.4. CD30

CD30 is a 120-kDa type I transmembrane protein that belongs to the tumor necrosis factor receptor family [156]. It plays a key role in lymphocyte activation and proliferation through the nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase pathways that have antiapoptotic and prosurvival benefits [157,158]. It is overexpressed in hematopoietic malignancies, including Hodgkin lymphoma (HL) and NHL, and is associated with the survival of these cells [159,160]. Several studies have suggested that CD30 is a target for antibody therapy against hematologic malignancies. For instance, in vivo studies on XmAb2513—a humanized mAb that targets CD30—showed a significant reduction in the tumor growth, and enhanced survival was observed in a mouse xenograft model implanted with CD30-expressing L540 human HL cells [161].

3.5.5. CD33

CD33—also known as the sialic acid-binding Ig-like lectin 3 (Siglec-3)—is a 67-kDa type I transmembrane protein [162]. It plays a crucial role in the modulation of immune-cell functions, such as phagocytosis, cytokine release, and apoptosis [163,164]. It is overexpressed in acute myeloid lymphoma (AML), and its overexpression is observed in >80% of patients with AML [165]. This increased CD33 expression is correlated with the poor prognosis of patients with AML. In patients with AML who were treated with chemotherapy, the OS rate has been reported to be 42.9% in patients with high CD33 expression, compared with 67.5% in those with low CD33 expression [166]. CD33 is a target for antibody therapy against AML. Gemtuzumab ozogamicin (Mylotarg)—a US FDA-approved ADC that targets CD33—showed promising clinical efficacy in patients with AML [167]. In a phase II clinical study, patients with AML in the first recurrence received gemtuzumab ozogamicin monotherapy; of the 277 patients, 35 showed a CR, and 36 showed a CR with incomplete platelet recovery [168].

3.5.6. CD38

CD38 is a 45-kDa type II transmembrane protein [169]. It is overexpressed in MM cells but shows a low expression in normal lymphoid and myeloid cells [170]. It participates in MM cell survival and proliferation [171]. Previous studies have elucidated that tumor growth decreased in a mouse xenograft model implanted with CD38-knockout RPMI 8226 human MM cells, compared with those nontargeting cells [172]. CD38 is a potential target for antibody therapy against MM. Daratumumab (Darzalex) is the first US FDA-approved human mAb that targets CD38 in the treatment of patients with R/R MM [173]. In a phase III clinical study, patients with R/R MM received chemotherapy (control group) or chemotherapy combined with daratumumab (daratumumab group); the CR rate was significantly higher in the daratumumab group (19.2%) than in the control group (9.0%) [174].

3.5.7. CD123

CD123—the alpha chain of the interleukin-3 (IL-3) receptor—is a 75-kDa type I transmembrane protein [175]. It is overexpressed in leukemic stem cells but shows low or no expression in normal hematopoietic stem cells [176]. CD123 binds to IL-3, which is a hematopoietic growth factor, which leads to the survival and proliferation of various hematologic cancers, such as AML, ALL, and HL [175,177,178,179]. Particularly in AML, increased CD123 expression is associated with a poor prognosis of patients with AML [180]. Previous studies have suggested that CD123 is a target for antibody therapy against AML. In vivo studies on IMGN632—an ADC that targets CD123—have revealed its antitumor activities in a mouse xenograft model implanted with MOLM-13 human AML cells; the mice received IMGN632 or control ADC, and IMGN632 increased the survival of mice compared with the vehicle treatment [181].

3.5.8. C-Type Lectin Domain Family 12 Member A

C-type lectin domain family 12 member A (CLEC12A)—a myeloid inhibitory receptor—is a 31-kDa type II transmembrane protein. It plays a crucial role in the negative regulation of inflammation [182,183]. CLEC12A is specifically expressed in AML and is observed in approximately 90% of patients with AML, but not in normal hematopoietic stem and progenitor cells [184,185]. It is closely associated with the poor prognosis of patients with AML. Previous studies have shown that CLEC12A-positive AML cells are more resistant to chemotherapy than CLEC12A-negative AML cells [186]. Furthermore, the administration of anti-CLEC12A chimeric mAb showed a significant tumor-growth delay of up to 38% in a mouse xenograft model implanted with HL-60 human AML cells [187]. These observations suggest that CLEC12A is a target for antibody therapy against AML.

3.5.9. FMS-Like Tyrosine Kinase 3

FMS-like tyrosine kinase 3 (FLT3)—a receptor tyrosine kinase—is a140–160-kDa type I transmembrane protein [188]. FLT3 promotes the proliferation and differentiation of hematopoietic cells by binding to its ligand [189,190]. FLT3 is overexpressed in AML, and its mutations have been detected in approximately 30% of patients with AML [191,192]. The mutation of FLT3 causes ligand-independent FLT3 signaling and leads to a poor prognosis of patients with AML [193,194]. FLT3 is a potential target for antibody therapy against AML. LY3012218 (IMC-EB10) is a human mAb that targets FLT3, which prevents FLT3 signaling [195]. In preclinical studies, LY3012218 has shown efficacy in a mouse xenograft model implanted with MOLM-14 human AML cells; LY3012218 exerts its effects by reducing the engraftment of leukemic cells and extending survival [196].

3.6. Multi-Pass Transmembrane Proteins as ICE Targets in Hematological Cancers

3.6.1. CD20

CD20 is a 33–37-kDa tetra-transmembrane protein [197]. It is involved in B-cell differentiation and it is overexpressed in most B-cell malignancies, such as follicular lymphoma (FL), but not in hematopoietic stem cells or plasma cells [198,199,200]. Several studies have shown that CD20 is a potential target for antibody therapy against FL. Rituximab (Rituxan) is a chimeric mAb that has been approved by the US FDA against CD20 [155]. In a phase III clinical study, patients with R/R FL received lenalidomide plus rituximab or placebo plus rituximab; the median PFS increased in the lenalidomide-plus-rituximab group (39.4 months), compared with that in the placebo-plus-rituximab group (14.1 months) [201].

3.6.2. GPCR Class C Group 5 Member D

GPCR class C group 5 member D (GPRC5D) is a 39-kDa seven-transmembrane protein [202]. It is an orphan receptor that is normally expressed only in the hair follicle. GPRC5D is overexpressed in MM and is unlikely to be shed from the membrane, which prevents the decrease in the efficacy of GPRC5D-targeted therapy [203,204,205]. The role of GPRC5D in cancers is yet to be defined; nonetheless, selective GPRC5D expression may be valuable as a target for antibody therapy against MM. Figure 2 summarizes the known and emerging targets for ICE therapy against cancers.

4. Design and Structure of bsAb-Based ICEs

BsAb-based ICEs are designed to contain two different antigen-binding sites that comprise determinants from the VH and VL chains of different antibodies that are specific to each target [206]. Thus far, various efforts have been made to increase their homogeneity, yield, and functional properties to generate desired bsAb-based ICEs [11]. On the basis of the bsAb-based ICE structures, they are divided into two categories: Fv-based ICEs and immunoglobulin G (IgG)-based ICEs (Figure 3). Fv-based ICEs are easy to produce and show lower immunogenicity, whereas IgG-based ICEs have higher solubility, stability, affinity, and extended half-life in serum [207].

4.1. Fv-Based ICEs

4.1.1. BiTE

BiTE comprises two single-chain Fvs (scFvs) combined with a flexible linker [29,208]. Blinatumomab (Blincyto) was the first US FDA-approved BiTE against CD19 and CD3 for the treatment of patients with R/R B-cell ALL [209]. It has a molecular weight of approximately 55 kDa and it is constructed by linking anti-CD19 scFv in a VL–VH orientation to anti-CD3 scFv in a VH–VL orientation through a short polypeptide (G4S) linker [29,208]. Furthermore, each scFv has two flexible long (G4S)3 linkers between the VH and VL to maintain the proper conformation of scFvs [210].

4.1.2. Dual-Affinity Retargeting Protein

Dual-affinity retargeting protein (DART) comprises two engineered heterogenous scFvs, which exchanged their VH regions, and it has a molecular weight of approximately 50 kDa [211]. Precisely, scFv1 comprises a VH from antibody A and a VL from antibody B, and scFv2 comprises a VH from antibody B and a VL from antibody A in the VL(B)–VH(A) and VL(A)–VH(B) orders, respectively [212]. This combination allows DART to mimic natural interactions within IgG molecules. The scFv1 of flotetuzumab—a CD3 × CD123 DART—comprises VL(CD123)—linker—VH(CD3), whereas scFv2 comprises VL(CD3)–linker–VH(CD123) [213]. The linker between VH and VL of the DART platform is as short as approximately five amino acids to prevent their association from forming an undesired scFv. Moreover, the C-terminal disulfide bridge between two VHs contributes to holding the molecule together in the correct orientation [214]. CD19 × CD3 DART molecules have been reported to be more stable and potent than CD19 × CD3 BiTE molecules in targeting and killing B-cell lymphoma cells [210,212,214].

4.1.3. Tandem Diabody

Tandem diabody (TandAb) is a novel tetravalent bsAb-based ICE with four binding sites: two for tumor antigens and the other two for immune cells [215]. It has a molecular weight of approximately 105 kDa. The configuration of TandAb is as follows: VH(A)–linker 1–VL(B)–linker 2–VH(B)–linker 3–VL(A) [216]. In this configuration, both linkers 1 and 3 comprise six amino acids (GGSGGS); however, linker 2 has one of the following three peptide sequences: GGSG, GGSGG, or GGSGGS [213]. AFM13—a CD16- and CD30-specific NK-cell engager—is currently being evaluated in a phase II clinical study for use in the treatment of patients with CD30-positive T-cell lymphoma [217].

4.2. IgG-Based ICEs: Symmetric Format

IgG-based ICEs are classified into symmetric and asymmetric architecture. Symmetric formats are mainly tetravalent (2 + 2) and are constructed by Fv or Fab fused with an IgG molecule [11].

4.2.1. Fabs-In-Tandem Ig

Fabs-In-Tandem Ig (FIT-Ig) is a tetravalent ICE, where Fab(A) is structurally fused in tandem with the N terminus of Fab(B), without any mutations or the use of peptide linkers [218]. It has a molecular weight of approximately 240 kDa. More specifically, it comprises one long chain and two short chains: the long chain where the light-chain (VL(A)–CL) domains are directly fused in tandem with the N terminus of a heavy chain (VH(B)–CH1–CH2–CH3), and two short chains (VH(A)–CH1 and VL(B)–CL) [218,219]. The resulting FIT-Ig may have activities that are similar to those of both parental mAbs [218]. EMB-06a—a CD3 × BCMA FIT-Ig—combines intact Fab fragments from two parental antibodies, and it exhibits favorable drug-like properties and manufacturing advantages that are similar to those of each parental mAb [218,220].

4.2.2. IgG–[L]–scFv2

IgG–[L]–scFv2 is a tetravalent ICE that is constructed by fusing the scFv with the C terminus of each IgG light chain [221]. This antibody is constructed by linking the human CD3-specific scFv on T-cells to the C terminus of the light chain of each antitumor IgG via a polypeptide linker [222,223]. Nivatrotamab is a 200-kDa (nearly) type of IgG–[L]–scFv2 antibody [224]. The heavy chain of nivatrotamab is identical to that of an anti-GD2 IgG, whereas its light chain is constructed by linking an anti-GD2 IgG light chain to a (G4S)3 linker, followed by the CD3-specific scFv [222,223,224]. Previous studies have indicated IgG–[L]–scFv2 as a promising platform for robust antitumor activity [221].

4.2.3. IgG–[H]–scFv2

IgG–[H]–scFv2 is a tetravalent ICE that is constructed by fusing the scFv with the C terminus of each IgG heavy chain. In this format, CD3-specific scFvs are covalently attached to the C terminus of each TAA-specific IgG heavy chain [210]. CC-1 (CD3 × PSMA) and CLN-049 (CD3 × FLT3) are the major ICEs of IgG–[H]–scFv2, with a molecular weight of approximately 200 kDa [225]. To construct the CC-1 ICE, anti-CD3 scFv is linked to an anti-PSMA IgG by a flexible (G4S)3 linker [226]. CC-1 ICE has been reported as a highly potent ICE with significant productivity and low aggregation [227].

4.2.4. scFv2–Fc–scFv2

scFv2–Fc–scFv2, including Aptevo’s ADAPTIR antibodies, is a type of tetravalent ICE that comprises two scFv pairs that are joined via the Fc region [226]. Antitumor scFvs are attached to the N terminus of the hinged domain, whereas anti-CD3 scFvs are fused at the C terminus of the Fc region with an unknown linker. The length and composition of the ADAPTIR linker vary to modulate the binding and activity [228]. APVO436 (CD3 × CD123), which is generated by using the ADAPTIR platform as a T-cell engager, is currently being evaluated in clinical studies [229].

4.3. IgG-Based ICEs: Asymmetric Format

The major challenge in the generation of IgG-based ICEs with asymmetric formats is ensuring the correct association between the heavy and light chains [11]. Thus far, various conventional and state-of-the-art technologies, including mouse–rat hybrid, knob-into-hole (KiH), charge pair, controlled Fab-arm exchange (cFAE), common light chain, and CrossMab technology, have been used to develop desirable IgG-based ICEs to avoid random associations between the heavy and light chains [230,231,232].

4.3.1. Mouse–Rat Hybrid IgG

Mouse–rat hybrid IgG is a bispecific chimeric antibody that is generated by using the quadroma technology that is based on the fusion of two distinct hybridomas [233]. This antibody is also called the trifunctional antibody or Triomab, owing to the retained effector function of the mouse/rat Fc part [234]. It comprises two different full-size IgG-like half antibodies, each with one light and one heavy chain, which originate from the parental-mouse-IgG2a and rat-IgG2b isotypes [235]. The use of the isotype combination enables the high yield of correctly paired bsAbs because of species-restricted heavy–light chain pairing [236]. By using this technology, catumaxomab—an EpCAM- and CD3-specific ICE—was developed and approved as the first bispecific ICE in 2009 by the EMA for the treatment of patients with malignant ascites [235]. However, catumaxomab was reported to have several issues that are related to the highly immunogenic nature of the mouse–rat hybrid antibody and large-scale commercial manufacturing. Catumaxomab was voluntarily withdrawn from the US and EU markets in 2013 and 2017, respectively [237].

4.3.2. KiH-Based IgG

The KiH technology invented by Genentech is the most widely used bispecific platform for the generation of asymmetric bsAbs [238]. The concept relies on interface modifications between the two CH3 domains that are crucial for reciprocal interactions. A bulky residue is introduced as a key into the CH3 domain of one antibody heavy chain, and a hole that accommodates this bulky residue acts as a lock in the other heavy chain. Precisely, a knob is designed by replacing T366 with a bulky W residue on one heavy chain, and the three amino acid residues on the partner heavy chain are changed into T366S, L368A, and Y407V for the hole formation [238,239]. This technology produces >90% of the desired asymmetric bsAbs under coexpression conditions [240]. More recently, the yield of the heterodimeric bsAb has been increased to >97% by the introduction of two additional mutations: S354C in the knob chain, and Y349C in the hole chain [241]. Furthermore, these KiH mutations are known to not significantly affect the antibody properties, such as the immunogenicity, thermal stability, FcɤR binding, Fc effector function, and pharmacokinetic behavior [235].

4.3.3. Charge-Pair-Based IgG

The charge-pair-based technology is a novel platform to generate electrostatically matched Fc domains by altering the charge polarity between the CH3 interfaces of an IgG antibody [242]. Previous studies have reported that the point mutations of three specific charged residue pairs in the heavy chains (E356K-K439E, E357K-K370E, and D399K-K409D) resulted in favorable attractive interactions [235]. This method uses the asymmetric re-engineering technology Ig (ART-Ig) platform [242]. ERY974, which is a CD3- and GPC3-specific ICE based on the ART-Ig platform, comprises E365K in one anti-CD3 heavy chain, and K439E in the other anti-GPC3 heavy chain [243]. However, the charge-pair approach may not result in the higher yield and purity of the respective heterodimeric antibodies than the KiH approach [244].

4.3.4. cFAE-Based IgG

cFAE technology is inspired by a natural immune phenomenon that is known as Fab-arm exchange, in which a half molecule of an IgG4 antibody is reassembled with that of another IgG4 antibody in vivo [232,245]. It is used to generate IgG-like bsAbs. One pair of these matching CH3 mutations (F405L in one antibody and K409R in the other) has been reported through extensive site-directed mutagenesis in IgG1 [246]. The cFAE platform does not need additional technology to correct the light-chain assembly because light-chain mispairing does not occur in this case [241]. Epcoritamab—a CD3 × CD20 ICE—is generated by individually introducing an F405L mutation into an anti-CD3 heavy chain, and a K409R mutation into a CD20-specific heavy chain [247]. This technology includes the DuoBody platform that was developed by Genmab. Recently, amivantamab—a bispecific molecule that targets EGFR and mesenchymal–epithelial transition factor (c-MET)—which was produced by Johnson & Johnson by using the aforementioned platform, was approved by the US FDA for the treatment of patients with locally advanced or metastatic non-SCLC [248].

4.3.5. Common Light-Chain-Based IgG

Common light-chain technology is developed to overcome the mispairing of light chains and is used in combination with an Fc-modified technology, such as KiH and charge-pair-based technologies [249]. This technology is based on the findings that antibodies against various antigens often share the same VL domain, in which antibodies were identified from phage display libraries with a very limited size of the light-chain repertoire [210]. bsAbs that are designed by using the common light-chain technology have identical light chains that can be paired with two different heavy chains. This antibody can prevent the mispairing between light and heavy chains [238]. By using this technology, REGN1979—a CD20- and CD3-specific ICE—was developed and is currently being evaluated in a phase II clinical study for use in the treatment of patients with B-cell NHL [250]. The common light-chain technology is based on simplifying antibody engineering, but this approach may cause less flexibility for antibody engineering [210].

4.3.6. CrossMab-Based IgG

CrossMab technology is developed to inhibit the mispairing of light chains [251]. However, it takes a different approach from that of the common light-chain technology. This technology is based on the crossover of the constant region (CL and CH1 domains) of one Fab arm, whereas the other Fab arm undergoes no change [241]. This approach employs the KiH technology for correct heavy-chain pairing, as well as domain swap to enable orthogonal light–heavy chain pairing [207]. Glofitamab—a CD20- and CD3-specific ICE—was developed by using this technology and is currently being evaluated in a phase III clinical study for use in the treatment of patients with diffuse large B-cell lymphoma (DLBCL) [252].

4.3.7. Fab–scFv–Fc

The Fab–scFv–Fc format is another strategy to correct the pairing of the light chain. The issue of light-chain mispairing is resolved by exchanging one Fab arm with scFv [253]. Fab–scFv–Fc comprises one Fab arm and one scFv that is fused with an Fc domain [254]. This antibody is used by several companies and it has various names, such as Xmab, Ybody, and BEAT [254,255]. Tidutamab (XmAb18087)—a CD3- and SSTR2-specific ICE—is currently being evaluated in a phase I/II clinical study for use in the treatment of patients with SCLC [256]. Additionally, six different ICEs that have the Fab–scFv–Fc format are currently being evaluated in clinical studies [257,258].

4.3.8. Fc-Fused Fvs

The small size of Fv-based ICEs contributes to high renal clearance, which results in a half-life shorter than that noted in the case of IgG-based ICEs. BiTE and DART ICEs have recently been engineered to improve their pharmacokinetics (PK) [210]. Several BiTEs are linked to the Fc domain to generate half-life-extended BiTE-Fc molecules (HLE-BiTE), which are compatible with once-weekly dosing for treatments. DART is fused with the Fc region, which generates a DART–Fc complex. MGD007 (CD3 × GPA33) was constructed as a DART–Fc complex that results in a significant extension of the half-life in serum [259].

5. Current Development Status of bsAb-Based ICEs

5.1. ICEs Targeting Tumor Antigens in Solid Cancers

5.1.1. CEA-Specific ICE

RO6958688 is a CD3- and CEA-specific bsAb-based T-cell engager that is used for the treatment of patients with advanced CEA-positive solid tumors. In phase I clinical studies (NCT02324257 and NCT02650713), RO6958688 has been administered as monotherapy or in combination with atezolizumab—a US FDA-approved anti-PD-ligand 1 mAb—in patients with advanced CEA-positive solid tumors. Two patients showed a PR after monotherapy, and two after its use in combination with atezolizumab. Antitumor activity was observed with RO6958688 monotherapy, which was enhanced when this engager was used in combination with atezolizumab, with a manageable safety profile [260].

5.1.2. CLDN18.2-Specific ICE

AMG 910 is a bsAb-based T-cell engager against CD3 and CLDN18.2 that is used for the treatment of patients with gastric or GEJ adenocarcinoma [261]. A phase I clinical study (NCT04260191) on AMG 910 is currently being conducted to evaluate its dose-limiting toxicity, treatment-related adverse event, PK, response duration, time to progression, and recommended phase 2 dose (RP2D) [261,262].

5.1.3. DLL3-Specific ICE

Tarlatamab (AMG 757) is a CD3- and DLL3-specific bsAb-based T-cell engager that is used for the treatment of patients with DLL3-expressing SCLC. In a phase I clinical study (NCT03319940) on tarlatamab, a confirmed PR was reported across all dose levels in 8 out of 60 patients (13%), and an unconfirmed PR in 5 out of 8 patients (63%), at the highest dose level [263]. A phase II clinical study (NCT05060016) is currently ongoing to evaluate the safety and efficacy of tarlatamab [264].

5.1.4. EGFR-Specific ICE

REGN7075 is a CD28- and EGFR-specific bsAb-based T-cell engager that is used for the treatment of patients with advanced solid cancers. A phase I/II clinical trial (NCT04626635) is currently being conducted to assess the safety and tolerability of REGN7075 in combination with cemiplimab—a US FDA-approved antihuman programmed cell death receptor-1 (PD-1) mAb—in patients with advanced solid tumors [265].

5.1.5. EpCAM-Specific ICE

Catumaxomab (Removab) is the first bsAb-based T-cell engager that was approved by the EMA against CD3 and EpCAM for use in the treatment of malignant ascites in patients with EpCAM-positive cancer [14]. In a phase II/III clinical study (NCT00836654), the puncture-free survival was significantly longer in the catumaxomab group (median: 46 days) than in the control group (median: 11 days) [266]. M701 is another type of CD3- and EpCAM-specific bsAb-based T-cell engager that is used for the treatment of malignant ascites that are caused by advanced solid tumors. The interim results of a phase I clinical study (NCT04501744) on M701 revealed promising efficacy results: of 16 patients treated with M701, 3 showed a CR, 7 showed a PR, and 6 had SD [267].

5.1.6. GD2-Specific ICE

Nivatrotamab is a CD3- and GD2-specific bsAb-based T-cell engager that is used for the treatment of patients with metastatic SCLC [268]. A phase I/II clinical study (NCT04750239) is currently being conducted to evaluate the safety and tolerability of nivatrotamab in patients with metastatic SCLC [269].

5.1.7. GPA33-Specific ICE

MGD007 is a bsAb-based T-cell engager against CD3 and GPA33 that is based on the DART platform and that is used for the treatment of patients with metastatic CRCs [270]. The safety, tolerability, and efficacy of MGD007 in combination with retifanlimab (previously known as MGA012)—an investigational anti-PD-1 humanized mAb—were assessed in a phase I/II clinical study (NCT03531632) [271].

5.1.8. GPC3-Specific ICE

ERY974 is a CD3- and GPC3-specific bsAb-based T-cell engager that is used for the treatment of patients with advanced or metastatic HCCs [272]. A phase I clinical study (NCT05022927) is currently being conducted to evaluate the safety, tolerability, PK, and antitumor activity of ERY974 in combination with atezolizumab and bevacizumab—a US FDA-approved anti-vascular endothelial growth factor mAb—in patients with locally advanced or metastatic HCC [273].

5.1.9. HER2-Specific ICE

M802 is a bsAb-based T-cell engager against CD3 and HER2 that is used for the treatment of patients with HER2-positive advanced solid cancers [255,274]. M802 has been approved for phase I clinical study (NCT04501770) to evaluate its safety and tolerability in patients with HER2-positive advanced solid cancers [274].

5.1.10. MUC16-Specific ICE

Ubamatamab (REGN4018) is a bsAb-based T-cell engager against CD3 and MUC16 that is used for the treatment of patients with advanced ovarian cancer. A phase I/II clinical study (NCT03564340) is currently being conducted to assess the PK, safety, tolerability, and preliminary antitumor activity of ubamatamab monotherapy and its combined use with cemiplimab, which is a US FDA-approved anti-PD-1 mAb [275].

5.1.11. MUC17-Specific ICE

AMG 199 is an HLE-BiTE molecule against CD3 and MUC17. It induces the activation and proliferation of CD3+ T-cells and activates T-cell-mediated tumor-cell lysis in MUC17-positive gastric cancer [81]. A phase I clinical study (NCT04117958) on AMG 199 is currently being conducted to evaluate the safety, tolerability, PK, and antitumor activity of AMG 199 in MUC17-positive solid tumors, and to determine the maximum tolerated dose and/or RP2D [276].

5.1.12. PSMA-Specific ICE

Acapatamab (AMG 160) is a CD3- and PSMA-specific bsAb-based T-cell engager that is used for the treatment of patients with mCRPC. Acapatamab induces the infiltration, activation, and expansion of T-cells, as well as the killing of tumor cells [277]. A phase I clinical study (NCT03792841) is currently being conducted to evaluate the safety and tolerability of acapatamab monotherapy or its combined use with pembrolizumab (Keytruda)—an anti-PD-1 mAb—in patients with mCRPC [278]. Additionally, other anti-PSMA bsAb-based ICEs, such as CC-1, JNJ-63898081, TNB-585, REGN5678, and MOR209/ES414, are currently being evaluated in clinical studies conducted on patients with mCRPC [226,279].

5.1.13. STEAP1-Specific ICE

AMG 509 is a CD3- and STEAP1-specific bsAb-based T-cell engager that is used for the treatment of patients with STEAP1-expressing prostate cancers. A phase I clinical study (NCT04221542) on AMG 509 is currently being conducted to evaluate its safety, tolerability, PK, and efficacy in patients with mCRPC [280].

5.1.14. SSTR2-Specific ICE

Tidutamab (XmAb18087) is a bsAb-based T-cell engager against CD3 and SSTR2 for the treatment of patients with advanced NETs [281]. A phase I/II clinical study (NCT04590781) is currently being conducted to evaluate the safety, tolerability, PK, and potential efficacy of tidutamab in patients with extensive-stage SCLC [256].

5.2. ICEs Targeting Tumor Antigens on Hematological Cancers

5.2.1. BCMA-Specific ICE

Teclistamab (JNJ-64007957) is a CD3- and BCMA-specific bsAb-based T-cell engager based on the DuoBody platform [282]. In a phase I clinical study (NCT03145181), patients with R/R MM received teclistamab monotherapy at the RP2D. Of the 40 patients, 58% showed a very good PR (VGPR) or better outcome, and 30% showed a CR [283]. A phase III clinical study (NCT05083169) aimed at evaluating the efficacy of teclistamab in combination with daratumumab, which is a US FDA-approved anti-CD38 mAb, in the treatment of patients with R/R MM is ongoing [284]. Other CD31- and BCMA-specific bsAb-based T-cell engagers, such as elranatamab (PF-06863135), linvoseltamab (REGN5458), REGN5459, EMB-06, CC-93269 (EM801), TNB-383B, and AMG 701, are being actively assessed in various clinical studies [251,285].

5.2.2. CD19-Specific ICE

Blinatumomab (Blincyto) is a CD3- and CD19-specific bsAb-based T-cell engager that was approved by the US FDA on the basis of the BiTE platform [286]. In a phase II/III clinical study (NCT02910063), patients with R/R aggressive B-cell NHL received blinatumomab as the second salvage therapy. Of the 41 patients, 9 showed complete metabolic responses, and 6 showed partial metabolic responses [287,288]. Then, in a phase II clinical study (NCT03023878), patients with high-risk DLBCL who received first-line treatment with rituximab plus chemotherapy were treated with blinatumomab. Of them, 7 of 8 patients with PR and SD after treatment with rituximab plus chemotherapy showed CR following treatment with one cycle of blinatumomab [289]. In addition, TNB-486—another anti-CD19 bsAb-based T-cell engager—is currently being evaluated in a phase I clinical study (NCT04594642) for its safety and clinical activity in patients with R/R B-cell NHL [290].

5.2.3. CD20-Specific ICE

Mosunetuzumab (RG7828) is a CD3- and CD20-specific bsAb-based T-cell engager that is based on the CrossMab platform [291]. In a phase I/II clinical trial (NCT02500407), 90 patients with R/R FL who received ≥2 prior lines of therapy were treated with mosunetuzumab monotherapy. Of them, 57.8% showed a CR. The 12-month event-free rates were 80.1% in CR patients [292]. A phase III clinical study (NCT04712097) is currently being conducted to evaluate the safety and efficacy of mosunetuzumab in combination with lenalidomide in patients with R/R FL [269]. Additionally, other CD3- and CD20-specific bsAb-based T-cell engagers, such as glofitamab (RG6026 and RO7082859), epcoritamab (GEN3013), odronextamab (REGN1979), and plamotamab (XmAb13676), are being actively assessed in various clinical studies [293,294].

5.2.4. CD22-Specific ICE

JNJ-75348780 is a CD3- and CD22-specific bsAb-based T-cell engager. It has been evaluated in a phase I clinical study (NCT04540796) to assess its safety and RP2D in patients with R/R B-cell NHL and CLL [295].

5.2.5. CD30-Specific ICE

AFM13 is a bsAb-based NK-cell engager against CD16A and CD30 [296]. In a phase I/II clinical study (NCT04074746), patients with R/R HL and NHL were treated with precomplexed cord-blood NK cells with AFM13. Of the 13 patients, 8 showed a CR and 5 showed a PR after two cycles of treatment. Of 8 patients who showed a CR, 7 remained in CR at the median follow up of 6.5 months [297]. A phase II clinical study (NCT04101331) is currently being conducted to evaluate AFM13 monotherapy in patients with R/R peripheral T-cell lymphoma or transformed mycosis fungoides that are classified as a subtype of NHL [217].

5.2.6. CD33-Specific ICE

AMG 330 is a CD3- and CD33-specific bsAb-based T-cell engager. A phase I clinical study (NCT02520427) enrolled 40 patients with R/R AML to receive AMG 330 monotherapy. Of them, two showed a CR, two showed a CR with incomplete hematologic recovery (CRi), and one achieved the morphologic leukemia-free stage (MLFS) [298]. In addition, JNJ-67571244—another CD3- and CD33-specific bsAb-based T-cell engager—has been included in a phase I clinical study (NCT03915379) to evaluate its safety and clinical activity in patients with R/R AML, as well as those with high-risk or very-high-risk myelodysplastic syndrome [299].

5.2.7. CD38-Specific ICE

ISB 1342 (GBR 1342) is a CD3- and CD38-specific bsAb-based T-cell engager. A phase I clinical study (NCT03309111) is currently being conducted to evaluate the safety and efficacy of ISB 1342 in patients with R/R MM [300].

5.2.8. CD123-Specific ICE

Flotetuzumab (MGD006) is a bsAb-based T-cell engager against CD3 and CD123 that is based on the DART platform [301]. In a phase I/II clinical study (NCT02152956), patients with primary induction failure (PIF) or early relapsed (ER) AML received flotetuzumab monotherapy. Among the patients with PIF/ER AML who were treated by using the RP2D, 31.8% showed a CR, CRi, and CR with partial hematologic recovery [302]. Flotetuzumab is in phase II clinical trial (NCT04582864) to evaluate its safety and efficacy in patients with relapsed AML following allogeneic hematopoietic-stem-cell transplantation [303]. Additionally, other CD3- and CD123-specific bsAb-based T-cell engagers, such as JNJ-63709178, vibecotamab (XmAb14045), and APVO436, are also being evaluated in clinical studies for their use in the treatment of CD123-expressing hematological malignancies [304].

5.2.9. CLEC12A-Specific ICE

Tepoditamab (MCLA-117) is a bsAb-based T-cell engager against CD3 and CLEC12A [305]. A phase I clinical study (NCT03038230) enrolled 58 patients with AML to receive tepoditamab monotherapy; the result showed that >50% of bone-marrow blast reduction was observed in six patients, including one patient who achieved the MLFS [306].

5.2.10. FLT3-Specific ICE

CLN-049 is a CD3- and FLT3-specific bsAb-based T-cell engager [307]. In preclinical studies, CLN-049 showed dose-dependent tumor reduction in the blood, and prolonged survival in a mouse xenograft model implanted with MOLM-13 human AML cells [308]. CLN-049 is currently being evaluated in a phase I clinical study (NCT05143996) for use in the treatment of patients with R/R AML [309]. In addition, AMG 427—another CD3- and FLT3-specific bsAb-based T-cell engager—is also in a phase I clinical study (NCT03541369) for use in the treatment of patients with R/R AML [310].

5.2.11. GPRC5D-Specific ICE

Talquetamab (JNJ-64407564) is a bsAb-based T-cell engager against CD3 and GPRC5D that is based on the DuoBody platform. In a phase I clinical study (NCT03399799), the patients with R/R MM received talquetamab monotherapy at the RP2D, and 50% of the patients showed a VGPR or better outcome [311]. Talquetamab is currently being evaluated in a phase II clinical study (NCT04634552) for its safety and efficacy in patients with R/R MM [312].

6. Practical Considerations for the Development of Next-Generation ICEs

On the basis of the currently available evidence, this study proposes practical considerations for the development of the next-generation ICEs. First, the proximity of immune and tumor cells appears to underlie the improvement in the anticancer effects that are exerted by ICEs. A previous study compared (in vitro and in vivo) the efficacy of two different CD3 × GD2 ICEs that were designed by using the IgG–[L]–scFv2 or IgG–[H]–scFv2 format. The results show that IgG–[L]–scFv2 has an antitumor effect that is superior to that of IgG–[H]–scFv2 [221]. Second, another challenge in the ICE generation is to reinforce the PK of ICEs. Recently, several Fv-based ICEs, such as BiTE and DART, extend the in vivo half-life of antibodies. Thus, most bsAb-based ICEs are currently developed in the IgG-like or Fc-fused form [259,313]. Third, a high affinity of ICE for CD3 may not be necessary for achieving optimal T-cell activation without causing systemic toxicity. The systemic administration of anti-CD3 antibodies or catumaxomab (CD3 × EpCAM) often leads to systemic toxicity that is due to the uncontrolled T-cell activation in the peripheral blood [314]. Anti-CD3 antibodies of ICEs reportedly have various affinities that range from 1 to 200 nM [315]. Furthermore, a recent study reports that low-affinity anti-CD3 antibody shows better tumor distribution in vivo, without targeting T-cell-containing normal tissues, such as the spleen and lymph nodes [316]. Fourth, ICEs with silenced Fc are required for diminishing effector functions, such as ADCC, antibody-dependent T-cellular phagocytosis, and complement-dependent cytotoxicity. The binding of Fc to Fcγ receptors on immune cells may induce nonspecific immune activation that is associated with undesired toxicity [317]. Various ICEs with silenced Fc are currently being developed. For instance, the Fc domain of some ICEs was silenced by introducing various mutations: L234A-L235A-P329G (EM801), L234A-L235A-G237A-K322A (APVO436), E233P-L234V-L235A-G236del-S267K (Vibecotamab), N297G (AMG 330), or L234F-L235E-D265A (GEN3013) [11,247,318,319]. Furthermore, ICEs with silenced Fc improve the immune cells’ infiltration into solid tumors and enhance antitumor effects. Fifth, the optimal amino acid composition and ICE linkers’ length are the crucial factors for ensuring favorable physicochemical properties that are closely associated with manufacturing efficiencies [210]. Finally, despite recent advances in the identification of therapeutic targets of ICEs in cancer, cancers are highly heterogeneous diseases that harbor frequent mutations that are resistant to pre-existing drugs. Thus, the identification of novel potential targets for ICE therapy against cancers remains crucial for improving the clinical outcomes of patients with cancers.

7. Conclusions

Over several decades, cancer therapies have continuously evolved and have substantially improved the quality of life and the survival of patients with cancers. In particular, the advent of cancer immunotherapy has revolutionized the treatment paradigms in various cancers. Recently, bsAb-based ICEs have been established as a feasible modality for effective cancer immunotherapy and, consequently, have become the focus of several preclinical and clinical studies that are conducted globally to evaluate the efficacy and/or toxicity of bsAb-based ICEs to known and/or emerging therapeutic targets. There are still multiple challenges that are associated with the use of bsAb-based ICEs, such as drug resistance, manufacturing difficulties, and adverse effects on adjacent normal cells; nevertheless, these ICEs have potential in cancer treatments, as they are single-molecule anticancer agents that combine the advantages of two different mAbs that specifically target tumor-specific antigens or TAAs. Continuous research and development may help overcome the current pitfalls of these treatments, which may create new avenues for the successful treatment of patients with various cancers in the near future.

Author Contributions

H.G.S., H.R.Y., A.Y. and S.L. collected and analyzed data, discussed and commented on the manuscript, and wrote the manuscript. S.L. supervised the project. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by research grants from the Bio & Medical Technology Development Program of the National Research Foundation, funded by the Korean government (NRF-2019M3E5D5065844 and NRF-2020M3A9I2107093).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data collected in this study are available from the corresponding author upon reasonable request.

Acknowledgments

The authors acknowledge the contribution of the researchers whose experimental work has been cited in this article. We also acknowledge that the images were created by using BioRender (biorender.com accessed on 20 April 2022).

Conflicts of Interest

The authors declare no conflict of interest. The funders have no role in the design of the study; in the collection, analysis, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Estimated Number of New Cases in 2020, Worldwide, Both Sexes, All Ages. Available online: https://gco.iarc.fr/today/online-analysispie?v=2020&mode=cancer&mode_population=continents&population=900&populations=900&key=total&sex=0&cancer=39&type=0&statistic=5&prevalence=0&population_group=0&ages_group%5B%5D=0&ages_group%5B%5D=17&nb_items=7&group_cancer=1&include_nmsc=1&include_nmsc_other=1&half_pie=0&donut=0 (accessed on 21 February 2022).
  2. Estimated Number of Deaths in 2020, Worldwide, Both Sexes, All Ages. Available online: https://gco.iarc.fr/today/online-analysispie?v=2020&mode=cancer&mode_population=continents&population=900&populations=900&key=total&sex=0&cancer=39&type=1&statistic=5&prevalence=0&population_group=0&ages_group%5B%5D=0&ages_group%5B%5D=17&nb_items=7&group_cancer=1&include_nmsc=1&include_nmsc_other=1&half_pie=0&donut=0 (accessed on 21 February 2022).
  3. Estimated Number of New Cases From 2020 to 2040, Both Sexes, Ages (0–85). Available online: https://gco.iarc.fr/tomorrow/en/dataviz/bars?types=0&sexes=0&mode=population&group_populations=1&multiple_populations=1&multiple_cancers=1&cancers=39&populations=903_904_905_908_909_935&apc=cat_ca20v1.5_ca23v1.5&group_cancers=1 (accessed on 21 February 2022).
  4. Arruebo, M.; Vilaboa, N.; Sáez-Gutierrez, B.; Lambea, J.; Tres, A.; Valladares, M.; González-Fernández, A. Assessment of the evolution of cancer treatment therapies. Cancers 2011, 3, 3279–3330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Basak, D.; Arrighi, S.; Darwiche, Y.; Deb, S. Comparison of Anticancer Drug Toxicities: Paradigm Shift in Adverse Effect Profile. Life 2021, 12, 48. [Google Scholar] [CrossRef] [PubMed]
  6. Baldo, B.A. Immune- and Non-Immune-Mediated Adverse Effects of Monoclonal Antibody Therapy: A Survey of 110 Approved Antibodies. Antibodies 2022, 11, 17. [Google Scholar] [CrossRef] [PubMed]
  7. Janeway, C.A., Jr.; Travers, P.; Walport, M.; Shlomchik, M.J. The structure of a typical antibody molecule. In Immunobiology: The Immune System in Health and Disease, 5th ed.; Garland Science: New York, NY, USA, 2001. [Google Scholar]
  8. Vidarsson, G.; Dekkers, G.; Rispens, T. IgG Subclasses and Allotypes: From Structure to Effector Functions. Front. Immunol. 2014, 5, 520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Redman, J.M.; Hill, E.M.; AlDeghaither, D.; Weiner, L.M. Mechanisms of action of therapeutic antibodies for cancer. Mol. Immunol. 2015, 67, 28–45. [Google Scholar] [CrossRef] [Green Version]
  10. Van Erp, E.A.; Luytjes, W.; Ferwerda, G.; van Kasteren, P.B. Fc-Mediated Antibody Effector Functions During Respiratory Syncytial Virus Infection and Disease. Front. Immunol. 2019, 10, 548. [Google Scholar] [CrossRef] [Green Version]
  11. Labrijn, A.F.; Janmaat, M.L.; Reichert, J.M.; Parren, P. Bispecific antibodies: A mechanistic review of the pipeline. Nat. Rev. Drug Discov. 2019, 18, 585–608. [Google Scholar] [CrossRef]
  12. Wang, S.; Chen, K.; Lei, Q.; Ma, P.; Yuan, A.Q.; Zhao, Y.; Jiang, Y.; Fang, H.; Xing, S.; Fang, Y. The state of the art of bispecific antibodies for treating human malignancies. EMBO Mol. Med. 2021, 13, 14291. [Google Scholar] [CrossRef]
  13. Mazor, Y.; Oganesyan, V.; Yang, C.; Hansen, A.; Wang, J.; Liu, H.; Sachsenmeier, K.; Carlson, M.; Gadre, D.V.; Borrok, M.J.; et al. Improving target cell specificity using a novel monovalent bispecific IgG design. MAbs 2015, 7, 377–389. [Google Scholar] [CrossRef] [Green Version]
  14. Linke, R.; Klein, A.; Seimetz, D. Catumaxomab: Clinical development and future directions. mAbs 2010, 2, 129–136. [Google Scholar] [CrossRef] [Green Version]
  15. Ruf, P.; Bauer, H.W.; Schoberth, A.; Kellermann, C.; Lindhofer, H. First time intravesically administered trifunctional antibody catumaxomab in patients with recurrent non-muscle invasive bladder cancer indicates high tolerability and local immunological activity. Cancer Immunol. Immunother. 2021, 70, 2727–2735. [Google Scholar] [CrossRef] [PubMed]
  16. Nagorsen, D.; Kufer, P.; Baeuerle, P.A.; Bargou, R. Blinatumomab: A historical perspective. Pharmacol. Ther. 2012, 136, 334–342. [Google Scholar] [CrossRef] [PubMed]
  17. Garber, K. Bispecific antibodies rise again: Amgen’s blinatumomab is setting the stage for a bispecific-antibody revival, enabled by new formats that may solve the field’s long-standing problems. Nat. Rev. Drug Discov. 2014, 13, 799–802. [Google Scholar] [CrossRef] [PubMed]
  18. Zhou, S.; Liu, M.; Ren, F.; Meng, X.; Yu, J. The landscape of bispecific T cell engager in cancer treatment. Biomark. Res. 2021, 9, 38. [Google Scholar] [CrossRef] [PubMed]
  19. Tian, Z.; Liu, M.; Zhang, Y.; Wang, X. Bispecific T cell engagers: An emerging therapy for management of hematologic malignancies. J. Hematol. Oncol. 2021, 14, 75. [Google Scholar] [CrossRef]
  20. Grakoui, A.; Bromley, S.K.; Sumen, C.; Davis, M.M.; Shaw, A.S.; Allen, P.M.; Dustin, M.L. The immunological synapse: A molecular machine controlling T cell activation. Science 1999, 285, 221–227. [Google Scholar] [CrossRef] [Green Version]
  21. Dustin, M.L. The immunological synapse. Cancer Immunol. Res. 2014, 2, 1023–1033. [Google Scholar] [CrossRef] [Green Version]
  22. Thiery, J.; Keefe, D.; Boulant, S.; Boucrot, E.; Walch, M.; Martinvalet, D.; Goping, I.S.; Bleackley, R.C.; Kirchhausen, T.; Lieberman, J. Perforin pores in the endosomal membrane trigger the release of endocytosed granzyme B into the cytosol of target cells. Nat. Immunol. 2011, 12, 770–777. [Google Scholar] [CrossRef] [Green Version]
  23. Bacac, M.; Fauti, T.; Sam, J.; Colombetti, S.; Weinzierl, T.; Ouaret, D.; Bodmer, W.; Lehmann, S.; Hofer, T.; Hosse, R.J.; et al. A Novel Carcinoembryonic Antigen T-Cell Bispecific Antibody (CEA TCB) for the Treatment of Solid Tumors. Clin. Cancer Res. 2016, 22, 3286–3297. [Google Scholar] [CrossRef] [Green Version]
  24. Dain Moon, N.T.; Park, Y.; Lee, S.-W.; Kim, D.H. Development of bispecific antibody for cancer immunotherapy: Focus on T cell engaging antibody. Immune Netw. 2022, 22, 4. [Google Scholar] [CrossRef]
  25. Kamakura, D.; Asano, R.; Yasunaga, M. T Cell Bispecific Antibodies: An Antibody-Based Delivery System for Inducing Antitumor Immunity. Pharmaceuticals 2021, 14, 1172. [Google Scholar] [CrossRef] [PubMed]
  26. Rudolph, M.G.; Stanfield, R.L.; Wilson, I.A. How TCRs bind MHCs, peptides, and coreceptors. Annu. Rev. Immunol. 2006, 24, 419–466. [Google Scholar] [CrossRef] [PubMed]
  27. Aptsiauri, N.; Ruiz-Cabello, F.; Garrido, F. The transition from HLA-I positive to HLA-I negative primary tumors: The road to escape from T-cell responses. Curr. Opin. Immunol. 2018, 51, 123–132. [Google Scholar] [CrossRef] [PubMed]
  28. Smith-Garvin, J.E.; Koretzky, G.A.; Jordan, M.S. T cell activation. Annu. Rev. Immunol. 2009, 27, 591–619. [Google Scholar] [CrossRef]
  29. Huehls, A.M.; Coupet, T.A.; Sentman, C.L. Bispecific T-cell engagers for cancer immunotherapy. Immunol. Cell Biol. 2015, 93, 290–296. [Google Scholar] [CrossRef] [Green Version]
  30. Offner, S.; Hofmeister, R.; Romaniuk, A.; Kufer, P.; Baeuerle, P.A. Induction of regular cytolytic T cell synapses by bispecific single-chain antibody constructs on MHC class I-negative tumor cells. Mol. Immunol. 2006, 43, 763–771. [Google Scholar] [CrossRef]
  31. Nguyen, H.H.; Kim, T.; Song, S.Y.; Park, S.; Cho, H.H.; Jung, S.H.; Ahn, J.S.; Kim, H.J.; Lee, J.J.; Kim, H.O.; et al. Naive CD8(+) T cell derived tumor-specific cytotoxic effectors as a potential remedy for overcoming TGF-beta immunosuppression in the tumor microenvironment. Sci. Rep. 2016, 6, 28208. [Google Scholar] [CrossRef]
  32. Demaria, O.; Gauthier, L.; Debroas, G.; Vivier, E. Natural killer cell engagers in cancer immunotherapy: Next generation of immuno-oncology treatments. Eur. J. Immunol. 2021, 51, 1934–1942. [Google Scholar] [CrossRef]
  33. Poli, A.; Michel, T.; Thérésine, M.; Andrès, E.; Hentges, F.; Zimmer, J. CD56bright natural killer (NK) cells: An important NK cell subset. Immunology 2009, 126, 458–465. [Google Scholar] [CrossRef]
  34. Amand, M.; Iserentant, G.; Poli, A.; Sleiman, M.; Fievez, V.; Sanchez, I.P.; Sauvageot, N.; Michel, T.; Aouali, N.; Janji, B.; et al. Human CD56dimCD16dim Cells As an Individualized Natural Killer Cell Subset. Front. Immunol. 2017, 8, 699. [Google Scholar] [CrossRef]
  35. Gleason, M.K.; Verneris, M.R.; Todhunter, D.A.; Zhang, B.; McCullar, V.; Zhou, S.X.; Panoskaltsis-Mortari, A.; Weiner, L.M.; Vallera, D.A.; Miller, J.S. Bispecific and trispecific killer cell engagers directly activate human NK cells through CD16 signaling and induce cytotoxicity and cytokine production. Mol. Cancer Ther. 2012, 11, 2674–2684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Bartlett, N.L.; Herrera, A.F.; Domingo-Domenech, E.; Mehta, A.; Forero-Torres, A.; Garcia-Sanz, R.; Armand, P.; Devata, S.; Izquierdo, A.R.; Lossos, I.S.; et al. A phase 1b study of AFM13 in combination with pembrolizumab in patients with relapsed or refractory Hodgkin lymphoma. Blood 2020, 136, 2401–2409. [Google Scholar] [CrossRef] [PubMed]
  37. Owen, D.H.; Giffin, M.J.; Bailis, J.M.; Smit, M.D.; Carbone, D.P.; He, K. DLL3: An emerging target in small cell lung cancer. J. Hematol. Oncol. 2019, 12, 61. [Google Scholar] [CrossRef] [PubMed]
  38. Furuta, M.; Kikuchi, H.; Shoji, T.; Takashima, Y.; Kikuchi, E.; Kikuchi, J.; Kinoshita, I.; Dosaka-Akita, H.; Sakakibara-Konishi, J. DLL 3 regulates the migration and invasion of small cell lung cancer by modulating Snail. Cancer Sci. 2019, 110, 1599–1608. [Google Scholar] [CrossRef] [PubMed]
  39. Furuta, M.; Sakakibara, J.K.; Shoji, T.; Takashima, Y.; Kikuchi, H.; Kikuchi, E.; Kikuchi, J.; Kinoshita, I.; Akita, H.D.; Nishimura, M. Abstract 3158: DLL3 regulates migration and invasion of small cell lung cancer. Cancer Res. 2018, 78, 3158. [Google Scholar] [CrossRef]
  40. Vitorino, P.; Chuang, C.-H.; Iannello, A.; Zhao, X.; Anderson, W.; Ferrando, R.; Zhang, Z.; Madhavan, S.; Karsunky, H.; Saunders, L. Rova-T enhances the anti-tumor activity of anti-PD1 in a murine model of small cell lung cancer with endogenous Dll3 expression. Transl. Oncol. 2021, 14, 100883. [Google Scholar] [CrossRef] [PubMed]
  41. Ribeiro, F.A.P.; Noguti, J.; Oshima, C.T.F.; Ribeiro, D.A. Effective targeting of the epidermal growth factor receptor (EGFR) for treating oral cancer: A promising approach. Anticancer. Res. 2014, 34, 1547–1552. [Google Scholar]
  42. Gschwind, A.; Fischer, O.M.; Ullrich, A. The discovery of receptor tyrosine kinases: Targets for cancer therapy. Nat. Rev. Cancer 2004, 4, 361–370. [Google Scholar] [CrossRef]
  43. Sigismund, S.; Avanzato, D.; Lanzetti, L. Emerging functions of the EGFR in cancer. Mol. Oncol. 2018, 12, 3–20. [Google Scholar] [CrossRef]
  44. Yaish, P.; Gazit, A.; Gilon, C.; Levitzki, A. Blocking of EGF-dependent cell proliferation by EGF receptor kinase inhibitors. Science 1988, 242, 933–935. [Google Scholar] [CrossRef]
  45. Lu, Z.; Jiang, G.; Blume-Jensen, P.; Hunter, T. Epidermal growth factor-induced tumor cell invasion and metastasis initiated by dephosphorylation and downregulation of focal adhesion kinase. Mol. Cell Biol. 2001, 21, 4016–4031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Spano, J.P.; Lagorce, C.; Atlan, D.; Milano, G.; Domont, J.; Benamouzig, R.; Attar, A.; Benichou, J.; Martin, A.; Morere, J.F. Impact of EGFR expression on colorectal cancer patient prognosis and survival. Ann. Oncol. 2005, 16, 102–108. [Google Scholar] [CrossRef] [PubMed]
  47. Richard, J.; Sainsbury, C.; Needham, G.; Farndon, J.; Malcolm, A.; Harris, A. Epidermal-growth-factor receptor status as predictor of early recurrence of and death from breast cancer. Lancet 1987, 329, 1398–1402. [Google Scholar] [CrossRef]
  48. Ekstrand, A.J.; James, C.D.; Cavenee, W.K.; Seliger, B.; Pettersson, R.F.; Collins, V.P. Genes for epidermal growth factor receptor, transforming growth factor α, and epidermal growth factor and their expression in human gliomas in vivo. Cancer Res. 1991, 51, 2164–2172. [Google Scholar] [PubMed]
  49. Hendler, F.J.; Ozanne, B.W. Human squamous cell lung cancers express increased epidermal growth factor receptors. J. Clin. Investig. 1984, 74, 647–651. [Google Scholar] [CrossRef]
  50. Ishitoya, J.; Toriyama, M.; Oguchi, N.; Kitamura, K.; Ohshima, M.; Asano, K.; Yamamoto, T. Gene amplification and overexpression of EGF receptor in squamous cell carcinomas of the head and neck. Brit. J. Cancer 1989, 59, 559–562. [Google Scholar] [CrossRef] [Green Version]
  51. You, B.; Chen, E. Anti-EGFR Monoclonal antibodies for treatment of colorectal cancers: Development of cetuximab and panitumumab. J. Clin. Pharmacol. 2012, 52, 128–155. [Google Scholar] [CrossRef]
  52. Huang, L.; Yang, Y.; Yang, F.; Liu, S.; Zhu, Z.; Lei, Z.; Guo, J. Functions of EpCAM in physiological processes and diseases. Int. J. Mol. Med. 2018, 42, 1771–1785. [Google Scholar] [CrossRef] [Green Version]
  53. Trzpis, M.; McLaughlin, P.M.J.; de Leij, L.M.F.H.; Harmsen, M.C. Epithelial cell adhesion molecule: More than a carcinoma marker and adhesion molecule. Am. J. Pathol. 2007, 171, 386–395. [Google Scholar] [CrossRef] [Green Version]
  54. Münz, M.; Kieu, C.; Mack, B.; Schmitt, B.; Zeidler, R.; Gires, O. The carcinoma-associated antigen EpCAM upregulates c-myc and induces cell proliferation. Oncogene 2004, 23, 5748–5758. [Google Scholar] [CrossRef]
  55. Went, P.; Vasei, M.; Bubendorf, L.; Terracciano, L.; Tornillo, L.; Riede, U.; Kononen, J.; Simon, R.; Sauter, G.; Baeuerle, P.A. Frequent high-level expression of the immunotherapeutic target Ep-CAM in colon, stomach, prostate and lung cancers. Brit. J. Cancer 2006, 94, 128–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Kim, J.-H.; Herlyn, D.; Wong, K.-K.; Park, D.-C.; Schorge, J.O.; Lu, K.H.; Skates, S.J.; Cramer, D.W.; Berkowitz, R.S.; Mok, S.C. Identification of Epithelial Cell Adhesion Molecule Autoantibody in Patients with Ovarian Cancer. Clin. Cancer Res. 2003, 9, 4782–4791. [Google Scholar] [PubMed]
  57. Osta, W.A.; Chen, Y.; Mikhitarian, K.; Mitas, M.; Salem, M.; Hannun, Y.A.; Cole, D.J.; Gillanders, W.E. EpCAM Is Overexpressed in Breast Cancer and Is a Potential Target for Breast Cancer Gene Therapy. Cancer Res. 2004, 64, 5818–5824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Herlyn, M.; Steplewski, Z.; Herlyn, D.; Koprowski, H. Colorectal carcinoma-specific antigen: Detection by means of monoclonal antibodies. Proc. Natl. Acad. Sci. USA 1979, 76, 1438–1442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Schmidt, M.; Scheulen, M.E.; Dittrich, C.; Obrist, P.; Marschner, N.; Dirix, L.; Rüttinger, D.; Schuler, M.; Reinhardt, C.; Awada, A. An open-label, randomized phase II study of adecatumumab, a fully human anti-EpCAM antibody, as monotherapy in patients with metastatic breast cancer. Ann. Oncol. 2010, 21, 275–282. [Google Scholar] [CrossRef]
  60. Lopes, N.; Bergsland, C.; Bruun, J.; Bjørnslett, M.; Vieira, A.F.; Mesquita, P.; Pinto, R.; Gomes, R.; Cavadas, B.; Bennett, E. A panel of intestinal differentiation markers (CDX2, GPA33, and LI-cadherin) identifies gastric cancer patients with favourable prognosis. Gastric Cancer 2020, 23, 811–823. [Google Scholar] [CrossRef]
  61. Heath Joan , K.; White Sara , J.; Johnstone, C.N.; Catimel, B.; Simpson, R.J.; Moritz, R.L.; Tu, G.-F.; Ji, H.; Whitehead, R.H.; Groenen, L.C.; et al. The human A33 antigen is a transmembrane glycoprotein and a novel member of the immunoglobulin superfamily. Proc. Natl. Acad. Sci. USA 1997, 94, 469–474. [Google Scholar] [CrossRef] [Green Version]
  62. Wei, D.; Fan, Q.; Cai, H.; Yang, H.; Wan, L.; Li, L.; Lu, X. CF750-A33scFv-Fc-based optical imaging of subcutaneous and orthotopic xenografts of GPA33-positive colorectal cancer in mice. BioMed Res. Int. 2015, 2015, 505183. [Google Scholar] [CrossRef]
  63. Sawada, N.; Taguchi, E.; Takahashi, M. In vitro and in vivo activities of KRN330, a fully human monoclonal antibody against colon cancer. J. Clin. Oncol. 2011, 29, 432. [Google Scholar] [CrossRef]
  64. Berlin, J.D.; Infante, J.R.; Taguchi, E.; Goff, L.W.; Jones, S.F.; Chan, E.; Bendell, J.C.; Rothenberg, M.L.; Burris, H.A. In vivo antibody binding to tumor in xenograft rodent models and colorectal cancer patients treated with anti-A33 antibody KRN330. Cancer Res. 2010, 70, 2431. [Google Scholar]
  65. Iqbal, N.; Iqbal, N.J.M. Human epidermal growth factor receptor 2 (HER2) in cancers: Overexpression and therapeutic implications. Mol. Biol. Int. 2014, 2014, 852748. [Google Scholar] [CrossRef] [PubMed]
  66. Tzahar, E.; Waterman, H.; Chen, X.; Levkowitz, G.; Karunagaran, D.; Lavi, S.; Ratzkin, B.J.; Yarden, Y. A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Mol. Cell. Biol. 1996, 16, 5276–5287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Graus-Porta, D.; Beerli, R.R.; Daly, J.M.; Hynes, N.E. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J. 1997, 16, 1647–1655. [Google Scholar] [CrossRef] [PubMed]
  68. SiShi, L.; Buchbinder, E.; Wu, L.; Bjorge, J.D.; Fujita, D.J.; Zhu, S. EGFR and HER2 levels are frequently elevated in colon cancer cells. Discov. Rep. 2014, 1, 1. [Google Scholar]
  69. Lemoine, N.R.; Jain, S.; Silvestre, F.; Lopes, C.; Hughes, C.M.; McLelland, E.; Gullick, W.J.; Filipe, M.I. Amplification and overexpression of the EGF receptor and c-erbB-2 proto-oncogenes in human stomach cancer. Brit. J. Cancer 1991, 64, 79. [Google Scholar] [CrossRef] [Green Version]
  70. Oh, J.J.; Grosshans, D.R.; Wong, S.G.; Slamon, D.J. Identification of differentially expressed genes associated with HER-2/neu overexpression in human breast cancer cells. Nucleic Acids Res. 1999, 27, 4008–4017. [Google Scholar] [CrossRef] [Green Version]
  71. Tai, W.; Qin, B.; Cheng, K. Inhibition of breast cancer cell growth and invasiveness by dual silencing of HER-2 and VEGF. Mol. Pharm. 2010, 7, 543–556. [Google Scholar] [CrossRef]
  72. Roh, H.; Pippin, J.; Drebin, J.A. Down-regulation of HER2/neu expression induces apoptosis in human cancer cells that overexpress HER2/neu. Cancer Res. 2000, 60, 560–565. [Google Scholar]
  73. Slamon, D.J.; Leyland-Jones, B.; Shak, S.; Fuchs, H.; Paton, V.; Bajamonde, A.; Fleming, T.; Eiermann, W.; Wolter, J.; Pegram, M. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 2001, 344, 783–792. [Google Scholar] [CrossRef]
  74. Haridas, D.; Ponnusamy, M.P.; Chugh, S.; Lakshmanan, I.; Seshacharyulu, P.; Batra, S.K. MUC16: Molecular analysis and its functional implications in benign and malignant conditions. FASEB J. 2014, 28, 4183–4199. [Google Scholar] [CrossRef]
  75. Aithal, A.; Rauth, S.; Kshirsagar, P.; Shah, A.; Lakshmanan, I.; Junker, W.M.; Jain, M.; Ponnusamy, M.P.; Batra, S.K. MUC16 as a novel target for cancer therapy. Expert Opin. Tar. 2018, 22, 675–686. [Google Scholar] [CrossRef] [PubMed]
  76. Felder, M.; Kapur, A.; Gonzalez-Bosquet, J.; Horibata, S.; Heintz, J.; Albrecht, R.; Fass, L.; Kaur, J.; Hu, K.; Shojaei, H.; et al. MUC16 (CA125): Tumor biomarker to cancer therapy, a work in progress. Mol. Cancer 2014, 13, 129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Comamala, M.; Pinard, M.; Thériault, C.; Matte, I.; Albert, A.; Boivin, M.; Beaudin, J.; Piché, A.; Rancourt, C. Downregulation of cell surface CA125/MUC16 induces epithelial-to-mesenchymal transition and restores EGFR signalling in NIH:OVCAR3 ovarian carcinoma cells. Brit. J. Cancer 2011, 104, 989–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Liu, J.; Li, L.; Luo, N.; Liu, Q.; Liu, L.; Chen, D.; Cheng, Z.; Xi, X.J.E.; Medicine, T. Inflammatory signals induce MUC16 expression in ovarian cancer cells via NF-κB activation. Exp. Ther. Med. 2021, 21, 1. [Google Scholar] [CrossRef] [PubMed]
  79. Berek, J.S.; Taylor, P.T.; Gordon, A.; Cunningham, M.J.; Finkler, N.; Orr, J., Jr.; Rivkin, S.; Schultes, B.C.; Whiteside, T.L.; Nicodemus, C.F. Randomized, placebo-controlled study of oregovomab for consolidation of clinical remission in patients with advanced ovarian cancer. J. Clin. Oncol. 2004, 22, 3507–3516. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, B.; Wu, A.W.; Hu, Y.Q.; Tao, C.J.; Wang, J.M.; Lu, Y.Y.; Xing, R. Mucin 17 inhibits the progression of human gastric cancer by limiting inflammatory responses through a MYH9-p53-RhoA regulatory feedback loop. J. Exp. Clin. Canc. Res. 2019, 38, 283. [Google Scholar] [CrossRef]
  81. Lordick, F.; Orra, E.B.; Cervantes, A.; Dayyani, F.; Rocha-Lima, C.; Greil, R.; van Laarhoven, H.; Lorenzen, S.; Kischel, R.; Shitara, K. P-76 A phase 1 study of AMG 199, a half-life extended bispecific T-cell engager (HLE BiTE®) immune therapy, targeting MUC17 in patients with gastric and gastroesophageal junction cancer. Ann. Oncol. 2020, 31, S114. [Google Scholar] [CrossRef]
  82. Panwar, H.; Rokana, N.; Aparna, S.V.; Kaur, J.; Singh, A.; Singh, J.; Singh, K.S.; Chaudhary, V.; Puniya, A.K. Gastrointestinal stress as innate defence against microbial attack. J. Appl. Microbiol. 2021, 130, 1035–1061. [Google Scholar] [CrossRef]
  83. Junker, W.M. Molecular and Biological Studies of MUC17; University of Nebraska Medical Center: Omaha, NE, USA, 2008. [Google Scholar]
  84. MUC17. Available online: https://www.amgenoncology.com/targets/MUC17.html (accessed on 14 April 2022).
  85. Chang, S.S. Overview of prostate-specific membrane antigen. Rev. Urol. 2004, 6, S13. [Google Scholar]
  86. Wang, F.; Li, Z.; Feng, X.; Yang, D.; Lin, M. Advances in PSMA-targeted therapy for prostate cancer. Prostate Cancer Prostatic Dis. 2021, 25, 1–16. [Google Scholar]
  87. Oh, S.W.; Cheon, G.J. Prostate-specific membrane antigen PET imaging in prostate cancer: Opportunities and challenges. Korean J. Radiol. 2018, 19, 819–831. [Google Scholar] [CrossRef] [PubMed]
  88. Bostwick, D.G.; Pacelli, A.; Blute, M.; Roche, P.; Murphy, G.P. Prostate specific membrane antigen expression in prostatic intraepithelial neoplasia and adenocarcinoma. Cancer 1998, 82, 2256–2261. [Google Scholar] [CrossRef]
  89. Ross, J.S.; Sheehan, C.E.; Fisher, H.A.G.; Kaufman, R.P., Jr.; Kaur, P.; Gray, K.; Webb, I.; Gray, G.S.; Mosher, R.; Kallakury, B.V.S. Correlation of Primary Tumor Prostate-Specific Membrane Antigen Expression with Disease Recurrence in Prostate Cancer. Clin. Cancer Res. 2003, 9, 6357–6362. [Google Scholar]
  90. Trover, J.K.; Beckett, M.L.; Wright, G.L., Jr. Detection and characterization of the prostate-specific membrane antigen (PSMA) in tissue extracts and body fluids. Int. J. Cancer 1995, 62, 552–558. [Google Scholar] [CrossRef] [PubMed]
  91. Tagawa, S.T.; Vallabhajosula, S.; Christos, P.J.; Jhanwar, Y.S.; Batra, J.S.; Lam, L.; Osborne, J.; Beltran, H.; Molina, A.M.; Goldsmith, S.J.J.C. Phase 1/2 study of fractionated dose lutetium-177–labeled anti–prostate-specific membrane antigen monoclonal antibody J591 (177Lu-J591) for metastatic castration-resistant prostate cancer. Cancer 2019, 125, 2561–2569. [Google Scholar] [CrossRef]
  92. Baek, J.H.; Park, D.J.; Kim, G.Y.; Cheon, J.; Kang, B.W.; Cha, H.J.; Kim, J. Clinical implications of Claudin18. 2 expression in patients with gastric cancer. Anticancer. Res. 2019, 39, 6973–6979. [Google Scholar] [CrossRef]
  93. Hashimoto, Y.; Yagi, K.; Kondoh, M. Current progress in a second-generation claudin binder, anti-claudin antibody, for clinical applications. Drug Discov. Today 2016, 21, 1711–1718. [Google Scholar] [CrossRef] [Green Version]
  94. Lin, H.; Zhang, R. Abstract B73: Development of anti-human CLDN18. 2 monoclonal antibody as cancer therapeutics. Cancer Immunol. Res. 2020, 8, B73. [Google Scholar]
  95. Sanada, Y.; Oue, N.; Mitani, Y.; Yoshida, K.; Nakayama, H.; Yasui, W. Down-regulation of the claudin-18 gene, identified through serial analysis of gene expression data analysis, in gastric cancer with an intestinal phenotype. J. Pathol. 2006, 208, 633–642. [Google Scholar] [CrossRef]
  96. Singh, P.; Toom, S.; Huang, Y.J. Anti-claudin 18.2 antibody as new targeted therapy for advanced gastric cancer. J. Hematol. Oncol. 2017, 10, 105. [Google Scholar] [CrossRef]
  97. Barroca-Ferreira, J.; Cruz-Vicente, P.; Santos, M.F.; Rocha, S.M.; Santos-Silva, T.; Maia, C.J.; Passarinha, L.A. Enhanced Stability of Detergent-Free Human Native STEAP1 Protein from Neoplastic Prostate Cancer Cells upon an Innovative Isolation Procedure. Int. J. Mol. Sci. 2021, 22, 10012. [Google Scholar] [CrossRef]
  98. Hubert Rene, S.; Vivanco, I.; Chen, E.; Rastegar, S.; Leong, K.; Mitchell Steve, C.; Madraswala, R.; Zhou, Y.; Kuo, J.; Raitano Arthur, B.; et al. STEAP: A prostate-specific cell-surface antigen highly expressed in human prostate tumors. Proc. Natl. Acad. Sci. USA 1999, 96, 14523–14528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Wu, Y.-Y.; Jiang, J.-N.; Fang, X.-D.; Ji, F.-J.J. STEAP1 regulates tumorigenesis and chemoresistance during peritoneal metastasis of gastric cancer. Front. Physiol. 2018, 9, 1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Chen, W.-J.; Wu, H.-T.; Li, C.-L.; Lin, Y.-K.; Fang, Z.-X.; Lin, W.-T.; Liu, J. Regulatory Roles of Six-Transmembrane Epithelial Antigen of the Prostate Family Members in the Occurrence and Development of Malignant Tumors. Front. Cell Dev. Biol. 2021, 9, 2988. [Google Scholar] [CrossRef] [PubMed]
  101. Rocha, S.; Barroca-Ferreira, J.; Passarinha, L.; Socorro, S.; Maia, C.J.E.P. The Usefulness of STEAP Proteins in Prostate Cancer Clinical Practice. Exon Publ. 2021, 10, 139–153. [Google Scholar]
  102. Activity of Anti-STEAP1 Antibody-Drug Conjugate in Patients With Metastatic Castration-Resistant Prostate Cancer. Available online: https://ascopost.com/news/november-2019/activity-of-anti-steap1-antibody-drug-conjugate-in-patients-with-mcrpc/ (accessed on 30 March 2022).
  103. Lehman, J.; Hoeksema, M.; Chen, H.; Shi, C.; Eisenberg, R.; Massion, P.P. Loss of somatostatin receptor 2 expression and lung cancer growth. J. Clin. Oncol. 2015, 33, 7569. [Google Scholar] [CrossRef]
  104. Lehman, J.M.; Hoeksema, M.D.; Staub, J.; Qian, J.; Harris, B.; Callison, J.C.; Miao, J.; Shi, C.; Eisenberg, R.; Chen, H. Somatostatin receptor 2 signaling promotes growth and tumor survival in small-cell lung cancer. Int. J. Cancer 2019, 144, 1104–1114. [Google Scholar] [CrossRef]
  105. Reisine, T.; Bell, G.I. Molecular biology of somatostatin receptors. Endocr. Rev. 1995, 16, 427–442. [Google Scholar]
  106. Si, Y.; Kim, S.; Ou, J.; Lu, Y.; Ernst, P.; Chen, K.; Whitt, J.; Carter, A.M.; Markert, J.M.; Bibb, J.A. Anti-SSTR2 antibody-drug conjugate for neuroendocrine tumor therapy. Cancer Gene Ther. 2021, 28, 799–812. [Google Scholar] [CrossRef]
  107. Kammerer, R.; Zimmermann, W. Coevolution of activating and inhibitory receptors within mammalian carcinoembryonic antigen families. BMC Biol. 2010, 8, 12. [Google Scholar] [CrossRef] [Green Version]
  108. Benchimol, S.; Fuks, A.; Jothy, S.; Beauchemin, N.; Shirota, K.; Stanners, C.P. Carcinoembryonic antigen, a human tumor marker, functions as an intercellular adhesion molecule. Cell 1989, 57, 327–334. [Google Scholar] [CrossRef]
  109. Oikawa, S.; Inuzuka, C.; Kuroki, M.; Matsuoka, Y.; Kosaki, G.; Nakazato, H. Cell adhesion activity of non-specific cross-reacting antigen (NCA) and carcinoembryonic antigen (CEA) expressed on CHO cell surface: Homophilic and heterophilic adhesion. Biochem. Biophys. Res. Commun. 1989, 164, 39–45. [Google Scholar] [CrossRef]
  110. Lee, J.H.; Lee, S.-W. The roles of carcinoembryonic antigen in liver metastasis and therapeutic approaches. Gastroenterol. Res. Pract. 2017, 2017, 7521987. [Google Scholar] [CrossRef]
  111. Deng, K.; Yang, L.; Hu, B.; Wu, H.; Zhu, H.; Tang, C. The Prognostic Significance of Pretreatment Serum CEA Levels in Gastric Cancer: A Meta-Analysis Including 14651 Patients. PLoS ONE 2015, 10, e0124151. [Google Scholar] [CrossRef]
  112. Pakdel, A.; Malekzadeh, M.; Naghibalhossaini, F. The association between preoperative serum CEA concentrations and synchronous liver metastasis in colorectal cancer patients. Cancer Biomark. 2016, 16, 245–252. [Google Scholar] [CrossRef]
  113. Auclin, E.; Taieb, J.; Lepage, C.; Aparicio, T.; Faroux, R.; Mini, E.; Folprecht, G.; Salazar, R.; Benetkiewicz, M.; Banzi, M.J.C.E.; et al. Carcinoembryonic antigen levels and survival in stage III colon cancer: Post hoc analysis of the MOSAIC and PETACC-8 trials. Cancer Epidemiol. Prev. Biomark. 2019, 28, 1153–1161. [Google Scholar] [CrossRef] [PubMed]
  114. Campos-da-Paz, M.; Dórea, J.G.; Galdino, A.S.; Lacava, Z.G.M.; de Fatima, M.A.S. Carcinoembryonic antigen (CEA) and hepatic metastasis in colorectal cancer: Update on biomarker for clinical and biotechnological approaches. Recent Pat. Biotechnol. 2018, 12, 269–279. [Google Scholar] [CrossRef] [PubMed]
  115. Beauchemin, N.; Arabzadeh, A. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev. 2013, 32, 643–671. [Google Scholar] [CrossRef]
  116. Thomas, P.; Gangopadhyay, A.; Steele, G., Jr.; Andrews, C.; Nakazato, H.; Oikawa, S.; Jessup, J.M. The effect of transfection of the CEA gene on the metastatic behavior of the human colorectal cancer cell line MIP-101. Cancer Lett. 1995, 92, 59–66. [Google Scholar] [CrossRef]
  117. Stein, R.; Goldenberg, D.M. A humanized monoclonal antibody to carcinoembryonic antigen, labetuzumab, inhibits tumor growth and sensitizes human medullary thyroid cancer xenografts to dacarbazine chemotherapy. Mol. Cancer Ther. 2004, 3, 1559–1564. [Google Scholar]
  118. Segal, N.H.; Verghis, J.; Govindan, S.; Maliakal, P.; Sharkey, R.M.; Wegener, W.A.; Goldenberg, D.M.; Saltz, L.B. Abstract LB-159: A Phase I study of IMMU-130 (labetuzumab-SN38) anti-CEACAM5 antibody-drug conjugate (ADC) in patients with metastatic colorectal cancer (mCRC). Cancer Res. 2013, 73, LB-159. [Google Scholar]
  119. Govindan, S.V.; Cardillo, T.M.; Rossi, E.A.; McBride, W.J.; Sharkey, R.M.; Goldenberg, D.M. IMMU-130, a unique antibody-drug conjugate (ADC) of SN-38 targeting CEACAM5 antigen: Preclinical basis for clinical activity in metastatic colorectal cancer (mCRC). J. Clin. Oncol. 2015, 33, 625. [Google Scholar] [CrossRef]
  120. Li, N.; Spetz, M.R.; Ho, M. The Role of Glypicans in Cancer Progression and Therapy. J. Histochem. Cytochem. 2020, 68, 841–862. [Google Scholar] [CrossRef] [PubMed]
  121. Ofuji, K.; Saito, K.; Yoshikawa, T.; Nakatsura, T. Critical analysis of the potential of targeting GPC3 in hepatocellular carcinoma. J. Hepatocell. Carcinoma 2014, 1, 35. [Google Scholar] [PubMed] [Green Version]
  122. Capurro, M.I.; Xiang, Y.-Y.; Lobe, C.; Filmus, J. Glypican-3 Promotes the Growth of Hepatocellular Carcinoma by Stimulating Canonical Wnt Signaling. Cancer Res. 2005, 65, 6245–6254. [Google Scholar] [CrossRef] [Green Version]
  123. Aviel-Ronen, S.; Lau, S.K.; Pintilie, M.; Lau, D.; Liu, N.; Tsao, M.S.; Jothy, S. Glypican-3 is overexpressed in lung squamous cell carcinoma, but not in adenocarcinoma. Mod. Pathol. 2008, 21, 817–825. [Google Scholar] [CrossRef] [Green Version]
  124. Maeda, D.; Ota, S.; Takazawa, Y.; Aburatani, H.; Nakagawa, S.; Yano, T.; Taketani, Y.; Kodama, T.; Fukayama, M. Glypican-3 expression in clear cell adenocarcinoma of the ovary. Mod. Pathol. 2009, 22, 824–832. [Google Scholar] [CrossRef] [Green Version]
  125. Ishiguro, T.; Sugimoto, M.; Kinoshita, Y.; Miyazaki, Y.; Nakano, K.; Tsunoda, H.; Sugo, I.; Ohizumi, I.; Aburatani, H.; Hamakubo, T. Anti–glypican 3 antibody as a potential antitumor agent for human liver cancer. Cancer Res. 2008, 68, 9832–9838. [Google Scholar] [CrossRef] [Green Version]
  126. Doronin, I.I.; Vishnyakova, P.A.; Kholodenko, I.V.; Ponomarev, E.D.; Ryazantsev, D.Y.; Molotkovskaya, I.M.; Kholodenko, R.V. Ganglioside GD2 in reception and transduction of cell death signal in tumor cells. BMC Cancer 2014, 14, 295. [Google Scholar] [CrossRef] [Green Version]
  127. Nazha, B.; Inal, C.; Owonikoko, T.K. Disialoganglioside GD2 expression in solid tumors and role as a target for cancer therapy. Front. Oncol. 2020, 10, 1000. [Google Scholar] [CrossRef]
  128. Dobrenkov, K.; Cheung, N.-K.V. GD2-targeted immunotherapy and radioimmunotherapy. Semin. Oncol. 2014, 41, 589–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. GD2 Ganglioside. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/GD2-Ganglioside (accessed on 15 April 2022).
  130. Yoshida, S.; Fukumoto, S.; Kawaguchi, H.; Sato, S.; Ueda, R.; Furukawa, K. Ganglioside GD2 in small cell lung cancer cell lines: Enhancement of cell proliferation and mediation of apoptosis. Cancer Res. 2001, 61, 4244–4252. [Google Scholar]
  131. Navid, F.; Santana, V.M.; Barfield, R.C. Anti-GD2 antibody therapy for GD2-expressing tumors. Curr. Cancer Drug Targets 2010, 10, 200–209. [Google Scholar] [CrossRef] [PubMed]
  132. Cheresh, D.A.; Pierschbacher, M.D.; Herzig, M.A.; Mujoo, K. Disialogangliosides GD2 and GD3 are involved in the attachment of human melanoma and neuroblastoma cells to extracellular matrix proteins. J. Cell Biol. 1986, 102, 688–696. [Google Scholar] [CrossRef]
  133. Cheung, N.; Kushner, B.H.; Yeh, S.; Larson, S.M. 3F8 monoclonal antibody treatment of patients with stage 4 neuroblastoma: A phase II study. Int. J. Oncol. 1998, 12, 1299–1605. [Google Scholar] [CrossRef] [PubMed]
  134. Shah, N.; Chari, A.; Scott, E.; Mezzi, K.; Usmani, S.Z. B-cell maturation antigen (BCMA) in multiple myeloma: Rationale for targeting and current therapeutic approaches. Leukemia 2020, 34, 985–1005. [Google Scholar] [CrossRef]
  135. Schiemann, B.; Gommerman Jennifer, L.; Vora, K.; Cachero Teresa, G.; Shulga-Morskaya, S.; Dobles, M.; Frew, E.; Scott Martin, L. An Essential Role for BAFF in the Normal Development of B Cells Through a BCMA-Independent Pathway. Science 2001, 293, 2111–2114. [Google Scholar] [CrossRef]
  136. Yu, G.; Boone, T.; Delaney, J.; Hawkins, N.; Kelley, M.; Ramakrishnan, M.; McCabe, S.; Qiu, W.-R.; Kornuc, M.; Xia, X.-Z.; et al. APRIL and TALL-1 and receptors BCMA and TACI: System for regulating humoral immunity. Nat. Immunol. 2000, 1, 252–256. [Google Scholar] [CrossRef] [Green Version]
  137. Tai, Y.-T.; Acharya, C.; An, G.; Moschetta, M.; Zhong, M.Y.; Feng, X.; Cea, M.; Cagnetta, A.; Wen, K.; van Eenennaam, H.; et al. APRIL and BCMA promote human multiple myeloma growth and immunosuppression in the bone marrow microenvironment. Blood 2016, 127, 3225–3236. [Google Scholar] [CrossRef] [Green Version]
  138. Miao, Y.R.; Cenik, C.; Jiang, D.; Mizuno, K.; Li, G.C.; Zhao, H.; Thakker, K.; Diep, A.; Xu, J.Y.; Zhang, X.E.; et al. Aberrant BCMA Signaling Promotes Tumor Growth by Altering Protein Translation Machinery, a Therapeutic Target for the Treatment of Relapse/Refractory Multiple Myeloma. bioRxiv 2021. [Google Scholar] [CrossRef]
  139. Sanchez, E.; Li, M.; Kitto, A.; Li, J.; Wang, C.S.; Kirk, D.T.; Yellin, O.; Nichols, C.M.; Dreyer, M.P.; Ahles, C.P.; et al. Serum B-cell maturation antigen is elevated in multiple myeloma and correlates with disease status and survival. Br. J. Haematol. 2012, 158, 727–738. [Google Scholar] [CrossRef] [PubMed]
  140. Ketchum, E.B.; Clarke, A.; Clemmons, A.B. Belantamab Mafodotin-blmf: A Novel Antibody-Drug Conjugate for Treatment of Patients With Relapsed/Refractory Multiple Myeloma. J. Adv. Pract. Oncol. 2022, 13, 77–85. [Google Scholar] [CrossRef] [PubMed]
  141. Tai, Y.-T.; Mayes, P.A.; Acharya, C.; Zhong, M.Y.; Cea, M.; Cagnetta, A.; Craigen, J.; Yates, J.; Gliddon, L.; Fieles, W.; et al. Novel anti–B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma. Blood 2014, 123, 3128–3138. [Google Scholar] [CrossRef] [PubMed]
  142. Tedder, T.F.; Zhou, L.-J.; Engel, P. The CD19/CD21 signal transduction complex of B lymphocytes. Immunol. Today 1994, 15, 437–442. [Google Scholar] [CrossRef]
  143. Van Zelm, M.C.; Reisli, I.; van der Burg, M.; Castaño, D.; van Noesel, C.J.M.; van Tol, M.J.D.; Woellner, C.; Grimbacher, B.; Patiño, P.J.; van Dongen, J.J.M. An antibody-deficiency syndrome due to mutations in the CD19 gene. N. Engl. J. Med. 2006, 354, 1901–1912. [Google Scholar] [CrossRef] [Green Version]
  144. Carter, R.H.; Fearon, D.T. CD19: Lowering the threshold for antigen receptor stimulation of B lymphocytes. Science 1992, 256, 105–107. [Google Scholar] [CrossRef]
  145. Anderson, K.C.; Bates, M.P.; Slaughenhoupt, B.L.; Pinkus, G.S.; Schlossman, S.F.; Nadler, L.M. Expression of human B cell-associated antigens on leukemias and lymphomas: A model of human B cell differentiation. Blood 1984, 63, 1424–1433. [Google Scholar] [CrossRef] [Green Version]
  146. He, X.; Kläsener, K.; Iype, J.M.; Becker, M.; Maity, P.C.; Cavallari, M.; Nielsen, P.J.; Yang, J.; Reth, M. Continuous signaling of CD 79b and CD 19 is required for the fitness of Burkitt lymphoma B cells. EMBO J. 2018, 37, e97980. [Google Scholar] [CrossRef]
  147. Jurczak, W.; Zinzani, P.L.; Gaidano, G.; Goy, A.; Provencio, M.; Nagy, Z.; Robak, T.; Maddocks, K.; Buske, C.; Ambarkhane, S.; et al. Phase IIa study of the CD19 antibody MOR208 in patients with relapsed or refractory B-cell non-Hodgkin’s lymphoma. Ann. Oncol. 2018, 29, 1266–1272. [Google Scholar] [CrossRef]
  148. Walker, J.A.; Smith, K.G.C. CD22: An inhibitory enigma. Immunology 2008, 123, 314–325. [Google Scholar] [CrossRef]
  149. Nitschke, L. The role of CD22 and other inhibitory co-receptors in B-cell activation. Curr. Opin. Immunol. 2005, 17, 290–297. [Google Scholar] [CrossRef] [PubMed]
  150. Shah, N.N.; Stevenson, M.S.; Yuan, C.M.; Richards, K.; Delbrook, C.; Kreitman, R.J.; Pastan, I.; Wayne, A.S. Characterization of CD22 expression in acute lymphoblastic leukemia. Pediatric Blood Cancer 2015, 62, 964–969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Jasper, G.A.; Arun, I.; Venzon, D.; Kreitman, R.J.; Wayne, A.S.; Yuan, C.M.; Marti, G.E.; Stetler-Stevenson, M. Variables affecting the quantitation of CD22 in neoplastic B cells. Cytom. Part B Clin. Cytom. 2011, 80B, 83–90. [Google Scholar] [CrossRef] [PubMed]
  152. Schweizer, A.; Wöhner, M.; Prescher, H.; Brossmer, R.; Nitschke, L. Targeting of CD 22-positive B-cell lymphoma cells by synthetic divalent sialic acid analogues. Eur. J. Immunol. 2012, 42, 2792–2802. [Google Scholar] [CrossRef]
  153. Ereño-Orbea, J.; Sicard, T.; Cui, H.; Mazhab-Jafari, M.T.; Benlekbir, S.; Guarné, A.; Rubinstein, J.L.; Julien, J.-P. Molecular basis of human CD22 function and therapeutic targeting. Nat. Commun. 2017, 8, 764. [Google Scholar] [CrossRef]
  154. Leonard, J.P.; Coleman, M.; Ketas, J.; Ashe, M.; Fiore, J.M.; Furman, R.R.; Niesvizky, R.; Shore, T.; Chadburn, A.; Horne, H.; et al. Combination Antibody Therapy With Epratuzumab and Rituximab in Relapsed or Refractory Non-Hodgkin’s Lymphoma. J. Clin. Oncol. 2005, 23, 5044–5051. [Google Scholar] [CrossRef]
  155. Pierpont, T.M.; Limper, C.B.; Richards, K.L. Past, Present, and Future of Rituximab—The World’s First Oncology Monoclonal Antibody Therapy. Front. Oncol. 2018, 8, 168. [Google Scholar] [CrossRef]
  156. Nagata, S.; Ise, T.; Onda, M.; Nakamura, K.; Ho, M.; Raubitschek, A.; Pastan, I.H. Cell membrane-specific epitopes on CD30: Potentially superior targets for immunotherapy. Proc. Natl. Acad. Sci. USA 2005, 102, 7946–7951. [Google Scholar] [CrossRef] [Green Version]
  157. Horie, R.; Watanabe, T.; Morishita, Y.; Ito, K.; Ishida, T.; Kanegae, Y.; Saito, I.; Higashihara, M.; Mori, S.; Kadin, M.E.; et al. Ligand-independent signaling by overexpressed CD30 drives NF-κB activation in Hodgkin–Reed-Sternberg cells. Oncogene 2002, 21, 2493–2503. [Google Scholar] [CrossRef] [Green Version]
  158. Guo, F.; Sun, A.; Wang, W.; He, J.; Hou, J.; Zhou, P.; Chen, Z. TRAF1 is involved in the classical NF-κB activation and CD30-induced alternative activity in Hodgkin’s lymphoma cells. Mol. Immunol. 2009, 46, 2441–2448. [Google Scholar] [CrossRef]
  159. Barta, S.K.; Gong, J.Z.; Porcu, P. Brentuximab vedotin in the treatment of CD30+ PTCL. Blood 2019, 134, 2339–2345. [Google Scholar] [CrossRef] [PubMed]
  160. Ranuhardy, D.; Suzanna, E.; Sari, R.M.; Hadisantoso, D.W.; Andalucia, R.; Abdillah, A. CD30, CD15, CD50, and PAX5 expressions as diagnostic markers for Hodgkin lymphoma (HL) and systemic anaplastic large cell lymphoma (sALCL). Acta Med. Indones. 2018, 50, 104–109. [Google Scholar] [PubMed]
  161. Lawrence, C.E.; Hammond, P.W.; Zalevsky, J.; Horton, H.; Chu, S.; Karki, S.; Desjarlais, J.R.; Carmichael, D.F. XmAb™2513, an Fc Engineered Humanized Anti-CD30 Monoclonal Antibody, Has Potent In Vitro and In Vivo Activities, and Has the Potential for Treating Hematologic Malignancies. Blood 2007, 110, 2340. [Google Scholar] [CrossRef]
  162. Álvarez, B.; Escalona, Z.; Uenishi, H.; Toki, D.; Revilla, C.; Yuste, M.; del Moral, M.G.; Alonso, F.; Ezquerra, A.; Domínguez, J. Molecular and functional characterization of porcine Siglec-3/CD33 and analysis of its expression in blood and tissues. Dev. Comp. Immunol. 2015, 51, 238–250. [Google Scholar] [CrossRef]
  163. Crocker, P.R.; McMillan, S.J.; Richards, H.E. CD33-related siglecs as potential modulators of inflammatory responses. Ann. New York Acad. Sci. 2012, 1253, 102–111. [Google Scholar] [CrossRef]
  164. Läubli, H.; Alisson-Silva, F.; Stanczak, M.A.; Siddiqui, S.S.; Deng, L.; Verhagen, A.; Varki, N.; Varki, A. Lectin galactoside-binding soluble 3 binding protein (LGALS3BP) is a tumor-associated immunomodulatory ligand for CD33-related Siglecs. J. Biol. Chem. 2014, 289, 33481–33491. [Google Scholar] [CrossRef] [Green Version]
  165. De Propris, M.S.; Raponi, S.; Diverio, D.; Milani, M.L.; Meloni, G.; Falini, B.; Foà, R.; Guarini, A. High CD33 expression levels in acute myeloid leukemia cells carrying the nucleophosmin (NPM1) mutation. Haematologica 2011, 96, 1548. [Google Scholar] [CrossRef] [Green Version]
  166. Liu, J.; Tong, J.; Yang, H. Targeting CD33 for acute myeloid leukemia therapy. BMC Cancer 2022, 22, 24. [Google Scholar] [CrossRef]
  167. Williams, B.A.; Law, A.; Hunyadkurti, J.; Desilets, S.; Leyton, J.V.; Keating, A. Antibody therapies for acute myeloid leukemia: Unconjugated, toxin-conjugated, radio-conjugated and multivalent formats. J. Clin. Med. 2019, 8, 1261. [Google Scholar] [CrossRef] [Green Version]
  168. Larson, R.A.; Sievers, E.L.; Stadtmauer, E.A.; Löwenberg, B.; Estey, E.H.; Dombret, H.; Theobald, M.; Voliotis, D.; Bennett, J.M.; Richie, M. Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence. Cancer 2005, 104, 1442–1452. [Google Scholar] [CrossRef]
  169. Gao, Y.; Mehta, K. N-linked glycosylation of CD38 is required for its structure stabilization but not for membrane localization. Mol. Cell. Biochem. 2007, 295, 1–7. [Google Scholar] [CrossRef] [PubMed]
  170. Malavasi, F.; Deaglio, S.; Funaro, A.; Ferrero, E.; Horenstein, A.L.; Ortolan, E.; Vaisitti, T.; Aydin, S. Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol. Rev. 2008, 88, 841–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Hu, Y.; Liu, H.; Fang, C.; Li, C.; Xhyliu, F.; Dysert, H.; Bodo, J.; Habermehl, G.; Russell, B.E.; Li, W. Targeting of CD38 by the tumor suppressor miR-26a serves as a novel potential therapeutic agent in multiple myeloma. Cancer Res. 2020, 80, 2031–2044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Irimia, R.M.; Gerke, M.B.; Thakar, M.; Ren, Z.; Helmenstine, E.; Imus, P.H.; Ghiaur, G.; Leone, R.; Gocke, C.B. CD38 Is a Key Regulator of Tumor Growth By Modulating the Metabolic Signature of Malignant Plasma Cells. Blood 2021, 138, 2652. [Google Scholar] [CrossRef]
  173. Sanchez, L.; Wang, Y.; Siegel, D.S.; Wang, M.L. Daratumumab: A first-in-class CD38 monoclonal antibody for the treatment of multiple myeloma. J. Hematol. Oncol. 2016, 9, 51. [Google Scholar] [CrossRef] [Green Version]
  174. Palumbo, A.; Chanan-Khan, A.; Weisel, K.; Nooka, A.K.; Masszi, T.; Beksac, M.; Spicka, I.; Hungria, V.; Munder, M.; Mateos, M.V. Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N. Engl. J. Med. 2016, 375, 754–766. [Google Scholar] [CrossRef]
  175. Hoseini, S.S.; Cheung, N.K. Acute myeloid leukemia targets for bispecific antibodies. Blood Cancer J. 2017, 7, e522. [Google Scholar] [CrossRef]
  176. Angelova, E.; Audette, C.; Kovtun, Y.; Daver, N.; Wang, S.A.; Pierce, S.; Konoplev, S.N.; Khogeer, H.; Jorgensen, J.L.; Konopleva, M.; et al. CD123 expression patterns and selective targeting with a CD123-targeted antibody-drug conjugate (IMGN632) in acute lymphoblastic leukemia. Haematologica 2019, 104, 749–755. [Google Scholar] [CrossRef]
  177. Patnaik, M.M.; Mughal, T.I.; Brooks, C.; Lindsay, R.; Pemmaraju, N. Targeting CD123 in hematologic malignancies: Identifying suitable patients for targeted therapy. Leuk. Lymphoma 2021, 62, 2568–2586. [Google Scholar] [CrossRef]
  178. Muñoz, L.; Nomdedéu, J.F.; López, O.; Carnicer, M.J.; Bellido, M.; Aventín, A.; Brunet, S.; Sierra, J. Interleukin-3 receptor alpha chain (CD123) is widely expressed in hematologic malignancies. Haematologica 2001, 86, 1261–1269. [Google Scholar]
  179. Aldinucci, D.; Poletto, D.; Gloghini, A.; Nanni, P.; Degan, M.; Perin, T.; Ceolin, P.; Rossi, F.M.; Gattei, V.; Carbone, A.; et al. Expression of Functional Interleukin-3 Receptors on Hodgkin and Reed-Sternberg Cells. Am. J. Pathol. 2002, 160, 585–596. [Google Scholar] [CrossRef] [Green Version]
  180. Jin, L.; Lee, E.M.; Ramshaw, H.S.; Busfield, S.J.; Peoppl, A.G.; Wilkinson, L.; Guthridge, M.A.; Thomas, D.; Barry, E.F.; Boyd, A. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor α chain, eliminates human acute myeloid leukemic stem cells. Cell. Stem. Cell. 2009, 5, 31–42. [Google Scholar] [CrossRef] [PubMed]
  181. Kovtun, Y.; Jones, G.E.; Adams, S.; Harvey, L.; Audette, C.A.; Wilhelm, A.; Bai, C.; Rui, L.; Laleau, R.; Liu, F.; et al. A CD123-targeting antibody-drug conjugate, IMGN632, designed to eradicate AML while sparing normal bone marrow cells. Blood Adv. 2018, 2, 848–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Ma, H.; Padmanabhan, I.S.; Parmar, S.; Gong, Y. Targeting CLL-1 for acute myeloid leukemia therapy. J. Hematol. Oncol. 2019, 12, 41. [Google Scholar] [CrossRef] [PubMed]
  183. Neumann, K.; Castiñeiras-Vilariño, M.; Höckendorf, U.; Hannesschläger, N.; Lemeer, S.; Kupka, D.; Meyermann, S.; Lech, M.; Anders, H.-J.; Kuster, B.; et al. Clec12a Is an Inhibitory Receptor for Uric Acid Crystals that Regulates Inflammation in Response to Cell Death. Immunity 2014, 40, 389–399. [Google Scholar] [CrossRef] [Green Version]
  184. Bakker, A.B.H.; van den Oudenrijn, S.; Bakker, A.Q.; Feller, N.; van Meijer, M.; Bia, J.A.; Jongeneelen, M.A.C.; Visser, T.J.; Bijl, N.; Geuijen, C.A.W.; et al. C-Type Lectin-Like Molecule-1: A Novel Myeloid Cell Surface Marker Associated with Acute Myeloid Leukemia. Cancer Res. 2004, 64, 8443–8450. [Google Scholar] [CrossRef] [Green Version]
  185. Haubner, S.; Perna, F.; Köhnke, T.; Schmidt, C.; Berman, S.; Augsberger, C.; Schnorfeil, F.M.; Krupka, C.; Lichtenegger, F.S.; Liu, X.; et al. Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML. Leukemia 2019, 33, 64–74. [Google Scholar] [CrossRef]
  186. Kenderian, S.S.; Ruella, M.; Shestova, O.; Klichinsky, M.; Kim, M.; Soderquist, C.; Bagg, A.; Singh, R.; Richardson, C.; Young, R. Targeting CLEC12A with chimeric antigen receptor T cells can overcome the chemotherapy refractoriness of leukemia stem cells. Biol. Blood Marrow Transplant. 2017, 23, S247–S248. [Google Scholar] [CrossRef]
  187. Xiaoxian, Z.; Shweta, S.; Cecile, P.; Jingsong, Z.; Eric, D.H.; Arie, A.; Wouter, K. Targeting C-type lectin-like molecule-1 for antibody-mediated immunotherapy in acute myeloid leukemia. Haematologica 2010, 95, 71–78. [Google Scholar] [CrossRef]
  188. Gilliland, D.G.; Griffin, J.D. The roles of FLT3 in hematopoiesis and leukemia. Blood 2002, 100, 1532–1542. [Google Scholar] [CrossRef] [Green Version]
  189. Rasko, J.E.; Metcalf, D.; Rossner, M.T.; Begley, C.G.; Nicola, N.A. The flt3/flk-2 ligand: Receptor distribution and action on murine haemopoietic cell survival and proliferation. Leukemia 1995, 9, 2058–2066. [Google Scholar] [PubMed]
  190. Rusten, L.S.; Lyman, S.D.; Veiby, O.P.; Jacobsen, S.E. The FLT3 ligand is a direct and potent stimulator of the growth of primitive and committed human CD34+ bone marrow progenitor cells in vitro. Blood 1996, 87, 1317–1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Poubel, C.P.; Mansur, M.B.; Boroni, M.; Emerenciano, M. FLT3 overexpression in acute leukaemias: New insights into the search for molecular mechanisms. Biochim. Et Biophys. Acta (BBA)-Rev. Cancer 2019, 1872, 80–88. [Google Scholar] [CrossRef]
  192. Radich, J.P.; Kopecky, K.J.; Willman, C.L.; Weick, J.; Head, D.; Appelbaum, F.; Collins, S.J. N-ras mutations in adult de novo acute myelogenous leukemia: Prevalence and clinical significance. Blood 1990, 76, 801–807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Kennedy, V.E.; Smith, C.C. FLT3 Mutations in Acute Myeloid Leukemia: Key Concepts and Emerging Controversies. Front. Oncol. 2020, 10. [Google Scholar] [CrossRef]
  194. Daver, N.; Schlenk, R.F.; Russell, N.H.; Levis, M.J. Targeting FLT3 mutations in AML: Review of current knowledge and evidence. Leukemia 2019, 33, 299–312. [Google Scholar] [CrossRef] [Green Version]
  195. Sanford, D.; Blum, W.G.; Ravandi, F.; Klisovic, R.B.; Borthakur, G.; Walker, A.R.; Garcia-Manero, G.; Marcucci, G.; Wierda, W.G.; Whitman, S.P.; et al. Efficacy and safety of an anti-FLT3 antibody (LY3012218) in patients with relapsed acute myeloid leukemia. J. Clin. Oncol. 2015, 33, 7059. [Google Scholar] [CrossRef]
  196. Piloto, O.; Griesemer, M.; Nguyen, B.; Li, L.; Li, Y.; Witte, L.; Hicklin, D.J.; Small, D. The Anti-FLT3 Monoclonal Antibody EB10 Is Cytotoxic to FLT3 Inhibitor Resistant Cells In Vivo. Blood 2005, 106, 1511. [Google Scholar] [CrossRef]
  197. Furusawa, Y.; Kaneko, M.K.; Kato, Y. Establishment of C20Mab-11, a novel anti-CD20 monoclonal antibody, for the detection of B cells. Oncol. Lett. 2020, 20, 1961–1967. [Google Scholar] [CrossRef]
  198. Stashenko, P.; Nadler, L.M.; Hardy, R.; Schlossman, S.F. Characterization of a human B lymphocyte-specific antigen. J. Immunol. 1980, 125, 1678–1685. [Google Scholar]
  199. Prevodnik, V.K.; Lavrenčak, J.; Horvat, M.; Novakovič, B.J. The predictive significance of CD20 expression in B-cell lymphomas. Diagn. Pathol. 2011, 6, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Liu, Q.; Weaver, L.S.; Liewehr, D.; Venzon, D.; Stetler-Stevenson, M.; Yuan, C.M. Increased expression of CD20 and CD45 and diminished expression of CD19 are features of follicular lymphoma. Pathol. Lab. Med. Int. 2013, 5, 21. [Google Scholar]
  201. Leonard, J.P.; Trneny, M.; Izutsu, K.; Fowler, N.H.; Hong, X.; Zhu, J.; Zhang, H.; Offner, F.; Scheliga, A.; Nowakowski, G.S.; et al. AUGMENT: A Phase III Study of Lenalidomide Plus Rituximab Versus Placebo Plus Rituximab in Relapsed or Refractory Indolent Lymphoma. J. Clin. Oncol. 2019, 37, 1188–1199. [Google Scholar] [CrossRef] [PubMed]
  202. Bräuner-Osborne, H.; Jensen, A.A.; Sheppard, P.O.; Brodin, B.; Krogsgaard-Larsen, P.; O’Hara, P. Cloning and characterization of a human orphan family C G-protein coupled receptor GPRC5D1GenBank accession Nos. for GPRC5C: AF207989, for Gprc5d: AF218809 and for GPRC5D: AF209923.1. Biochim. Et Biophys. Acta (BBA)-Gene Struct. Expr. 2001, 1518, 237–248. [Google Scholar] [CrossRef]
  203. Pillarisetti, K.; Edavettal, S.; Mendonça, M.; Li, Y.; Tornetta, M.; Babich, A.; Majewski, N.; Husovsky, M.; Reeves, D.; Walsh, E.; et al. A T-cell–redirecting bispecific G-protein–coupled receptor class 5 member D x CD3 antibody to treat multiple myeloma. Blood 2020, 135, 1232–1243. [Google Scholar] [CrossRef]
  204. Smith Eric, L.; Harrington, K.; Staehr, M.; Masakayan, R.; Jones, J.; Long Thomas, J.; Ng Khong, Y.; Ghoddusi, M.; Purdon Terence, J.; Wang, X.; et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. Sci. Transl. Med. 2019, 11, eaau7746. [Google Scholar] [CrossRef]
  205. Gao, Y.; Wang, X.; Yan, H.; Zeng, J.; Ma, S.; Niu, Y.; Zhou, G.; Jiang, Y.; Chen, Y. Comparative Transcriptome Analysis of Fetal Skin Reveals Key Genes Related to Hair Follicle Morphogenesis in Cashmere Goats. PLoS ONE 2016, 11, e0151118. [Google Scholar] [CrossRef] [Green Version]
  206. Suurs, F.V.; Lub-de Hooge, M.N.; de Vries, E.G.E.; de Groot, D.J.A. A review of bispecific antibodies and antibody constructs in oncology and clinical challenges. Pharmacol. Ther. 2019, 201, 103–119. [Google Scholar] [CrossRef]
  207. Ma, J.; Mo, Y.; Tang, M.; Shen, J.; Qi, Y.; Zhao, W.; Huang, Y.; Xu, Y.; Qian, C. Bispecific Antibodies: From Research to Clinical Application. Front. Immunol. 2021, 12, 626616. [Google Scholar] [CrossRef]
  208. Loffler, A.; Kufer, P.; Lutterbuse, R.; Zettl, F.; Daniel, P.T.; Schwenkenbecher, J.M.; Riethmuller, G.; Dorken, B.; Bargou, R.C. A recombinant bispecific single-chain antibody, CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes. Blood 2000, 95, 2098–2103. [Google Scholar] [CrossRef]
  209. Frankel, S.R.; Baeuerle, P.A. Targeting T cells to tumor cells using bispecific antibodies. Curr. Opin. Chem. Biol. 2013, 17, 385–392. [Google Scholar] [CrossRef] [PubMed]
  210. Wang, Q.; Chen, Y.; Park, J.; Liu, X.; Hu, Y.; Wang, T.; McFarland, K.; Betenbaugh, M.J. Design and production of bispecific antibodies. Antibodies 2019, 8, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Holliger, P.; Prospero, T.; Winter, G. “Diabodies”: Small bivalent and bispecific antibody fragments. Proc. Natl. Acad. Sci. USA 1993, 90, 6444–6448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Moore, P.A.; Zhang, W.; Rainey, G.J.; Burke, S.; Li, H.; Huang, L.; Gorlatov, S.; Veri, M.C.; Aggarwal, S.; Yang, Y.; et al. Application of dual affinity retargeting molecules to achieve optimal redirected T-cell killing of B-cell lymphoma. Blood 2011, 117, 4542–4551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Yang, F.; Wen, W.; Qin, W. Bispecific Antibodies as a Development Platform for New Concepts and Treatment Strategies. Int. J. Mol. Sci. 2016, 18, 48. [Google Scholar] [CrossRef] [Green Version]
  214. Rader, C. DARTs take aim at BiTEs. Blood 2011, 117, 4403–4404. [Google Scholar] [CrossRef]
  215. Reusch, U.; Burkhardt, C.; Fucek, I.; Le Gall, F.; Le Gall, M.; Hoffmann, K.; Knackmuss, S.H.; Kiprijanov, S.; Little, M.; Zhukovsky, E.A. A novel tetravalent bispecific TandAb (CD30/CD16A) efficiently recruits NK cells for the lysis of CD30+ tumor cells. mAbs 2014, 6, 728–739. [Google Scholar] [CrossRef] [Green Version]
  216. Reusch, U.; Duell, J.; Ellwanger, K.; Herbrecht, C.; Knackmuss, S.H.; Fucek, I.; Eser, M.; McAleese, F.; Molkenthin, V.; Gall, F.L.; et al. A tetravalent bispecific TandAb (CD19/CD3), AFM11, efficiently recruits T cells for the potent lysis of CD19(+) tumor cells. mAbs 2015, 7, 584–604. [Google Scholar] [CrossRef] [Green Version]
  217. Choe-Juliak, C.; Alexis, K.M.; Schwarz, S.; Garcia, L.; Sawas, A. A phase II open-label multicenter study to assess the efficacy and safety of AFM13 in patients with relapsed or refractory CD30-positive peripheral T-cell lymphoma or transformed mycosis fungoides: The REDIRECT study design and rationale. J. Clin. Oncol. 2020, 38, TPS3148. [Google Scholar] [CrossRef]
  218. Gong, S.; Wu, C. Generation of Fabs-in-tandem immunoglobulin molecules for dual-specific targeting. Methods 2019, 154, 87–92. [Google Scholar] [CrossRef]
  219. Gong, S.; Ren, F.; Wu, D.; Wu, X.; Wu, C. Fabs-in-tandem immunoglobulin is a novel and versatile bispecific design for engaging multiple therapeutic targets. mAbs 2017, 9, 1118–1128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. A Ph1/2 Study of EMB-06 in Participants with Relapsed or Refractory Myeloma. Available online: https://clinicaltrials.gov/ct2/show/NCT04735575 (accessed on 19 April 2022).
  221. Santich, B.H.; Park, J.A.; Tran, H.; Guo, H.F.; Huse, M.; Cheung, N.V. Interdomain spacing and spatial configuration drive the potency of IgG-[L]-scFv T cell bispecific antibodies. Sci. Transl. Med. 2020, 12, eaax1315. [Google Scholar] [CrossRef] [PubMed]
  222. Wu, Z.; Guo, H.-F.; Xu, H.; Cheung, N.-K.V. Development of a Tetravalent Anti-GPA33/Anti-CD3 Bispecific Antibody for Colorectal Cancers. Mol. Cancer Ther. 2018, 17, 2164–2175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Hoseini, S.S.; Guo, H.; Wu, Z.; Hatano, M.N.; Cheung, N.-K.V. A potent tetravalent T-cell–engaging bispecific antibody against CD33 in acute myeloid leukemia. Blood Adv. 2018, 2, 1250–1258. [Google Scholar] [CrossRef] [PubMed]
  224. Xu, H.; Cheng, M.; Guo, H.; Chen, Y.; Huse, M.; Cheung, N.K. Retargeting T cells to GD2 pentasaccharide on human tumors using Bispecific humanized antibody. Cancer Immunol. Res. 2015, 3, 266–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Zekri, L.; Vogt, F.; Osburg, L.; Müller, S.; Kauer, J.; Manz, T.; Pflügler, M.; Maurer, A.; Heitmann, J.S.; Hagelstein, I. An IgG-based bispecific antibody for improved dual targeting in PSMA-positive cancer. EMBO Mol. Med. 2021, 13, e11902. [Google Scholar] [CrossRef]
  226. Heitmann, J.S.; Pfluegler, M.; Jung, G.; Salih, H.R. Bispecific Antibodies in Prostate Cancer Therapy: Current Status and Perspectives. Cancers 2021, 13, 549. [Google Scholar] [CrossRef]
  227. Heitmann, J.S.; Walz, J.S.; Pflügler, M.; Kauer, J.; Schlenk, R.F.; Jung, G.; Salih, H.R. Protocol of a prospective, multicentre phase I study to evaluate the safety, tolerability and preliminary efficacy of the bispecific PSMAxCD3 antibody CC-1 in patients with castration-resistant prostate carcinoma. BMJ Open 2020, 10, e039639. [Google Scholar] [CrossRef]
  228. Uckun, F.M.; Lin, T.L.; Mims, A.S.; Patel, P.; Lee, C.; Shahidzadeh, A.; Shami, P.J.; Cull, E.; Cogle, C.R.; Watts, J. A Clinical Phase 1B Study of the CD3xCD123 Bispecific Antibody APVO436 in Patients with Relapsed/Refractory Acute Myeloid Leukemia or Myelodysplastic Syndrome. Cancers 2021, 13, 4113. [Google Scholar] [CrossRef]
  229. Watts, J.M.; Lin, T.; Wang, E.S.; Mims, A.S.; Cull, E.H.; Patel, P.A.; Shami, P.J.; Walter, R.B.; Cogle, C.R.; Chenault, R.A.; et al. Preliminary Results from a Phase 1 Study of APVO436, a Novel Anti-CD123 x Anti-CD3 Bispecific Molecule, in Relapsed/Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome. Blood 2020, 136, 11–12. [Google Scholar] [CrossRef]
  230. Han, L.; Chen, J.; Ding, K.; Zong, H.; Xie, Y.; Jiang, H.; Zhang, B.; Lu, H.; Yin, W.; Gilly, J.; et al. Efficient generation of bispecific IgG antibodies by split intein mediated protein trans-splicing system. Sci. Rep. 2017, 7, 8360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Duivelshof, B.L.; Beck, A.; Guillarme, D.; D’Atri, V. Bispecific antibody characterization by a combination of intact and site-specific/chain-specific LC/MS techniques. Talanta 2022, 236, 122836. [Google Scholar] [CrossRef] [PubMed]
  232. Labrijn, A.F.; Meesters, J.I.; de Goeij, B.E.; van den Bremer, E.T.; Neijssen, J.; van Kampen, M.D.; Strumane, K.; Verploegen, S.; Kundu, A.; Gramer, M.J.; et al. Efficient generation of stable bispecific IgG1 by controlled Fab-arm exchange. Proc. Natl. Acad. Sci. USA 2013, 110, 5145–5150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Suresh, M.R.; Cuello, A.C.; Milstein, C. Bispecific monoclonal antibodies from hybrid hybridomas. Methods Enzymol. 1986, 121, 210–228. [Google Scholar] [CrossRef]
  234. Shuptrine, C.W.; Surana, R.; Weiner, L.M. Monoclonal antibodies for the treatment of cancer. Semin. Cancer Biol. 2012, 22, 3–13. [Google Scholar] [CrossRef] [Green Version]
  235. Chelius, D.; Ruf, P.; Gruber, P.; Ploscher, M.; Liedtke, R.; Gansberger, E.; Hess, J.; Wasiliu, M.; Lindhofer, H. Structural and functional characterization of the trifunctional antibody catumaxomab. mAbs 2010, 2, 309–319. [Google Scholar] [CrossRef] [Green Version]
  236. Lindhofer, H.; Mocikat, R.; Steipe, B.; Thierfelder, S. Preferential species-restricted heavy/light chain pairing in rat/mouse quadromas. Implications for a single-step purification of bispecific antibodies. J. Immunol. 1995, 155, 219–225. [Google Scholar]
  237. Viardot, A.; Bargou, R. Bispecific antibodies in haematological malignancies. Cancer Treat. Rev. 2018, 65, 87–95. [Google Scholar] [CrossRef]
  238. Merchant, A.M.; Zhu, Z.; Yuan, J.Q.; Goddard, A.; Adams, C.W.; Presta, L.G.; Carter, P. An efficient route to human bispecific IgG. Nat. Biotechnol. 1998, 16, 677–681. [Google Scholar] [CrossRef]
  239. Carter, P. Bispecific human IgG by design. J. Immunol. Methods 2001, 248, 7–15. [Google Scholar] [CrossRef]
  240. Xu, Y.; Lee, J.; Tran, C.; Heibeck, T.H.; Wang, W.D.; Yang, J.; Stafford, R.L.; Steiner, A.R.; Sato, A.K.; Hallam, T.J.; et al. Production of bispecific antibodies in “knobs-into-holes” using a cell-free expression system. mAbs 2015, 7, 231–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Klein, C.; Sustmann, C.; Thomas, M.; Stubenrauch, K.; Croasdale, R.; Schanzer, J.; Brinkmann, U.; Kettenberger, H.; Regula, J.T.; Schaefer, W. Progress in overcoming the chain association issue in bispecific heterodimeric IgG antibodies. mAbs 2012, 4, 653–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Shiraiwa, H.; Narita, A.; Kamata-Sakurai, M.; Ishiguro, T.; Sano, Y.; Hironiwa, N.; Tsushima, T.; Segawa, H.; Tsunenari, T.; Ikeda, Y.; et al. Engineering a bispecific antibody with a common light chain: Identification and optimization of an anti-CD3 epsilon and anti-GPC3 bispecific antibody, ERY974. Methods 2019, 154, 10–20. [Google Scholar] [CrossRef] [PubMed]
  243. Ishiguro, T.; Sano, Y.; Komatsu, S.-I.; Kamata-Sakurai, M.; Kaneko, A.; Kinoshita, Y.; Shiraiwa, H.; Azuma, Y.; Tsunenari, T.; Kayukawa, Y. An anti–glypican 3/CD3 bispecific T cell–redirecting antibody for treatment of solid tumors. Sci. Transl. Med. 2017, 9, eaal4291. [Google Scholar] [CrossRef] [Green Version]
  244. Gunasekaran, K.; Pentony, M.; Shen, M.; Garrett, L.; Forte, C.; Woodward, A.; Ng, S.B.; Born, T.; Retter, M.; Manchulenko, K. Enhancing antibody Fc heterodimer formation through electrostatic steering effects: Applications to bispecific molecules and monovalent IgG. J. Biol. Chem. 2010, 285, 19637–19646. [Google Scholar] [CrossRef] [Green Version]
  245. Van der Neut Kolfschoten, M.; Schuurman, J.; Losen, M.; Bleeker Wim, K.; Martínez-Martínez, P.; Vermeulen, E.; den Bleker Tamara, H.; Wiegman, L.; Vink, T.; Aarden Lucien, A.; et al. Anti-Inflammatory Activity of Human IgG4 Antibodies by Dynamic Fab Arm Exchange. Science 2007, 317, 1554–1557. [Google Scholar] [CrossRef] [Green Version]
  246. Van den Bremer, E.T.J.; Labrijn, A.F.; van den Boogaard, R.; Priem, P.; Scheffler, K.; Melis, J.P.M.; Schuurman, J.; Parren, P.W.H.I.; de Jong, R.N. Cysteine-SILAC Mass Spectrometry Enabling the Identification and Quantitation of Scrambled Interchain Disulfide Bonds: Preservation of Native Heavy-Light Chain Pairing in Bispecific IgGs Generated by Controlled Fab-arm Exchange. Anal. Chem. 2017, 89, 10873–10882. [Google Scholar] [CrossRef]
  247. Engelberts, P.J.; Hiemstra, I.H.; de Jong, B.; Schuurhuis, D.H.; Meesters, J.; Beltran Hernandez, I.; Oostindie, S.C.; Neijssen, J.; van den Brink, E.N.; Horbach, G.J.; et al. DuoBody-CD3xCD20 induces potent T-cell-mediated killing of malignant B cells in preclinical models and provides opportunities for subcutaneous dosing. EbioMedicine 2020, 52, 102625. [Google Scholar] [CrossRef]
  248. Syed, Y.Y. Amivantamab: First Approval. Drugs 2021, 81, 1349–1353. [Google Scholar] [CrossRef]
  249. Brinkmann, U.; Kontermann, R.E. The making of bispecific antibodies. mAbs 2017, 9, 182–212. [Google Scholar] [CrossRef]
  250. Kim, T.M.; Alonso, A.; Prince, M.; Taszner, M.; Cho, S.-G.; Stevens, D.A.; Poon, M.; Lim, F.; Le Gouill, S.; Carpio, C.; et al. A Phase 2 Study of Odronextamab (REGN1979), a CD20 x CD3 Bispecific Antibody, in Patients with Relapsed/Refractory B-Cell Non-Hodgkin Lymphoma. Blood 2020, 136, 28–29. [Google Scholar] [CrossRef]
  251. Surowka, M.; Schaefer, W.; Klein, C. Ten years in the making: Application of CrossMab technology for the development of therapeutic bispecific antibodies and antibody fusion proteins. mAbs 2021, 13, 1967714. [Google Scholar] [CrossRef] [PubMed]
  252. Hertzberg, M.; Ku, M.; Catalani, O.; Althaus, B.; Simko, S.; Gregory, G.P. A phase III trial evaluating glofitamab in combination with gemcitabine plus oxaliplatin versus rituximab in combination with gemcitabine and oxaliplatin in patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL). J. Clin. Oncol. 2021, 39, TPS7575. [Google Scholar] [CrossRef]
  253. Li, Y. IgG-like bispecific antibody platforms with built-in purification-facilitating elements. Protein Expr. Purif. 2021, 188, 105955. [Google Scholar] [CrossRef] [PubMed]
  254. Ha, J.-H.; Kim, J.-E.; Kim, Y.-S. Immunoglobulin Fc Heterodimer Platform Technology: From Design to Applications in Therapeutic Antibodies and Proteins. Front. Immunol. 2016, 7, 394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Zhang, J.; Yi, J.; Zhou, P. Development of bispecific antibodies in China: Overview and prospects. Antib. Ther. 2020, 3, 126–145. [Google Scholar] [CrossRef]
  256. Safety and Efficacy of XmAb18087 ± Pembrolizumab in Advanced Merkel Cell Carcinoma or Extensive-Stage Small Cell Lung Cancer. Available online: https://clinicaltrials.gov/ct2/show/NCT04590781?term=NCT04590781&draw=2&rank=1 (accessed on 15 April 2022).
  257. You, G.; Won, J.; Lee, Y.; Moon, D.; Park, Y.; Lee, S.H.; Lee, S.-W. Bispecific Antibodies: A Smart Arsenal for Cancer Immunotherapies. Vaccines 2021, 9, 724. [Google Scholar] [CrossRef]
  258. Voynov, V.; Adam, P.J.; Nixon, A.E.; Scheer, J.M. Discovery Strategies to Maximize the Clinical Potential of T-Cell Engaging Antibodies for the Treatment of Solid Tumors. Antibodies 2020, 9, 65. [Google Scholar] [CrossRef]
  259. Moore, P.A.; Shah, K.; Yang, Y.; Alderson, R.; Roberts, P.; Long, V.; Liu, D.; Li, J.C.; Burke, S.; Ciccarone, V. Development of MGD007, a gpA33 x CD3-bispecific DART protein for T-cell immunotherapy of metastatic colorectal cancer. Mol. Cancer Ther. 2018, 17, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
  260. Tabernero, J.; Melero, I.; Ros, W.; Argiles, G.; Marabelle, A.; Rodriguez-Ruiz, M.E.; Albanell, J.; Calvo, E.; Moreno, V.; Cleary, J.M. Phase Ia and Ib studies of the novel carcinoembryonic antigen (CEA) T-cell bispecific (CEA CD3 TCB) antibody as a single agent and in combination with atezolizumab: Preliminary efficacy and safety in patients with metastatic colorectal cancer (mCRC). J. Clin. Oncol. 2017, 35, 3002. [Google Scholar] [CrossRef]
  261. Bailis, J.M.; Lutterbuese, P.; Thomas, O.; Locher, K.; Harrold, J.; Boyle, M.; Wahl, J.; Li, S.; Sternjak, A.; Henn, A. Preclinical evaluation of BiTE® immune therapy targeting MUC17 or CLDN18. 2 for gastric cancer. Cancer Res. 2020, 80, 3364. [Google Scholar]
  262. Lordick, F.; Chao, J.; Buxò, E.; van Laarhoven, H.; Lima, C.; Lorenzen, S.; Dayyani, F.; Heinemann, V.; Greil, R.; Stienen, S. 1496TiP Phase I study evaluating safety and tolerability of AMG 910, a half-life extended bispecific T cell engager targeting claudin-18.2 (CLDN18. 2) in gastric and gastroesophageal junction (G/GEJ) adenocarcinoma. Ann. Oncol. 2020, 31, S928–S929. [Google Scholar] [CrossRef]
  263. Owonikoko, T.K.; Champiat, S.; Johnson, M.L.; Govindan, R.; Izumi, H.; Lai, W.V.V.; Borghaei, H.; Boyer, M.J.; Boosman, R.J.; Hummel, H.-D. Updated results from a phase 1 study of AMG 757, a half-life extended bispecific T-cell engager (BiTE) immuno-oncology therapy against delta-like ligand 3 (DLL3), in small cell lung cancer (SCLC). J. Clin. Oncol. 2021, 39, 8510. [Google Scholar] [CrossRef]
  264. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727. [Google Scholar]
  265. Lakhani, N.; Johnson, M.; Groisberg, R.; Han, H.; Casey, K.; Li, S.; Skokos, D.; Seebach, F.; Lowy, I.; Fury, M. 535 A phase I/II study of REGN7075 (EGFRxCD28 costimulatory bispecific antibody) in combination with cemiplimab (anti–PD-1) in patients with advanced solid tumors. J. Immunother. Cancer 2021, 9, A565. [Google Scholar] [CrossRef]
  266. Heiss, M.M.; Murawa, P.; Koralewski, P.; Kutarska, E.; Kolesnik, O.O.; Ivanchenko, V.V.; Dudnichenko, A.S.; Aleknaviciene, B.; Razbadauskas, A.; Gore, M. The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: Results of a prospective randomized phase II/III trial. Int. J. Cancer 2010, 127, 2209–2221. [Google Scholar] [CrossRef]
  267. Xu, J.; Zhao, C.; Jia, Y.; Wang, S.; Ma, X.; Wang, T.; Huang, S.; Pei, M.; Wang, X.; Zhou, P. 539P Interim results of a phase I study of M701, a recombinant anti-EpCAM and anti-CD3 bispecific antibody in EpCAM-positive cancer patients with malignant ascites. Annals of Oncology 2021, 32, S603. [Google Scholar] [CrossRef]
  268. Yankelevich, M.; Kondadasula, S.V.; Thakur, A.; Buck, S.; Cheung, N.-K.V.; Lum, L.G. Anti-CD3 × anti-GD2 bispecific antibody redirects T-cell cytolytic activity to neuroblastoma targets. Pediatric Blood Cancer 2012, 59, 1198–1205. [Google Scholar] [CrossRef] [Green Version]
  269. Elshiaty, M.; Schindler, H.; Christopoulos, P. Principles and Current Clinical Landscape of Multispecific Antibodies against Cancer. Int. J. Mol. Sci. 2021, 22, 5632. [Google Scholar] [CrossRef]
  270. Hurwitz, H.; Crocenzi, T.; Lohr, J.; Bonvini, E.; Johnson, S.; Moore, P.; Wigginton, J. A Phase I, first-in-human, open label, dose escalation study of MGD007, a humanized gpA33× CD3 dual-affinity re-targeting (DART®) protein in patients with relapsed/refractory metastatic colorectal carcinoma. J. Immunother. Cancer 2014, 2, 86. [Google Scholar] [CrossRef] [Green Version]
  271. Singh, A.; Dees, S.; Grewal, I. Overcoming the challenges associated with CD3+ T-cell redirection in cancer. Br. J. Cancer 2021, 124, 1037–1048. [Google Scholar] [CrossRef] [PubMed]
  272. Ogita, Y.; Weiss, D.; Sugaya, N.; Nakamura, M.; Ito, H.; Ishiguro, T.; Shimada, S.; Ueda, M.; Matsushima, J.; Gianella-Borradori, A. A phase 1 dose escalation (DE) and cohort expansion (CE) study of ERY974, an anti-Glypican 3 (GPC3)/CD3 bispecific antibody, in patients with advanced solid tumors. J. Clin. Oncol. 2018, 36, TPS2599. [Google Scholar] [CrossRef]
  273. A Phase I Study of ERY974 in Patients With Hepatocellular Carcinoma. Available online: https://clinicaltrials.gov/ct2/show/NCT05022927?term=NCT05022927&draw=2&rank=1 (accessed on 15 April 2022).
  274. Yu, S.; Zhang, J.; Yan, Y.; Yao, X.; Fang, L.; Xiong, H.; Liu, Y.; Chu, Q.; Zhou, P.; Wu, K.; et al. A novel asymmetrical anti-HER2/CD3 bispecific antibody exhibits potent cytotoxicity for HER2-positive tumor cells. J. Exp. Clin. Cancer Res. 2019, 38, 355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Study of REGN4018 Administered Alone or in Combination with Cemiplimab in Adult Patients with Recurrent Ovarian Cancer. Available online: https://clinicaltrials.gov/ct2/show/NCT03564340?term=NCT03564340&draw=2&rank=1 (accessed on 18 February 2022).
  276. Einsele, H.; Borghaei, H.; Orlowski, R.Z.; Subklewe, M.; Roboz, G.J.; Zugmaier, G.; Kufer, P.; Iskander, K.; Kantarjian, H.M.J.C. The BiTE (bispecific T-cell engager) platform: Development and future potential of a targeted immuno-oncology therapy across tumor types. Cancer 2020, 126, 3192–3201. [Google Scholar] [CrossRef]
  277. Subudhi, S.K.; Siddiqui, B.A.; Maly, J.J.; Nandagopal, L.; Lam, E.T.; Whang, Y.E.; Minocha, M.; Gupta, V.; Penny, X.; Cooner, F. Safety and efficacy of AMG 160, a half-life extended BiTE immune therapy targeting prostate-specific membrane antigen (PSMA), and other therapies for metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2021, 39, TPS5088. [Google Scholar] [CrossRef]
  278. Tran, B.; Horvath, L.; Dorff, T.B.; Greil, R.; Machiels, J.-P.H.; Roncolato, F.; Autio, K.A.; Rettig, M.; Fizazi, K.; Lolkema, M.P. Phase I study of AMG 160, a half-life extended bispecific T-cell engager (HLE BiTE) immune therapy targeting prostate-specific membrane antigen (PSMA), in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2020, 38, TPS261. [Google Scholar] [CrossRef]
  279. Buelow, B.; Dalvi, P.; Dang, K.; Patel, A.; Johal, K.; Pham, D.; Panchal, S.; Liu, Y.; Fong, L.; Sartor, A.O. TNB585. 001: A multicenter, phase 1, open-label, dose-escalation and expansion study of tnb-585, a bispecific T-cell engager targeting PSMA in subjects with metastatic castrate resistant prostate cancer. J. Clin. Oncol. 2021, 39, TPS5092. [Google Scholar] [CrossRef]
  280. Nolan-Stevaux, O. Abstract DDT02-03: AMG 509: A novel, humanized, half-Life extended, bispecific STEAP1 × CD3 T cell recruiting XmAb® 2+1 antibody. Cancer Res. 2020, 80, DDT02–DDT03. [Google Scholar] [CrossRef]
  281. Lee, S.-H.; Chu, S.Y.; Rashid, R.; Phung, S.; Leung, I.W.; Muchhal, U.S.; Moore, G.L.; Bernett, M.J.; Schubbert, S.; Ardila, C.J.C.R. Anti-SSTR2× anti-CD3 bispecific antibody induces potent killing of human tumor cells in vitro and in mice, and stimulates target-dependent T cell activation in monkeys: A potential immunotherapy for neuroendocrine tumors. Cancer Res. 2017, 77, 3633. [Google Scholar]
  282. Pillarisetti, K.; Powers, G.; Luistro, L.; Babich, A.; Baldwin, E.; Li, Y.; Zhang, X.; Mendonça, M.; Majewski, N.; Nanjunda, R.; et al. Teclistamab is an active T cell–redirecting bispecific antibody against B-cell maturation antigen for multiple myeloma. Blood Adv. 2020, 4, 4538–4549. [Google Scholar] [CrossRef]
  283. Krishnan, A.Y.; Garfall, A.L.; Mateos, M.-V.; van de Donk, N.W.C.J.; Nahi, H.; San-Miguel, J.; Oriol, A.; Rosiñol, L.; Chari, A.; Bhutani, M. Updated phase 1 results of teclistamab, a B-cell maturation antigen (BCMA)× CD3 bispecific antibody, in relapsed/refractory multiple myeloma (MM). Blood 2021, 136, 27. [Google Scholar] [CrossRef]
  284. A Study of Teclistamab in Combination With Daratumumab Subcutaneously (SC) (Tec-Dara) Versus Daratumumab SC, Pomalidomide, and Dexamethasone (DPd) or Daratumumab SC, Bortezomib, and Dexamethasone (DVd) in Participants With Relapsed or Refractory Multiple Myeloma (MajesTEC-3). Available online: https://clinicaltrials.gov/ct2/show/NCT05083169?term=NCT05083169&draw=2&rank=1 (accessed on 18 February 2022).
  285. Caraccio, C.; Krishna, S.; Phillips, D.J.; Schürch, C.M. Bispecific Antibodies for Multiple Myeloma: A Review of Targets, Drugs, Clinical Trials, and Future Directions. Front. Immunol. 2020, 11, 501. [Google Scholar] [CrossRef] [PubMed]
  286. Burt, R.; Warcel, D.; Fielding, A.K. Blinatumomab, a bispecific B-cell and T-cell engaging antibody, in the treatment of B-cell malignancies. Hum. Vaccines Immunother. 2019, 15, 594–602. [Google Scholar] [CrossRef] [PubMed]
  287. Viardot, A.; Hess, G.; Bargou, R.C.; Morley, N.; Gritti, G.; Iskander, K.; Cohan, D.; Zhang, A.; Franklin, J.; Coyle, L. Durability of complete response after blinatumomab therapy for refractory/relapsed aggressive B-cell non-Hodgkin lymphoma. J. Clin. Oncol. 2019, 37, e19041. [Google Scholar] [CrossRef]
  288. Study to Evaluate Safety and Efficacy of Blinatumomab in Subjects with Relapsed/Refractory (R/R) Aggressive B-Cell NHL. Available online: https://clinicaltrials.gov/ct2/show/NCT02910063 (accessed on 15 April 2022).
  289. Katz, D.A.; Morris, J.D.; Chu, M.P.; David, K.A.; Thieblemont, C.; Morley, N.J.; Khan, S.S.; Viardot, A.; Martín García-Sancho, A.; Rodríguez-García, G.; et al. Open-label, phase 2 study of blinatumomab after frontline R-chemotherapy in adults with newly diagnosed, high-risk DLBCL. Leuk. Lymphoma 2022, 1–11. [Google Scholar] [CrossRef]
  290. A Study of TNB-486 in Subjects With Relapsed or Refractory B-Cell Non-Hodgkin Lymphoma. Available online: https://clinicaltrials.gov/ct2/show/NCT04594642?term=NCT04594642&draw=2&rank=1 (accessed on 18 February 2022).
  291. Schuster, S.J. Bispecific antibodies for the treatment of lymphomas: Promises and challenges. Hematol. Oncol. 2021, 39, 113–116. [Google Scholar] [CrossRef]
  292. Budde, L.E.; Sehn, L.H.; Matasar, M.J.; Schuster, S.J.; Assouline, S.; Giri, P.; Kuruvilla, J.; Canales, M.; Dietrich, S.; Fay, K.; et al. Mosunetuzumab Monotherapy Is an Effective and Well-Tolerated Treatment Option for Patients with Relapsed/Refractory (R/R) Follicular Lymphoma (FL) Who Have Received ≥2 Prior Lines of Therapy: Pivotal Results from a Phase I/II Study. Blood 2021, 138, 127. [Google Scholar] [CrossRef]
  293. Salvaris, R.; Ong, J.; Gregory, G.P. Bispecific Antibodies: A Review of Development, Clinical Efficacy and Toxicity in B-Cell Lymphomas. J. Pers. Med. 2021, 11, 355. [Google Scholar] [CrossRef]
  294. Patel, K.; Michot, J.-M.; Chanan-Khan, A.; Ghesquieres, H.; Bouabdallah, K.; Byrd, J.C.; Cartron, G.; Portell, C.A.; Solh, M.; Tilly, H.; et al. Safety and Anti-Tumor Activity of Plamotamab (XmAb13676), an Anti-CD20 x Anti-CD3 Bispecific Antibody, in Subjects with Relapsed/Refractory Non-Hodgkin’s Lymphoma. Blood 2021, 138, 2494. [Google Scholar] [CrossRef]
  295. A Study of JNJ-75348780 in Participants With Non-Hodgkin Lymphoma (NHL) and Chronic Lymphocytic Leukemia (CLL). Available online: https://clinicaltrials.gov/ct2/show/NCT04540796?term=NCT04540796&draw=2&rank=1 (accessed on 19 February 2022).
  296. Wu, J.; Fu, J.; Zhang, M.; Liu, D. AFM13: A first-in-class tetravalent bispecific anti-CD30/CD16A antibody for NK cell-mediated immunotherapy. J. Hematol. Oncol. 2015, 8, 96. [Google Scholar] [CrossRef] [Green Version]
  297. Affimed Announces 100% Objective Response Rate at Highest Dose in Phase 1-2 Study of Cord Blood-Derived Natural Killer Cells Pre-Complexed with Innate Cell Engager AFM13 for CD30-Positive Lyphomas. Available online: https://www.affimed.com/affimed-announces-100-objective-response-rate-at-highest-dose-in-phase-1-2-study-of-cord-blood-derived-natural-killer-cells-pre-complexed-with-innate-cell-engager-afm13-for-cd30-positive-lymphomas/ (accessed on 19 February 2022).
  298. Ravandi, F.; Stein, A.S.; Kantarjian, H.M.; Walter, R.B.; Paschka, P.; Jongen-Lavrencic, M.; Ossenkoppele, G.J.; Yang, Z.; Mehta, B.; Subklewe, M. A phase 1 first-in-human study of AMG 330, an anti-CD33 bispecific T-cell engager (BiTE®) antibody construct, in relapsed/refractory acute myeloid leukemia (R/R AML). Blood 2018, 132, 25. [Google Scholar] [CrossRef]
  299. Nair-Gupta, P.; Diem, M.; Reeves, D.; Wang, W.; Schulingkamp, R.; Sproesser, K.; Mattson, B.; Heidrich, B.; Mendonça, M.; Joseph, J.; et al. A novel C2 domain binding CD33xCD3 bispecific antibody with potent T-cell redirection activity against acute myeloid leukemia. Blood Adv. 2020, 4, 906–919. [Google Scholar] [CrossRef] [PubMed]
  300. Richter, J.R.; Landgren, C.O.; Kauh, J.S.; Back, J.; Salhi, Y.; Reddy, V.; Bayever, E.; Berdeja, J.G. Phase 1, multicenter, open-label study of single-agent bispecific antibody t-cell engager GBR 1342 in relapsed/refractory multiple myeloma. J. Clin. Oncol. 2018, 36, TPS3132. [Google Scholar] [CrossRef]
  301. Simoneaux, R. Positive Results for Bispecific DART Flotetuzumab in Patients With AML. Oncol. Times 2021, 43, 15. [Google Scholar] [CrossRef]
  302. Uy, G.L.; Aldoss, I.; Foster, M.C.; Sayre, P.H.; Wieduwilt, M.J.; Advani, A.S.; Godwin, J.E.; Arellano, M.L.; Sweet, K.L.; Emadi, A.; et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood 2021, 137, 751–762. [Google Scholar] [CrossRef]
  303. Kaplon, H.; Reichert, J.M. Antibodies to watch in 2021. mAbs 2021, 12, 1860476. [Google Scholar] [CrossRef]
  304. Slade, M.J.; Uy, G.L. CD123 bi-specific antibodies in development in AML: What do we know so far? Best Pract. Res. Clin. Haematol. 2020, 33, 101219. [Google Scholar] [CrossRef]
  305. Van Loo, P.F.; Hangalapura, B.N.; Thordardottir, S.; Gibbins, J.D.; Veninga, H.; Hendriks, L.J.A.; Kramer, A.; Roovers, R.C.; Leenders, M.; de Kruif, J.; et al. MCLA-117, a CLEC12AxCD3 bispecific antibody targeting a leukaemic stem cell antigen, induces T cell-mediated AML blast lysis. Expert Opin. Biol. Ther. 2019, 19, 721–733. [Google Scholar] [CrossRef]
  306. Mascarenhas, J.; Huls, G.; Venditti, A.; Breems, D.; De Botton, S. Update from the ongoing phase I multinational study of MCLA-117, a bispecific CLEC12A x CD3 T-cell engager, in patients (pts) with acute myelogenous leukemia (AML). Eur. Hematol. Assoc. 2020. [Google Scholar]
  307. Mehta, N.K.; Pfluegler, M.; Meetze, K.; Li, B.; Sindel, I.; Vogt, F.; Marklin, M.; Heitmann, J.S.; Kauer, J.; Osburg, L.; et al. A novel IgG-based FLT3xCD3 bispecific antibody for the treatment of AML and B-ALL. J. Immunother. Cancer 2022, 10, e003882. [Google Scholar] [CrossRef]
  308. Meetze, K.; Mehta, N.K.; Pfluegler, M.; Li, B.; Baeuerle, P.A.; Michaelson, J.S.; Jung, G.; Salih, H. CLN-049 is a bispecific T cell engaging IgG-like antibody targeting FLT3 on AML cells [abstract]. In Proceedings of the 113th Annual Meeting of the American Association for Cancer Research, New Orleans, LA, USA, 8–13 April 2021. [Google Scholar]
  309. CLN-049 in Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML). Available online: https://clinicaltrials.gov/ct2/show/NCT05143996 (accessed on 14 April 2022).
  310. Safety, Tolerability, PK, PD, and Efficacy of AMG 427 in Subjects with Relapsed/Refractory Acute Myeloid Leukemia (20170528). Available online: https://clinicaltrials.gov/ct2/show/NCT03541369 (accessed on 19 April 2022).
  311. Berdeja, J.G.; Krishnan, A.Y.; Oriol, A.; van de Donk, N.W.C.J.; Rodríguez-Otero, P.; Askari, E.; Mateos, M.-V.; Minnema, M.C.; Costa, L.J.; Verona, R.; et al. Updated results of a phase 1, first-in-human study of talquetamab, a G protein-coupled receptor family C group 5 member D (GPRC5D) × CD3 bispecific antibody, in relapsed/refractory multiple myeloma (MM). J. Clin. Oncol. 2021, 39, 8008. [Google Scholar] [CrossRef]
  312. A Study of Talquetamab in Participants With Relapsed or Refractory Multiple Myeloma. Available online: https://clinicaltrials.gov/ct2/show/NCT04634552?term=NCT04634552&draw=2&rank=1 (accessed on 18 February 2022).
  313. Lorenczewski, G.; Friedrich, M.; Kischel, R.; Dahlhoff, C.; Anlahr, J.; Balazs, M.; Rock, D.; Boyle, M.C.; Goldstein, R.; Coxon, A. Generation of a half-life extended anti-CD19 BiTE® antibody construct compatible with once-weekly dosing for treatment of CD19-positive malignancies. Blood 2017, 130, 2815. [Google Scholar]
  314. Borlak, J.; Länger, F.; Spanel, R.; Schöndorfer, G.; Dittrich, C. Immune-mediated liver injury of the cancer therapeutic antibody catumaxomab targeting EpCAM, CD3 and Fcγ receptors. Oncotarget 2016, 7, 28059. [Google Scholar] [CrossRef] [PubMed]
  315. Leong, S.R.; Sukumaran, S.; Hristopoulos, M.; Totpal, K.; Stainton, S.; Lu, E.; Wong, A.; Tam, L.; Newman, R.; Vuillemenot, B.R.; et al. An anti-CD3/anti–CLL-1 bispecific antibody for the treatment of acute myeloid leukemia. Blood 2017, 129, 609–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  316. Mandikian, D.; Takahashi, N.; Lo, A.A.; Li, J.; Eastham-Anderson, J.; Slaga, D.; Ho, J.; Hristopoulos, M.; Clark, R.; Totpal, K. Relative target affinities of T-cell–dependent bispecific antibodies determine biodistribution in a solid tumor mouse model. Mol. Cancer Ther. 2018, 17, 776–785. [Google Scholar] [CrossRef] [Green Version]
  317. Nimmerjahn, F.; Ravetch, J.V. Fcγ receptors as regulators of immune responses. Nat. Rev. Immunol. 2008, 8, 34–47. [Google Scholar] [CrossRef]
  318. Schlothauer, T.; Herter, S.; Koller, C.F.; Grau-Richards, S.; Steinhart, V.; Spick, C.; Kubbies, M.; Klein, C.; Umaña, P.; Mössner, E. Novel human IgG1 and IgG4 Fc-engineered antibodies with completely abolished immune effector functions. Protein Eng. Des. Sel. 2016, 29, 457–466. [Google Scholar] [CrossRef] [Green Version]
  319. Moore, G.L.; Bernett, M.J.; Rashid, R.; Pong, E.W.; Nguyen, D.-H.T.; Jacinto, J.; Eivazi, A.; Nisthal, A.; Diaz, J.E.; Chu, S.Y. A robust heterodimeric Fc platform engineered for efficient development of bispecific antibodies of multiple formats. Methods 2019, 154, 38–50. [Google Scholar] [CrossRef]
Figure 1. Action mechanism of bispecific antibody (bsAb)-based immune-cell engagers (ICEs) in cancers. The schematic drawing represents bsAb-based ICEs, including T- and natural killer-cell engagers, that bind simultaneously to tumor-associated antigens on cancer cells and specific antigens, such as CD3, CD28, and CD16 on immune cells. These interactions result in the formation of an immune synapse and the activation of immune cells that release cytokines, perforins, and granzymes to induce the cytotoxic effects on cancer cells.
Figure 1. Action mechanism of bispecific antibody (bsAb)-based immune-cell engagers (ICEs) in cancers. The schematic drawing represents bsAb-based ICEs, including T- and natural killer-cell engagers, that bind simultaneously to tumor-associated antigens on cancer cells and specific antigens, such as CD3, CD28, and CD16 on immune cells. These interactions result in the formation of an immune synapse and the activation of immune cells that release cytokines, perforins, and granzymes to induce the cytotoxic effects on cancer cells.
Ijms 23 05686 g001
Figure 2. Known and emerging targets for immune-cell engager (ICE) therapy against cancers. The schematic representation shows known and emerging therapeutic targets of bispecific antibody-based ICEs in solid (red background) and hematological (blue background) cancers. All therapeutic targets listed in this figure are grouped on the basis of their relationship with the bilayer and transmembrane topology.
Figure 2. Known and emerging targets for immune-cell engager (ICE) therapy against cancers. The schematic representation shows known and emerging therapeutic targets of bispecific antibody-based ICEs in solid (red background) and hematological (blue background) cancers. All therapeutic targets listed in this figure are grouped on the basis of their relationship with the bilayer and transmembrane topology.
Ijms 23 05686 g002
Figure 3. Immune-cell engager (ICE) structures in clinical studies, or ICEs approved by the United States Food and Drug Administration (US FDA) and/or European Medicines Agency (EMA). The schematic drawing depicts the structures of bispecific antibody (bsAb)-based ICEs that are currently evaluated in clinical studies or that have been approved by the US FDA and/or EMA. The structures of bsAb-based ICEs are subdivided into three major classes: variable fragments -based, immunoglobulin G (IgG)-based symmetric, and IgG-based asymmetric ICEs. Variable heavy-chain domains (VHs) of two different antibodies, designated as antibodies A, B, or C, are shown in dark blue, red, or pink, respectively. The variable light-chain domains (VLs) are shown in light blue, red, and pink, respectively. Moreover, rat or mouse antibody is depicted in purple or green, respectively.
Figure 3. Immune-cell engager (ICE) structures in clinical studies, or ICEs approved by the United States Food and Drug Administration (US FDA) and/or European Medicines Agency (EMA). The schematic drawing depicts the structures of bispecific antibody (bsAb)-based ICEs that are currently evaluated in clinical studies or that have been approved by the US FDA and/or EMA. The structures of bsAb-based ICEs are subdivided into three major classes: variable fragments -based, immunoglobulin G (IgG)-based symmetric, and IgG-based asymmetric ICEs. Variable heavy-chain domains (VHs) of two different antibodies, designated as antibodies A, B, or C, are shown in dark blue, red, or pink, respectively. The variable light-chain domains (VLs) are shown in light blue, red, and pink, respectively. Moreover, rat or mouse antibody is depicted in purple or green, respectively.
Ijms 23 05686 g003
Table 1. Current statuses of bispecific antibody (bsAb)-based immune-cell engagers (ICEs) in clinical studies or bsAb-based ICEs approved by the United States Food and Drug Administration or European Medicines Agency.
Table 1. Current statuses of bispecific antibody (bsAb)-based immune-cell engagers (ICEs) in clinical studies or bsAb-based ICEs approved by the United States Food and Drug Administration or European Medicines Agency.
BsAb NamesCompanyTargetRoleDevelopment Stage
(Selected)
AFM13AffimedCD16 × CD30NK cell engagerPhase II (NCT04101331)
AMG 160AmgenCD3 × PSMAT-cell engagerPhase I (NCT03792841)
AMG 199AmgenCD3 × MUC17T-cell engagerPhase I (NCT04117958)
AMG 330AmgenCD3 × CD33T-cell engagerPhase I (NCT02520427)
AMG 427AmgenCD3 × FLT3T-cell engagerPhase I (NCT03541369)
AMG 509AmgenCD3 × STEAP1T-cell engagerPhase I (NCT04221542)
AMG 701AmgenCD3 × BCMAT-cell engagerPhase I (NCT03287908)
AMG 910AmgenCD3 × CLDN18.2T-cell engagerPhase I (NCT04260191)
APVO414/ES414/MOR209Aptevo TherapeuticsCD3 × PSMAT-cell engagerPhase I (NCT02262910)
APVO436Aptevo TherapeuticsCD3 × CD123T-cell engagerPhase I (NCT03647800)
Blinatumomab/BlincytoAmgenCD3 × CD19T-cell engagerMarketed
Catumaxomab/RemovabFresenius Biotechand
and Trion Pharma
CD3 × EpCAMT-cell engagerWithdrawn from the
market
CC-1University of TubingenCD3 × PSMAT-cell engagerPhase I (NCT04104607)
CC-93269/EM801CelgeneCD3 × BCMAT-cell engagerPhase I (NCT03486067)
Cibisatamab/RG7802/RO6958688RocheCD3 × CEAT-cell engagerPhase I (NCT02324257, NCT02650713)
CLN-049Cullinan OncologyCD3 × FLT3T-cell engagerPhase I (NCT05143996)
Elranatamab/PF-06863135PfizerCD3 × BCMAT-cell engagerPhase II (NCT04649359)
EMB-06EpimAb BiotherapeuticsCD3 × BCMAT-cell engagerPhase I/II (NCT04735575)
Epcoritamab/GEN3013Genmab A/SCD3 × CD20T-cell engagerPhase III (NCT04628494)
ERY974ChugaiCD3 × GPC3T-cell engagerPhase I (NCT05022927)
Flotetuzumab/MGD006MacroGenicsCD3 × CD123T-cell engagerPhase II (NCT04582864)
Glofitamab/RG6026/RO7082859RocheCD3 × CD20T-cell engagerPhase III (NCT04408638)
ISB 1342/GBR 1342Ichnos SciencesCD3 × CD38T-cell engagerPhase I (NCT03309111)
JNJ-63709178Johnson & JohnsonCD3 × CD123T-cell engagerPhase I (NCT02715011)
JNJ-63898081Johnson & JohnsonCD3 × PSMAT-cell engagerPhase I (NCT03926013)
JNJ-67571244Johnson & JohnsonCD3 × CD33T-cell engagerPhase I (NCT03915379)
JNJ-75348780Johnson & JohnsonCD3 × CD22T-cell engagerPhase I (NCT04540796)
Linvoseltamab/REGN 5458Regeneron PharmaceuticalsCD3 × BCMAT-cell engagerPhase I/II (NCT03761108)
M701YZYBioCD3 × EpCAMT-cell engagerPhase I (NCT04501744)
M802YZYBioCD3 × HER2T-cell engagerPhase I (NCT04501770)
MGD007MacrogenicsCD3 × GPA33T-cell engagerPhase I/II (NCT03531632)
Mosunetuzumab/RG7828GenentechCD3 × CD20T-cell engagerPhase III (NCT04712097)
Nivatrotamab/Hu3F8-BsAbY-mAbsCD3 × GD2T-cell engagerPhase I/II (NCT04750239)
Odronextamab/REGN1979Regeneron PharmaceuticalsCD3 × CD20T-cell engagerPhase II (NCT03888105)
REGN4018Regeneron PharmaceuticalsCD3 × MUC16T-cell engagerPhase I/II (NCT03564340)
REGN5459Regeneron PharmaceuticalsCD3 × BCMAT-cell engagerPhase I/II (NCT04083534)
REGN5678Regeneron PharmaceuticalsCD28 × PSMAT-cell engagerPhase I/II (NCT03972657)
REGN7075Regeneron PharmaceuticalsCD28 × EGFRT-cell engagerPhase I/II (NCT04626635)
Talquetamab/JNJ-64407564Johnson & JohnsonCD3 × GPRC5DT-cell engagerPhase II (NCT04634552)
Tarlatamab/AMG 757AmgenCD3 × DLL-3T-cell engagerPhase II (NCT05060016)
Teclistamab/JNJ-64007957Johnson & JohnsonCD3 × BCMAT-cell engagerPhase III (NCT05083169)
Tepoditamab/MCLA-117MerusCD3 × CLEC12AT-cell engagerPhase I (NCT03038230)
TNB-383BTeneoOneCD3 × BCMAT-cell engagerPhase I (NCT03933735)
TNB-486TeneoTwoCD3 × CD19T-cell engagerPhase I (NCT04594642)
TNB-585AmgenCD3 × PSMAT-cell engagerPhase I (NCT04740034)
XmAb13676/PlamotamabXencorCD3 × CD20T-cell engagerPhase I (NCT02924402)
XmAb14045/VibecotamabXencorCD3 × CD123T-cell engagerPhase I (NCT02730312)
XmAb18087/TidutamabXencorCD3 × SSTR2T-cell engagerPhase I/II (NCT04590781)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shin, H.G.; Yang, H.R.; Yoon, A.; Lee, S. Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy. Int. J. Mol. Sci. 2022, 23, 5686. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23105686

AMA Style

Shin HG, Yang HR, Yoon A, Lee S. Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy. International Journal of Molecular Sciences. 2022; 23(10):5686. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23105686

Chicago/Turabian Style

Shin, Ha Gyeong, Ha Rim Yang, Aerin Yoon, and Sukmook Lee. 2022. "Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy" International Journal of Molecular Sciences 23, no. 10: 5686. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23105686

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop