Next Article in Journal
In Vivo Antihypertensive and Ex Vivo Vasodilatory Studies of Taxifolin
Previous Article in Journal
Integrative Study of Dipsaci Radix and Phlomidis Radix: Nomenclature, Morphology, DNA-Based Authentication, and Comparative Effects on Osteoclastogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Overview of Target Membrane Proteins for Near-Infrared Photoimmunotherapy

Division of Fundamental Technology Development, Near InfraRed Photo-ImmunoTherapy Research Institute at Kansai Medical University, 2-5-1, Shin-Machi, Hirakata, Osaka 573-1010, Japan
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(9), 1419; https://doi.org/10.3390/ph18091419
Submission received: 2 August 2025 / Revised: 2 September 2025 / Accepted: 17 September 2025 / Published: 21 September 2025
(This article belongs to the Section Radiopharmaceutical Sciences)

Abstract

Near-infrared photoimmunotherapy (NIR-PIT) is a recently developed cancer treatment that utilizes antibody–photoabsorber (IRDye700DX [IR700]) conjugates and NIR light. Necrotic cell death associated with lethal membrane damage is induced when this conjugate binds to an antigen on cancer cells, and it is exposed to NIR light. Therefore, various membrane proteins are potential therapeutic targets for NIR-PIT, and many studies have described various target molecules and specific antibodies. To develop future drugs for NIR-PIT, the selection of appropriate target membrane proteins and monoclonal antibodies will be important. In this review, we summarize the membrane targets and antibodies for NIR-PIT used in previous studies, focusing on the characteristics of each molecule.

1. Introduction

Near-infrared photoimmunotherapy (NIR-PIT) is a recently developed cancer treatment. The cell-killing effects of NIR-PIT are induced by the photochemical reaction between NIR-PIT agents and NIR light. NIR-PIT agents consist of conjugates of a tumor-targeting monoclonal antibody (mAb) and a photoabsorber (IRDye700DX [IR700]), which absorbs light at 690 nm in the NIR range [1,2]. Although these conjugates and NIR light alone are minimally toxic, lethal damage is induced when this conjugate binds to an antigen on cancer cells and is exposed to NIR light (Figure 1). Thus, NIR-PIT is a highly selective cancer treatment with few adverse effects. Furthermore, NIR-PIT induces immunogenic cell death (ICD) and activates local tumor immunity, meaning that NIR-PIT has the potential to control both the primary tumor and metastatic and recurrent tumors [3,4]. Therefore, NIR-PIT has attracted attention as a novel anti-cancer strategy.
NIR-PIT, using the anti-epidermal growth factor receptor (EGFR) antibody cetuximab conjugated with IR700 (cetuximab sarotalocan sodium, Akalux®: Rakuten Medical Inc., San Diego, CA, USA), was approved for clinical use in patients with unresectable locally advanced or recurrent head and neck squamous cell cancer (HNSCC) in Japan [5,6]. In addition, phase III clinical trials in patients with HNSCC are ongoing worldwide [7] (Table 1). To further develop NIR-PIT, it is essential to expand the indications for EGFR-targeting NIR-PIT and develop NIR-PIT using novel targets and antibodies.
Many studies have been performed since the introduction of NIR-PIT, and several review articles on NIR-PIT have been reported [1,8,9]. The mechanism by which NIR-PIT induced cell death is also being elucidated. The structural change in IR700 induced by NIR light exposure causes mAb-IR700 conjugates to aggregate. When this aggregation occurs when mAb is bound to membrane proteins, the cell membrane is damaged, leading to the influx of water into the cell and resulting in cell swelling and eventual cell death. This cell death mode is necrosis-like and differs from apoptosis, which is characterized by cell shrinkage. Thus, the type of antibody-membrane protein complexes formed on the membrane might have a significant impact on the therapeutic efficacy of NIR-PIT, and the properties of the antibody and membrane protein would be greatly involved in the formation of this complex. In addition, because NIR-PIT has a novel therapeutic mechanism, it is necessary to select target molecules and antibodies from a different perspective. This review specifically focuses on the target membrane proteins and the antibodies used in previous studies, which will help guide the future development of drugs for NIR-PIT.

2. Membrane Proteins as Targets of NIR-PIT

The cytotoxic mechanism of NIR-PIT is based on cellular membrane damage caused by the photochemical reaction of NIR-PIT agents induced by NIR light and aggregation of the drug–membrane protein complex [10]. Therefore, it is important to summarize the properties of membrane proteins that could influence therapeutic efficacy, such as the type, size, and antibody-binding site of the membrane proteins reported in previous NIR-PIT studies. The involvement of characteristics of membrane proteins in NIR-PIT is discussed in detail in the subsequent sections. In previous reports, the properties of membrane proteins, such as the type, size, and antibody-binding site of the membrane proteins, other than internalization, exert a negligible impact on the therapeutic efficacy of NIR-PIT.

2.1. Protein Type

A large variety of membrane proteins exist in eukaryotic cells, and they can be classified into two groups depending on their membrane interaction: integral and peripheral membrane proteins. In integral membrane proteins, one or multiple domains are embedded within the entire phospholipid bilayer. Conversely, peripheral membrane proteins interact with the membrane through indirect interactions (electrostatic, hydrophobic, and fatty acid modification) or direct interactions (hydrophobic tails or glycosylphosphatidylinositol [GPI] anchors) (Figure 2) [11,12]. Target molecules that have been implemented to date for NIR-PIT are summarized in Table 1. The therapeutic effects of NIR-PIT targeting both integral and peripheral membrane proteins have been reported, although GPI-anchored proteins are not expected to significantly interact with the plasma membrane. Thus, the efficacy of NIR-PIT might be less dependent on the protein type. Of course, several other membrane proteins exist, but it is not necessary to exclude them by type.

2.2. Protein Size

The size of membrane proteins, particularly the size of the extracellular domain of the protein, might influence the effect of NIR-PIT because its mechanism of action involves aggregation of the drug–membrane protein complex. The membrane proteins listed in Table 2 vary in size, with the smallest (e.g., podoplanin [PDPN], CD20) being in the 30 kDa range and the largest reaching nearly 200 kDa. In other words, some membrane proteins are slightly larger than IgG antibodies (150 kDa), whereas others are considerably smaller. Although there might be differences in efficacy depending on the size of the membrane protein, the therapeutic effects of NIR-PIT have been observed for small, medium, and large membrane proteins.
Most single-pass transmembrane proteins and GPI-anchored proteins possess a large extracellular domain consisting of several hundred amino acid residues. Among them, PDPN has a relatively small extracellular domain of 130 amino acids [41,42]. The cytotoxic effects of NIR-PIT targeting PDPN with NZ-1 antibody have been reported in malignant pleural mesothelioma [27].
In the case of multipass transmembrane proteins, the size of the extracellular domain is variable. In particular, CD29 and CD133 have large extracellular domains, and they are assumed to respond in the same manner as single-pass transmembrane proteins. Conversely, the extracellular domain of CD20 is shorter than 50 amino acids. However, NIR-PIT using rituximab, an anti-CD20 antibody, exhibited a sufficient therapeutic effect [30]. It is unknown whether NIR-PIT can exert therapeutic effects if the extracellular domain is even smaller, but it is worthy of investigation. However, if the extracellular domain is too small, it might be difficult to develop antibodies that bind to it.

2.3. Antibody-Binding Site

Considering membrane damage caused by aggregation of the drug–membrane protein complex, the location at which the antibody binds to the membrane protein is important. If aggregation occurs too distantly from the membrane, then insufficient membrane damage might be induced. Some of the antibodies used in NIR-PIT, listed in Table 1, have undisclosed antigen-binding sites. Considering only cases that are clear, some antibodies bind to sites close to the cell membrane, whereas others bind to more distant sites.
Anti-EGFR antibodies bind slightly away from the membrane, but they have achieved sufficient therapeutic efficacy. In particular, IR700-cetuximab is already in clinical use. Regarding HER2, two anti-HER2 antibodies, namely trastuzumab and pertuzumab, have been evaluated in HER2-targeting NIR-PIT. Trastuzumab binds to the extracellular domain IV of HER2, which is close to the membrane [43]. In contrast, the binding region of pertuzumab is domain II, which is about 300 amino acids away from the membrane [44]. However, the therapeutic effects of NIR-PIT using these antibodies are similar [33]. Further study is needed to clarify the relationship between binding sites and treatment efficacy.

2.4. Other Properties

Other properties of membrane proteins might be involved in the therapeutic effect of NIR-PIT. One factor affecting therapeutic efficacy is the internalization of drugs, as the cell-killing effect of NIR-PIT occurs on the membrane surface. It is assumed that the therapeutic effect is better if greater amounts of the drug remain on the membrane surface. Indeed, a previous report suggested that the cytotoxic effects of NIR-PIT with trastuzumab were reduced by internalization of the drug [45]. Conversely, in vivo therapeutic effects have been reported for NIR-PIT using trastuzumab and cetuximab, which are known to be internalized. This might be because the internalization of these antibodies is not rapid, and newly migrated drugs can bind to newly emerging target molecules on the surface of the membrane in vivo; therefore, a certain amount of drugs remains on the cell surface. Thus, membrane proteins that internalize quickly after antibody binding or are slowly recycled might not be suitable targets for NIR-PIT.
The expression levels of membrane proteins on the cell surface are important. If the density of expression is higher, then aggregation among multiple molecules might be induced, leading to greater membrane damage. This is because π-π stacking by phthalocyanine, which occurs in the chemical change of IR700 after light irradiation, is considered important for inducing aggregation, and stacking can occur even between molecules if they are close to each other [46]. Thus, targeting membrane proteins that form dimers or multimers might improve the therapeutic efficacy of NIR-PIT.
Glycosidation or other properties do not appear to significantly affect the therapeutic effect of NIR-PIT. Specific examples are presented in Section 4.

3. Antibodies

The high selectivity of NIR-PIT is ensured by antibodies. Antibodies are classified into five classes: IgA, IgD, IgE, IgG, and IgM. IgG is the most common isotype in the human serum antibody fraction, accounting for 75% of the circulating antibody pool. IgG is also mainly used as a clinical drug. IgG is further classified by its heavy chain into four subclasses: IgG1, IgG2, IgG3, and IgG4. Each subclass has a different profile, including differences in their binding affinity for crystallizable fragment gamma receptor and half-lives, and the majority of antibodies for tumor treatment are of the IgG1 isotype (Table 3) [47]. Moreover, therapeutic mAbs can be classified into four types by the manufacturing method: murine, chimeric (structural chimeras made by fusing mouse variable regions with human antibody constant regions), humanized (only complementarity-determining region from mice), and fully human antibodies (Figure 3) [48]. Humanization of mAbs can reduce their immunogenicity while maintaining their therapeutic efficacy. The type and subclass of these antibodies affect the therapeutic effects of antibody monotherapy and conjugated mAb therapy, including ADCs, radioimmunotherapy, and immunotoxins.
In NIR-PIT, the properties of the antibody itself have little or no effect on the cytotoxic mechanism. This is because in NIR-PIT, antibodies are used as carriers to deliver IR700 in a cancer-specific manner, and the effect of the antibody itself, such as antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC), is not a requirement. Indeed, NIR-PIT with cetuximab (IgG1) and panitumumab (IgG2) induces similar cytotoxic effects in vitro, proving that the IgG subclass has little effect on the cytotoxic mechanism [53]. Of course, the antibody itself is assumed to exert a therapeutic effect in vivo, but even without such an effect, a sufficient therapeutic effect is expected. Therapeutic efficacy has been observed even with IgG4, which induces little ADCC or CDC as previously described. For some target molecules, it might be better to select an antibody without pharmacological effects to avoid side effects in normal organs. Conversely, the biological half-life of antibodies affects their therapeutic efficacy. In mouse models, NIR-PIT employing the fully human antibody panitumumab produces better therapeutic efficacy than that employing the chimeric antibody cetuximab because of the longer biological half-life of panitumumab despite little difference in their in vitro efficacy. In addition, one of the factors affecting the therapeutic efficacy is mAb-IR700 conjugate internalization because the cell-killing effect of photoimmunotherapy is attributable to the aggregation of the drug on the membrane. This opposite is true for ADCs, for which antibody internalization is critical. A previous report suggested that the cytotoxic effects of NIR-PIT are greatest when photochemical reactions occur on the membrane and are attenuated by antibody internalization [45]. Even for the same target molecule, the biological half-life and the internalization rate should be considered when selecting an antibody, which could help optimize NIR-PIT in the future.
The therapeutic effects of NIR-PIT do not require Fc region-mediated effects; therefore, engineered antibody fragments such as those in Figure 3B might be useful as NIR-PIT moieties because of their advantages, such as rapid pharmacokinetics and high tissue penetration compared with whole IgG. Rapid clearance of the drug from the body would reduce concerns about photosensitivity associated with NIR-PIT. High tissue penetration is expected to increase the effectiveness of NIR-PIT by allowing it to reach deeper into the tumor. NIR-PIT using single-chain antibody variable fragments, minibodies, and Fab fragments achieved therapeutic efficacy in vitro and in vivo [21,54,55]. In addition, removal of the Fc region prevents cellular injury induced by ADCC or CDC, thereby avoiding adverse effects outside the target lesion [56]. Thus, fragment antibodies have potential utility in NIR-PIT. However, their short half-lives might affect their therapeutic efficacy as previously mentioned, and further validation is needed, including appropriate IR700 conjugate administration.

4. Typical NIR-PIT for Cancer Cells

Typical membrane proteins targeted in NIR-PIT for cancer are described in the subsequent sections. These specific examples illustrate the implementation of NIR-PIT against various targets.

4.1. EGFR

EGFR is a 170 kDa transmembrane tyrosine kinase belonging to the erythroblastosis oncogene B (ErbB) family. The EGFR gene encodes a 1186-amino acid transmembrane glycoprotein that consists of three regions: an extracellular ligand-binding domain, a transmembrane region, and an intracellular cytoplasmic tyrosine kinase region [57]. The extracellular domain consists of 621 amino acids, and it is divided into four subdomains, namely domains I (also known as L1, amino acids 1–133), II (also known as S1 or CR1, amino acids 134–312), III (also known as L2, amino acids 313–445), and IV (also known as S2 or CR2, amino acids 446–621) [58,59]. Domains I and III share 37% sequence identity, and they are leucine-rich fragments that interact with ligands [60]. Domain II is a cystine-rich region that contributes to the dimerization of two EGFR moieties [61]. Domain IV is also cystine-rich, and it links to transmembrane domains.
Currently, four EGFR-targeting mAbs, cetuximab (chimeric IgG1 antibody), panitumumab (humanized IgG2 antibody), nimotuzumab (humanized IgG1 antibody), and necitumumab (humanized IgG1 antibody), are already on the market, and they are currently under clinical investigation in various tumors [62]. All of these antibodies inhibit ligand binding to EGFR by binding to domain III, albeit through different binding sites (cetuximab, amino acids 408–468; panitumumab, amino acids 386–391; nimotuzumab, amino acids 353–358; and necitumumab, amino acids 384–409) [62,63]. These differences in binding sites might lead to differences in efficacy and resistance.
In NIR-PIT, the therapeutic efficacy of cetuximab-IR700 (Cet-IR700) and panitumumab-IR700 (Pan-IR700) conjugates was verified by previous studies [9]. The therapeutic efficacy of the Cet-IR700 and Pan-IR700 conjugates has been compared. The in vivo half-life of Pan-IR700 is longer than that of Cet-IR700; thus, NIR-PIT with Pan-IR700 is associated with greater therapeutic efficacy than that with Cet-IR700 [20]. Antibodies other than cetuximab and panitumumab could also be useful in NIR-PIT. A previous report suggested that necitumumab interacts with a similar epitope to cetuximab, although the paratopes are different [64]. Moreover, the paratope cavity of necitumumab is significantly larger than that of panitumumab; thus, necitumumab can bind even cetuximab- and panitumumab-resistant EGFR variants [65,66].

4.2. Human Epidermal Growth Factor Receptor 2 (HER2)

HER2, also known as ErbB2, c-erbB2, or HER2/neu, is a 185 kDa transmembrane glycoprotein and a member of the ErbB family. The HER2 gene encodes a 1255-amino acid transmembrane glycoprotein with three regions: an extracellular domain, a transmembrane lipophilic segment, and an intracellular tyrosine kinase domain. The extracellular domain of HER2 consists of 620 amino acids, and it is divided into four domains: two leucine-rich domains (I and III) and two cysteine-rich domains (II and IV). In the ErbB family, the extracellular domain can exist as a tethered form (inactive form) or an extended form (active form). In the tethered form, domain II is tethered to domain IV, rendering it unable to dimerize with other receptors [67,68].
HER2 has been studied as an effective therapeutic target, and two mAbs, namely trastuzumab and pertuzumab, have been developed as HER2-targeting therapies. Trastuzumab is a humanized recombinant monoclonal IgG1 antibody that binds to extracellular domain IV of HER2 [43]. By contrast, the binding region of pertuzumab is domain II, and this drug can inhibit the heterodimerization of HER2 with other receptors [44].
The therapeutic effect of NIR-PIT using trastuzumab-IR700 (Tra-IR700) conjugates has been reported in several tumors, including breast, esophageal, lung, ovarian, and bile duct cancers [2,22,23,69,70,71]. Furthermore, the therapeutic efficacy of NIR-PIT using the combination of Tra-IR700 and pertuzumab-IR700 (Per-IR700) is stronger than that using Tra-IR700 or Per-IR700 alone [24]. In their study, no difference in therapeutic efficacy was observed for NIR-PIT between Tra-IR700 and Per-IR700 in vitro; thus, the region to which the antibody binds might not affect the therapeutic effect.

4.3. CD133

CD133, also known as prominin-1 or AC133, is a 97 kDa pentaspan transmembrane glycoprotein that contains two extracellular loops with nine putative N-linked glycosylation sites [72]. After heavy glycosylation, the apparent molecular weight of CD133 increases to 120 kDa. In epithelial cells, CD133 is restricted to plasma membrane protrusions, and it accumulates in membrane microdomains (lipid rafts) [73,74].
The CD133 gene encodes an 865-amino acid transmembrane glycoprotein consisting of an N-terminal extracellular domain (amino acids 20–108), five transmembrane domains, cysteine-rich two small intracellular loops, two large extracellular loops (amino acids 179–433 and 508–792, respectively), and an intercellular cytoplasmic tail [75,76]. The extracellular loop contains nine glycosylation sites (Asn206, Asn220, Asn274, Asn395, Asn414, Asn548, Asn580, Asn729, and Asn730) [72].
To date, the mAbs CD133/1 (AC133) and CD133/2 (AC141), which bind to different glycosylated epitopes in the extracellular domain region of CD133, are the most studied. AC133 conjugated to the genetically modified cytolethal distending toxin AC141, conjugated to polymeric nanoparticles loaded with paclitaxel, and radiolabeled AC133 with iodine-131 exhibit cytotoxic effects in tumors [77,78,79].
NIR-PIT using AC133-IR700 conjugates induces cytotoxicity in glioblastoma stem cells and suppresses tumor growth in both subcutaneous and orthotopic tumor models [33].

4.4. CD44

CD44 is a single-chain transmembrane glycoprotein and a member of the cartilage link protein family [80]. In humans, the CD44 gene consists of 19 exons, and the first and last five exons are constant. The intermediate nine exons are subjected to alternative splicing, resulting in multiple CD44 variant (CD44v) isoforms [81]. Accordingly, the molecular weight of CD44 ranges from 85 to 230 kDa. The smallest CD44 isoform (85–95 kDa), which is only coded by the constant exons, is most common, and it is called standard CD44 (CD44s). CD44s is ubiquitously expressed on the membranes of vertebrate cells, and it plays essential roles in homeostasis [82]. By contrast, CD44v expression is mostly restricted to epithelia [83]. Importantly, CD44v isoforms are cancer stem cell markers, and they are associated with tumor progression and metastasis [84].
All CD44 isoforms consist of one extracellular domain, one transmembrane domain, and one intracellular domain [85]. Several ligands, including hyaluronic acid (HA) and osteopontin, bind to the extracellular domain and regulate the PI3K signaling pathway. CD44s is a 363-amino acid glycoprotein that consists of a 270-amino acid extracellular domain, a 21-amino acid transmembrane domain, and a 72-amino acid C-terminal cytoplasmic domain. The theoretical molecular weight of 37 kDa is increased to 85–95 kDa through the attachment of glycosaminoglycan.
CD44 is an attractive target, and more than 140 commercially available CD44-targeting mAbs have been developed [86]. H4C4 is an anti-human CD44 IgG1 mAb that reduces tumor growth, metastasis, and post-radiation recurrence in pancreatic cancer [87]. IM7, another anti-mouse/human pan-CD44 IgG2b mAb that recognizes the HA-binding domain of CD44, inhibits the cell migration and invasion of breast cancer cells [88,89]. Information on other antibodies is omitted from this review.
NIR-PIT using IM7 conjugated with IR700 has been reported. NIR-PIT targeting CD44 induces ICD and increases tumor-infiltrating CD8+ cell counts in syngeneic models of colorectal, lung, head and neck, and oral cancers [90,91,92,93,94].

4.5. Carcinoembryonic Antigen (CEA)

CEA, also known as CEA-related cell adhesion molecule 5 (CEACAM5) or CD66e, is a 150–200 kDa heavily glycosylated protein. CEA belongs to the immunoglobulin (Ig) superfamily, and it is expressed on the cellular membrane through a GPI anchor [95]. CEA belongs to the CEACAM family, which is a subgroup of the CEA family. It was first isolated from human colorectal cancer tissue, and it is commonly used in clinical practice as a serum biomarker [96].
CEA consists of 651 amino acids and contains seven Ig-like domains, including a single Ig variable region-like N-terminal domain followed by six Ig constant region type 2-like domains termed A1, B1, A2, B2, A3, and B3 [97,98,99]. The C-terminal domain of CEA is anchored to the cellular membrane through GPI [100].
In normal colon epithelial cells, CEA expression is strictly localized to the apical surface. Conversely, CEA is expressed over the entire cell surface of colorectal cancer cells [101]. Moreover, CEA is overexpressed in various tumors, including breast, pancreatic, lung, and thyroid tumors [102,103,104,105]. CEA is a GPI-anchored protein that has no intracellular domain. While the anti-CEA mAbs are useful as imaging carriers [106,107], but antibodies alone are unlikely to show therapeutic efficacy [108]. Anti-CEA mAbs have potential as antibody-drug conjugates (ADCs), and labetuzumab govitecan (IMMU 130), an anti-CEA/SN-38 conjugate, displayed therapeutic activity in heavily pretreated patients with metastatic colorectal cancer [109]. Moreover, several cytotoxic agents based on anti-CEA mAbs, such as PR1A3 and 15-1-32, have been developed [108,110,111].
NIR-PIT targeting CEA using chimeric anti-CEA mAb (Genara Biosciences LLC, Morgan Hill, CA, USA) or the anti-CEA IgG4 C2-45 suppressed tumor progression in gastric and pancreatic tumor models [35,112,113]. Moreover, NIR-PIT targeting CEA was effective in a patient-derived orthotopic xenograft model [34].

4.6. Prostate-Specific Membrane Antigen (PSMA)

PSMA, also known as glutamate carboxypeptidase II or N-acetylated alpha-linked acidic dipeptidase in the central/peripheral nervous system, is a type II transmembrane glycoprotein that is highly expressed in prostate cancer cells [114]. The predicted molecular weight of PSMA is 84 kDa, but it is increased to more than 100 kDa by glycosylation, which is necessary for proteolytic activity [115,116]. In addition, PSMA is expressed on the cell surface in both dimeric and monomeric forms, and dimerization is required for its enzymatic activity [117].
PSMA consists of a short N-terminal cytoplasmic domain (19 amino acids), a single hydrophobic transmembrane domain (24 amino acids), and a large extracellular domain (707 amino acids) [118]. The extracellular domain has nine N-glycosylation sites, two domains of unknown function (amino acids 44–150 and 151–274, respectively), proline- and glycine-rich regions (amino acids 145–172 and 249–273, respectively), a catalytic domain (amino acids 274–587), and a helical dimerization domain (amino acids 587–750) [115,119].
7E11, also known as CYT-356 and capromab, is the first developed anti-PSMA mAb, and [111In]-capromab pendetide has been used as a single-photon emission computed tomography agent in patients with prostate cancer [120,121]. However, clinical trials using [90Y]-capromab pendetide for radioimmunotherapy were discontinued because of poor efficacy [122]. Molecular mapping revealed that the reason for the limited therapeutic efficacy is that the 7E11 recognizes the intracellular domain of PSMA [123,124]. To improve the imaging quality and therapeutic efficacy of the PSMA-targeting strategy, several mAbs targeting the extracellular epitope of PSMA have been developed, and J591 (humanized IgG1 mAb) is the most commonly used anti-PSMA antibody for radioimmunotherapy in clinical trials [125].
NIR-PIT targeting PSMA employing a commercially available antibody (clone: YPSMA-1) induces cytotoxic effects associated with membrane damage [126]. Moreover, NIR-PIT using a fully human anti-PSMA mAb that binds a conformation-dependent epitope within the extracellular domain significantly suppresses tumor proliferation [29].

4.7. Podoplanin (PDPN)

PDPN, also known as T1α, aggrus, or gp36, is a 38 kDa type I transmembrane mucin-like glycoprotein. PDPN is expressed in normal tissue, such as kidney podocytes, lung type I alveolar epithelium, and lymphatic endothelium [127,128,129]. PDPN has no enzymatic activity motif, and it performs its function through interactions with other proteins. PDPN is an endogenous ligand for C-type lectin-like receptor 2, which is highly expressed in platelets.
PDPN consists of an N-terminal extracellular domain of 130 amino acids, followed by a single transmembrane domain of 25 amino acids, and its intracellular domain consists of only nine amino acids [41,42]. The extracellular domain contains a repeat sequence of EDxxVTPG, known as PLAG1, 2, 3, and PLAG-like domain (PLD or PLAG4) [130,131].
Currently, several anti-PDPN mAbs have been developed, including NZ-1, P2-0, and MS-1 (recognizing the PLAG2/3 domain); D2-40 (recognizing the PLAG1/2 domain); LpMab-2 (recognizing the glycopeptide Thr55–Leu64 of human PDPN); LpMab23 (recognizing the naked peptide Gly54–Leu64 of human PDPN); and chPMab and 2F7 (recognizing PLD) [131]. Some of them can inhibit PDPN-induced lung metastasis [132,133,134,135]. Moreover, chimeric antigen receptor-transduced T cells targeting PDPN using NZ-1 have displayed efficacy against glioblastoma in vitro and in orthotopic models [136].
The cytotoxic effects of NIR-PIT targeting PDPN with NZ-1 have been reported in malignant pleural mesothelioma [27]. Moreover, NIR-PIT induced death in PDPN-expressing cancer cells and cancer-associated fibroblasts, leading to an increase in the number of cytotoxic T cells in tumor tissue [137].

4.8. Intercellular Adhesion Molecule-1 (ICAM-1)

ICAM1, also known as CD54, is a 90 kDa Ig superfamily transmembrane protein. The molecular weight of human ICAM-1 varies up to 114 kDa because it contains eight putative N-glycosylation sites [138].
ICAM-1 consists of five extracellular Ig-like domains (D1–D5), a short hydrophobic transmembrane region, and a small carboxyl-terminal cytoplasmic tail that interacts with the actin cytoskeleton [139].
The therapeutic effects of NIR-PIT targeting ICAM-1 in triple-negative breast cancer have been reported [25]. As an NIR-PIT agent, a commercially available anti-ICAM-1 mAb (clone R6-5-D6) has been employed. Interestingly, drastic histological changes, namely cytoplasmic vacuolation and proliferation marker downregulation, are induced only 2 h after in vivo NIR-PIT targeting ICAM-1.

4.9. Nectin Cell Adhesion Molecule 4 (Nectin-4)

Nectin-4, also known as poliovirus receptor-related protein 4, is a 66 kDa Ca2+-independent Ig-like type I transmembrane protein. Nectin-4 is a member of Nectin family, which regulates cellular movement, proliferation, survival, differentiation, and polarization [140,141]. Although Nectin 1–3 are widely expressed in adult tissues, the expression of Nectin-4 is limited to the embryo and placenta [142,143].
Nectin-4 consists of an extracellular region, a transmembrane region, and a cytoplasmic tail that interacts with afadin, an F-actin-associated molecule, to bind to the actin cytoskeleton [144]. The extracellular domain contains two Ig-like C2-type domains and one Ig-like V-type domain.
The fully anti-human Nectin-4 mAb AGS-22M6, targeting the extracellular domain of human Nectin-4, was generated using the XenoMouse strains genetically engineered with a humanized humoral immune system [145]. Enfortumab vedotin, which consists of AGS-22M6 conjugated with monomethyl auristatin E, has been developed, and its therapeutic efficacy has been reported in various tumor models, including breast, bladder, pancreatic, and lung cancers [146]. The US Food and Drug Administration has approved enfortumab vedotin-ejfv (Padcev, Astellas Pharma) in combination with pembrolizumab (anti-PD1 mAb) for the treatment of patients with locally advanced or metastatic urothelial cancer who are ineligible for cisplatin-containing chemotherapy [147].
The cytotoxic effects of NIR-PIT targeting Nectin-4 using enfortumab biosimilars in luminal subtype bladder cancer cell lines have been reported [26]. Moreover, NIR-PIT has displayed efficacy in bladder cancer orthotopic models. Previously, the effectiveness of endoscopic NIR-PIT was reported; therefore, NIR light irradiation using cystoscopy might be effective and clinically available.

5. Limitation

This review concentrates on representative membrane proteins as well as those membrane proteins that have not been addressed in previous reviews, with an emphasis on the classifications of membrane proteins and the associated antibodies that may be utilized in NIR-PIT agents. Consequently, it does not encompass all proteins and antibodies that are relevant to NIR-PIT. Conversely, it encapsulates the categorization of membrane proteins and antibodies, which will prove advantageous for forthcoming investigations into NIR-PIT. Second, there are no reports of cases where NIR-PIT treatment was ineffective, so it is unclear which membrane proteins are not applicable to. However, NIR-PIT currently works on various types of membrane proteins and may be effective against all types of membrane proteins. Further knowledge will be accumulated as it is applied to various membrane proteins in the future.

6. Summary

In the decade since NIR-PIT was developed, its efficacy against various cancer types using different antibodies has been reported. Currently, only NIR-PIT using the EGFR antibody cetuximab is approved for clinical use. To apply NIR-PIT in additional cancer regimens, it is important to identify an appropriate set of cancer types and targeting proteins and develop useful antibodies. The cytotoxic mechanism of NIR-PIT is based on cellular membrane damage caused by the photochemical reaction of agents induced by NIR light and aggregation of the drug–membrane protein complex. Therefore, to attain sufficient efficacy for NIR-PIT, it appears important to select appropriate membrane proteins and antibodies. However, as previously mentioned, the therapeutic effects of NIR-PIT on various types of membrane proteins using all types of antibodies have been demonstrated. The properties of membrane proteins, such as the type, size, and antibody-binding site of the membrane proteins, exert a negligible impact on the therapeutic efficacy of NIR-PIT. Therefore, a variety of target molecules can be selected as long as they are sufficiently present on the cell membrane. It may be more suitable when expressed at high density. On the other hand, membrane proteins that internalize quickly after antibody binding and are slowly recycled might not be suitable targets. That is, if a target cell overexpresses a molecule on the cell surface and produces antibodies against it, then the cell can potentially be killed by NIR-PIT. NIR-PIT does not cause cytotoxicity without light exposure, so if the antibody itself has no pharmacological effect, it might be possible to target more molecules than ever before, since it is not limited to those expressed only in cancer. Indeed, specificity could be achieved by application of light to the cancer tissue only. In addition, because the pharmacological effect of the antibody itself is not required and accumulation at sites distant from the target lesion is not a problem, the development of antibodies could be easier than that for other antibody-based drugs. From the viewpoint of side effects, the pharmacological effects of the antibody itself must be considered. NIR-PIT has potential as a novel anti-cancer strategy because it is a highly effective and less invasive tumor treatment, and thus, further research and development are expected.

Author Contributions

M.S.: Conceptualization, Visualization, Writing—original draft. H.H.: Conceptualization, Project administration, Resources, Supervision, Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported, in part, by the Japan Science and Technology Agency FOREST Program (Grant number JPMJFR205I [HH]).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no competing interests in relation to this study.

Abbreviations

CDRcomplementarity-determining region
CEAcarcinoembryonic antigen
EGFRepidermal growth factor receptor
ErbBerythroblastosis oncogene B
GPIglycosylphosphatidylinositol
HER2human epidermal growth factor receptor
HNSCChead and neck squamous cell cancer
ICAM-1intercellular adhesion molecule-1
ICDimmunogenic cell death; mAb, monoclonal antibody
Nectin-4Nectin cell adhesion molecule 4
NIR-PITnear-infrared photoimmunotherapy
PDPNpodoplanin
PSMAprostate-specific membrane antigen
scFVsingle-chain antibody variable fragment

References

  1. Kobayashi, H.; Choyke, P.L. Near-Infrared Photoimmunotherapy of Cancer. Acc. Chem. Res. 2019, 52, 2332–2339. [Google Scholar] [CrossRef]
  2. Mitsunaga, M.; Ogawa, M.; Kosaka, N.; Rosenblum, L.T.; Choyke, P.L.; Kobayashi, H. Cancer cell-selective in vivo near infrared photoimmunotherapy targeting specific membrane molecules. Nat. Med. 2011, 17, 1685–1691. [Google Scholar] [CrossRef] [PubMed]
  3. Ogawa, M.; Tomita, Y.; Nakamura, Y.; Lee, M.J.; Lee, S.; Tomita, S.; Nagaya, T.; Sato, K.; Yamauchi, T.; Iwai, H.; et al. Immunogenic cancer cell death selectively induced by near infrared photoimmunotherapy initiates host tumor immunity. Oncotarget 2017, 8, 10425–10436. [Google Scholar] [CrossRef] [PubMed]
  4. Kobayashi, H.; Furusawa, A.; Rosenberg, A.; Choyke, P.L. Near-infrared photoimmunotherapy of cancer: A new approach that kills cancer cells and enhances anti-cancer host immunity. Int. Immunol. 2021, 33, 7–15. [Google Scholar] [CrossRef] [PubMed]
  5. Tahara, M.; Okano, S.; Enokida, T.; Ueda, Y.; Fujisawa, T.; Shinozaki, T.; Tomioka, T.; Okano, W.; Biel, M.A.; Ishida, K.; et al. A phase I, single-center, open-label study of RM-1929 photoimmunotherapy in Japanese patients with recurrent head and neck squamous cell carcinoma. Int. J. Clin. Oncol. 2021, 26, 1812–1821. [Google Scholar] [CrossRef]
  6. Miyazaki, N.L.; Furusawa, A.; Choyke, P.L.; Kobayashi, H. Review of RM-1929 Near-Infrared Photoimmunotherapy Clinical Efficacy for Unresectable and/or Recurrent Head and Neck Squamous Cell Carcinoma. Cancers 2023, 15, 5117. [Google Scholar] [CrossRef]
  7. Cognetti, D.M.; Johnson, J.M.; Curry, J.M.; Kochuparambil, S.T.; McDonald, D.; Mott, F.; Fidler, M.J.; Stenson, K.; Vasan, N.R.; Razaq, M.A.; et al. Phase 1/2a, open-label, multicenter study of RM-1929 photoimmunotherapy in patients with locoregional, recurrent head and neck squamous cell carcinoma. Head. Neck 2021, 43, 3875–3887. [Google Scholar] [CrossRef]
  8. Wei, D.; Qi, J.; Hamblin, M.R.; Wen, X.; Jiang, X.; Yang, H. Near-infrared photoimmunotherapy: Design and potential applications for cancer treatment and beyond. Theranostics 2022, 12, 7108–7131. [Google Scholar] [CrossRef]
  9. Kato, T.; Wakiyama, H.; Furusawa, A.; Choyke, P.L.; Kobayashi, H. Near Infrared Photoimmunotherapy; A Review of Targets for Cancer Therapy. Cancers 2021, 13, 2535. [Google Scholar] [CrossRef]
  10. Kobayashi, H.; Choyke, P.L.; Ogawa, M. The chemical basis of cytotoxicity of silicon-phthalocyanine-based near infrared photoimmunotherapy (NIR-PIT) and its implications for treatment monitoring. Curr. Opin. Chem. Biol. 2023, 74, 102289. [Google Scholar] [CrossRef]
  11. Whited, A.M.; Johs, A. The interactions of peripheral membrane proteins with biological membranes. Chem. Phys. Lipids 2015, 192, 51–59. [Google Scholar] [CrossRef] [PubMed]
  12. Boes, D.M.; Godoy-Hernandez, A.; McMillan, D.G.G. Peripheral Membrane Proteins: Promising Therapeutic Targets across Domains of Life. Membranes 2021, 11, 346. [Google Scholar] [CrossRef]
  13. Lum, Y.L.; Luk, J.M.; Staunton, D.E.; Ng, D.K.P.; Fong, W.P. Cadherin-17 Targeted Near-Infrared Photoimmunotherapy for Treatment of Gastrointestinal Cancer. Mol. Pharm. 2020, 17, 3941–3951. [Google Scholar] [CrossRef]
  14. Kato, T.; Okada, R.; Furusawa, A.; Inagaki, F.; Wakiyama, H.; Furumoto, H.; Okuyama, S.; Fukushima, H.; Choyke, P.L.; Kobayashi, H. Simultaneously Combined Cancer Cell- and CTLA4-Targeted NIR-PIT Causes a Synergistic Treatment Effect in Syngeneic Mouse Models. Mol. Cancer Ther. 2021, 20, 2262–2273. [Google Scholar] [CrossRef]
  15. Wei, W.; Jiang, D.; Ehlerding, E.B.; Barnhart, T.E.; Yang, Y.; Engle, J.W.; Luo, Q.Y.; Huang, P.; Cai, W. CD146-Targeted Multimodal Image-Guided Photoimmunotherapy of Melanoma. Adv. Sci. 2019, 6, 1801237. [Google Scholar] [CrossRef]
  16. Fujimoto, S.; Muguruma, N.; Okamoto, K.; Kurihara, T.; Sato, Y.; Miyamoto, Y.; Kitamura, S.; Miyamoto, H.; Taguchi, T.; Tsuneyama, K.; et al. A Novel Theranostic Combination of Near-infrared Fluorescence Imaging and Laser Irradiation Targeting c-KIT for Gastrointestinal Stromal Tumors. Theranostics 2018, 8, 2313–2328. [Google Scholar] [CrossRef]
  17. Isobe, Y.; Sato, K.; Nishinaga, Y.; Takahashi, K.; Taki, S.; Yasui, H.; Shimizu, M.; Endo, R.; Koike, C.; Kuramoto, N.; et al. Near infrared photoimmunotherapy targeting DLL3 for small cell lung cancer. EBioMedicine 2020, 52, 102632. [Google Scholar] [CrossRef] [PubMed]
  18. Takao, S.; Fukushima, H.; King, A.P.; Kato, T.; Furusawa, A.; Okuyama, S.; Kano, M.; Choyke, P.L.; Escorcia, F.E.; Kobayashi, H. Near-infrared photoimmunotherapy in the models of hepatocellular carcinomas using cetuximab-IR700. Cancer Sci. 2023, 114, 4654–4663. [Google Scholar] [CrossRef] [PubMed]
  19. Nagaya, T.; Sato, K.; Harada, T.; Nakamura, Y.; Choyke, P.L.; Kobayashi, H. Near Infrared Photoimmunotherapy Targeting EGFR Positive Triple Negative Breast Cancer: Optimizing the Conjugate-Light Regimen. PLoS ONE 2015, 10, e0136829. [Google Scholar] [CrossRef]
  20. Sato, K.; Watanabe, R.; Hanaoka, H.; Harada, T.; Nakajima, T.; Kim, I.; Paik, C.H.; Choyke, P.L.; Kobayashi, H. Photoimmunotherapy: Comparative effectiveness of two monoclonal antibodies targeting the epidermal growth factor receptor. Mol. Oncol. 2014, 8, 620–632. [Google Scholar] [CrossRef]
  21. Wei, D.; Tao, Z.; Shi, Q.; Wang, L.; Liu, L.; She, T.; Yi, Q.; Wen, X.; Liu, L.; Li, S.; et al. Selective Photokilling of Colorectal Tumors by Near-Infrared Photoimmunotherapy with a GPA33-Targeted Single-Chain Antibody Variable Fragment Conjugate. Mol. Pharm. 2020, 17, 2508–2517. [Google Scholar] [CrossRef] [PubMed]
  22. Sato, K.; Nagaya, T.; Choyke, P.L.; Kobayashi, H. Near infrared photoimmunotherapy in the treatment of pleural disseminated NSCLC: Preclinical experience. Theranostics 2015, 5, 698–709. [Google Scholar] [CrossRef] [PubMed]
  23. Sato, K.; Hanaoka, H.; Watanabe, R.; Nakajima, T.; Choyke, P.L.; Kobayashi, H. Near infrared photoimmunotherapy in the treatment of disseminated peritoneal ovarian cancer. Mol. Cancer Ther. 2015, 14, 141–150. [Google Scholar] [CrossRef] [PubMed]
  24. Ito, K.; Mitsunaga, M.; Nishimura, T.; Kobayashi, H.; Tajiri, H. Combination photoimmunotherapy with monoclonal antibodies recognizing different epitopes of human epidermal growth factor receptor 2: An assessment of phototherapeutic effect based on fluorescence molecular imaging. Oncotarget 2016, 7, 14143–14152. [Google Scholar] [CrossRef]
  25. Fukushima, H.; Kato, T.; Furusawa, A.; Okada, R.; Wakiyama, H.; Furumoto, H.; Okuyama, S.; Kondo, E.; Choyke, P.L.; Kobayashi, H. Intercellular adhesion molecule-1-targeted near-infrared photoimmunotherapy of triple-negative breast cancer. Cancer Sci. 2022, 113, 3180–3192. [Google Scholar] [CrossRef]
  26. Fukushima, H.; Takao, S.; Furusawa, A.; Valera Romero, V.; Gurram, S.; Kato, T.; Okuyama, S.; Kano, M.; Choyke, P.L.; Kobayashi, H. Near-infrared photoimmunotherapy targeting Nectin-4 in a preclinical model of bladder cancer. Cancer Lett. 2024, 585, 216606. [Google Scholar] [CrossRef]
  27. Nishinaga, Y.; Sato, K.; Yasui, H.; Taki, S.; Takahashi, K.; Shimizu, M.; Endo, R.; Koike, C.; Kuramoto, N.; Nakamura, S.; et al. Targeted Phototherapy for Malignant Pleural Mesothelioma: Near-Infrared Photoimmunotherapy Targeting Podoplanin. Cells 2020, 9, 1019. [Google Scholar] [CrossRef]
  28. Nishimura, T.; Mitsunaga, M.; Sawada, R.; Saruta, M.; Kobayashi, H.; Matsumoto, N.; Kanke, T.; Yanai, H.; Nakamura, K. Photoimmunotherapy targeting biliary-pancreatic cancer with humanized anti-TROP2 antibody. Cancer Med. 2019, 8, 7781–7792. [Google Scholar] [CrossRef]
  29. Nagaya, T.; Nakamura, Y.; Okuyama, S.; Ogata, F.; Maruoka, Y.; Choyke, P.L.; Kobayashi, H. Near-Infrared Photoimmunotherapy Targeting Prostate Cancer with Prostate-Specific Membrane Antigen (PSMA) Antibody. Mol. Cancer Res. 2017, 15, 1153–1162. [Google Scholar] [CrossRef]
  30. Nagaya, T.; Nakamura, Y.; Sato, K.; Harada, T.; Choyke, P.L.; Kobayashi, H. Near infrared photoimmunotherapy of B-cell lymphoma. Mol. Oncol. 2016, 10, 1404–1414. [Google Scholar] [CrossRef]
  31. Furusawa, A.; Okada, R.; Inagaki, F.; Wakiyama, H.; Kato, T.; Furumoto, H.; Fukushima, H.; Okuyama, S.; Choyke, P.L.; Kobayashi, H. CD29 targeted near-infrared photoimmunotherapy (NIR-PIT) in the treatment of a pigmented melanoma model. Oncoimmunology 2022, 11, 2019922. [Google Scholar] [CrossRef]
  32. Kiss, B.; van den Berg, N.S.; Ertsey, R.; McKenna, K.; Mach, K.E.; Zhang, C.A.; Volkmer, J.P.; Weissman, I.L.; Rosenthal, E.L.; Liao, J.C. CD47-Targeted Near-Infrared Photoimmunotherapy for Human Bladder Cancer. Clin. Cancer Res. 2019, 25, 3561–3571. [Google Scholar] [CrossRef]
  33. Jing, H.; Weidensteiner, C.; Reichardt, W.; Gaedicke, S.; Zhu, X.; Grosu, A.L.; Kobayashi, H.; Niedermann, G. Imaging and Selective Elimination of Glioblastoma Stem Cells with Theranostic Near-Infrared-Labeled CD133-Specific Antibodies. Theranostics 2016, 6, 862–874. [Google Scholar] [CrossRef]
  34. Hiroshima, Y.; Maawy, A.; Zhang, Y.; Guzman, M.G.; Heim, R.; Makings, L.; Luiken, G.A.; Kobayashi, H.; Tanaka, K.; Endo, I.; et al. Photoimmunotherapy Inhibits Tumor Recurrence After Surgical Resection on a Pancreatic Cancer Patient-Derived Orthotopic Xenograft (PDOX) Nude Mouse Model. Ann. Surg. Oncol. 2015, 22 (Suppl. S3), 1469–1474. [Google Scholar] [CrossRef] [PubMed]
  35. Maawy, A.A.; Hiroshima, Y.; Zhang, Y.; Heim, R.; Makings, L.; Garcia-Guzman, M.; Luiken, G.A.; Kobayashi, H.; Hoffman, R.M.; Bouvet, M. Near infra-red photoimmunotherapy with anti-CEA-IR700 results in extensive tumor lysis and a significant decrease in tumor burden in orthotopic mouse models of pancreatic cancer. PLoS ONE 2015, 10, e0121989. [Google Scholar] [CrossRef] [PubMed]
  36. Hollandsworth, H.M.; Amirfakhri, S.; Filemoni, F.; Molnar, J.; Hoffman, R.M.; Yazaki, P.; Bouvet, M. Near-infrared photoimmunotherapy is effective treatment for colorectal cancer in orthotopic nude-mouse models. PLoS ONE 2020, 15, e0234643. [Google Scholar] [CrossRef] [PubMed]
  37. Polikarpov, D.M.; Campbell, D.H.; Lund, M.E.; Lu, Y.; Lu, Y.; Wu, J.; Walsh, B.J.; Zvyagin, A.V.; Gillatt, D.A. The feasibility of Miltuximab(R)-IRDye700DX-mediated photoimmunotherapy of solid tumors. Photodiagn. Photodyn. Ther. 2020, 32, 102064. [Google Scholar] [CrossRef]
  38. Hanaoka, H.; Nagaya, T.; Sato, K.; Nakamura, Y.; Watanabe, R.; Harada, T.; Gao, W.; Feng, M.; Phung, Y.; Kim, I.; et al. Glypican-3 targeted human heavy chain antibody as a drug carrier for hepatocellular carcinoma therapy. Mol. Pharm. 2015, 12, 2151–2157. [Google Scholar] [CrossRef]
  39. Hanaoka, H.; Nakajima, T.; Sato, K.; Watanabe, R.; Phung, Y.; Gao, W.; Harada, T.; Kim, I.; Paik, C.H.; Choyke, P.L.; et al. Photoimmunotherapy of hepatocellular carcinoma-targeting Glypican-3 combined with nanosized albumin-bound paclitaxel. Nanomedicine 2015, 10, 1139–1147. [Google Scholar] [CrossRef]
  40. Nagaya, T.; Nakamura, Y.; Sato, K.; Zhang, Y.F.; Ni, M.; Choyke, P.L.; Ho, M.; Kobayashi, H. Near infrared photoimmunotherapy with an anti-mesothelin antibody. Oncotarget 2016, 7, 23361–23369. [Google Scholar] [CrossRef]
  41. Krishnan, H.; Rayes, J.; Miyashita, T.; Ishii, G.; Retzbach, E.P.; Sheehan, S.A.; Takemoto, A.; Chang, Y.W.; Yoneda, K.; Asai, J.; et al. Podoplanin: An emerging cancer biomarker and therapeutic target. Cancer Sci. 2018, 109, 1292–1299. [Google Scholar] [CrossRef]
  42. Quintanilla, M.; Montero-Montero, L.; Renart, J.; Martin-Villar, E. Podoplanin in Inflammation and Cancer. Int. J. Mol. Sci. 2019, 20, 707. [Google Scholar] [CrossRef]
  43. Molina, M.A.; Codony-Servat, J.; Albanell, J.; Rojo, F.; Arribas, J.; Baselga, J. Trastuzumab (herceptin), a humanized anti-Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells. Cancer Res. 2001, 61, 4744–4749. [Google Scholar] [PubMed]
  44. Franklin, M.C.; Carey, K.D.; Vajdos, F.F.; Leahy, D.J.; de Vos, A.M.; Sliwkowski, M.X. Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell 2004, 5, 317–328. [Google Scholar] [CrossRef] [PubMed]
  45. Nakajima, K.; Ogawa, M. Phototoxicity in near-infrared photoimmunotherapy is influenced by the subcellular localization of antibody-IR700. Photodiagn. Photodyn. Ther. 2020, 31, 101926. [Google Scholar] [CrossRef] [PubMed]
  46. Ogawa, M.; Takakura, H. Photoimmunotherapy: A new cancer treatment using photochemical reactions. Bioorg. Med. Chem. 2021, 43, 116274. [Google Scholar] [CrossRef]
  47. Yu, J.; Song, Y.; Tian, W. How to select IgG subclasses in developing anti-tumor therapeutic antibodies. J. Hematol. Oncol. 2020, 13, 45. [Google Scholar] [CrossRef]
  48. Wang, Z.; Wang, G.; Lu, H.; Li, H.; Tang, M.; Tong, A. Development of therapeutic antibodies for the treatment of diseases. Mol. Biomed. 2022, 3, 35. [Google Scholar] [CrossRef]
  49. 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] [CrossRef]
  50. Tijink, B.M.; Buter, J.; de Bree, R.; Giaccone, G.; Lang, M.S.; Staab, A.; Leemans, C.R.; van Dongen, G.A. A phase I dose escalation study with anti-CD44v6 bivatuzumab mertansine in patients with incurable squamous cell carcinoma of the head and neck or esophagus. Clin. Cancer Res. 2006, 12, 6064–6072. [Google Scholar] [CrossRef]
  51. Lyu, X.; Zhao, Q.; Hui, J.; Wang, T.; Lin, M.; Wang, K.; Zhang, J.; Shentu, J.; Dalby, P.A.; Zhang, H.; et al. The global landscape of approved antibody therapies. Antib. Ther. 2022, 5, 233–257. [Google Scholar] [CrossRef]
  52. Wichert, S.; Juliusson, G.; Johansson, A.; Sonesson, E.; Teige, I.; Wickenberg, A.T.; Frendeus, B.; Korsgren, M.; Hansson, M. A single-arm, open-label, phase 2 clinical trial evaluating disease response following treatment with BI-505, a human anti-intercellular adhesion molecule-1 monoclonal antibody, in patients with smoldering multiple myeloma. PLoS ONE 2017, 12, e0171205. [Google Scholar] [CrossRef] [PubMed]
  53. Okada, R.; Kato, T.; Furusawa, A.; Inagaki, F.; Wakiyama, H.; Fujimura, D.; Okuyama, S.; Furumoto, H.; Fukushima, H.; Choyke, P.L.; et al. Selection of antibody and light exposure regimens alters therapeutic effects of EGFR-targeted near-infrared photoimmunotherapy. Cancer Immunol. Immunother. 2022, 71, 1877–1887. [Google Scholar] [CrossRef] [PubMed]
  54. Mao, C.; Qu, P.; Miley, M.J.; Zhao, Y.; Li, Z.; Ming, X. P-glycoprotein targeted photodynamic therapy of chemoresistant tumors using recombinant Fab fragment conjugates. Biomater. Sci. 2018, 6, 3063–3074. [Google Scholar] [CrossRef] [PubMed]
  55. Watanabe, R.; Hanaoka, H.; Sato, K.; Nagaya, T.; Harada, T.; Mitsunaga, M.; Kim, I.; Paik, C.H.; Wu, A.M.; Choyke, P.L.; et al. Photoimmunotherapy targeting prostate-specific membrane antigen: Are antibody fragments as effective as antibodies? J. Nucl. Med. 2015, 56, 140–144. [Google Scholar] [CrossRef]
  56. Okada, R.; Furusawa, A.; Vermeer, D.W.; Inagaki, F.; Wakiyama, H.; Kato, T.; Nagaya, T.; Choyke, P.L.; Spanos, W.C.; Allen, C.T.; et al. Near-infrared photoimmunotherapy targeting human-EGFR in a mouse tumor model simulating current and future clinical trials. eBioMedicine 2021, 67, 103345. [Google Scholar] [CrossRef]
  57. Ullrich, A.; Coussens, L.; Hayflick, J.S.; Dull, T.J.; Gray, A.; Tam, A.W.; Lee, J.; Yarden, Y.; Libermann, T.A.; Schlessinger, J.; et al. Human epidermal growth factor receptor cDNA sequence and aberrant expression of the amplified gene in A431 epidermoid carcinoma cells. Nature 1984, 309, 418–425. [Google Scholar] [CrossRef]
  58. Bajaj, M.; Waterfield, M.D.; Schlessinger, J.; Taylor, W.R.; Blundell, T. On the tertiary structure of the extracellular domains of the epidermal growth factor and insulin receptors. Biochim. Biophys. Acta 1987, 916, 220–226. [Google Scholar] [CrossRef]
  59. Lax, I.; Johnson, A.; Howk, R.; Sap, J.; Bellot, F.; Winkler, M.; Ullrich, A.; Vennstrom, B.; Schlessinger, J.; Givol, D. Chicken epidermal growth factor (EGF) receptor: cDNA cloning, expression in mouse cells, and differential binding of EGF and transforming growth factor alpha. Mol. Cell Biol. 1988, 8, 1970–1978. [Google Scholar] [CrossRef]
  60. Ward, C.W.; Hoyne, P.A.; Flegg, R.H. Insulin and epidermal growth factor receptors contain the cysteine repeat motif found in the tumor necrosis factor receptor. Proteins 1995, 22, 141–153. [Google Scholar] [CrossRef]
  61. Dawson, J.P.; Berger, M.B.; Lin, C.C.; Schlessinger, J.; Lemmon, M.A.; Ferguson, K.M. Epidermal growth factor receptor dimerization and activation require ligand-induced conformational changes in the dimer interface. Mol. Cell Biol. 2005, 25, 7734–7742. [Google Scholar] [CrossRef] [PubMed]
  62. Cai, W.Q.; Zeng, L.S.; Wang, L.F.; Wang, Y.Y.; Cheng, J.T.; Zhang, Y.; Han, Z.W.; Zhou, Y.; Huang, S.L.; Wang, X.W.; et al. The Latest Battles Between EGFR Monoclonal Antibodies and Resistant Tumor Cells. Front. Oncol. 2020, 10, 1249. [Google Scholar] [CrossRef] [PubMed]
  63. Voigt, M.; Braig, F.; Gothel, M.; Schulte, A.; Lamszus, K.; Bokemeyer, C.; Binder, M. Functional dissection of the epidermal growth factor receptor epitopes targeted by panitumumab and cetuximab. Neoplasia 2012, 14, 1023–1031. [Google Scholar] [CrossRef] [PubMed]
  64. Li, S.; Kussie, P.; Ferguson, K.M. Structural basis for EGF receptor inhibition by the therapeutic antibody IMC-11F8. Structure 2008, 16, 216–227. [Google Scholar] [CrossRef]
  65. Sickmier, E.A.; Kurzeja, R.J.; Michelsen, K.; Vazir, M.; Yang, E.; Tasker, A.S. The Panitumumab EGFR Complex Reveals a Binding Mechanism That Overcomes Cetuximab Induced Resistance. PLoS ONE 2016, 11, e0163366. [Google Scholar] [CrossRef]
  66. Bagchi, A.; Haidar, J.N.; Eastman, S.W.; Vieth, M.; Topper, M.; Iacolina, M.D.; Walker, J.M.; Forest, A.; Shen, Y.; Novosiadly, R.D.; et al. Molecular Basis for Necitumumab Inhibition of EGFR Variants Associated with Acquired Cetuximab Resistance. Mol. Cancer Ther. 2018, 17, 521–531. [Google Scholar] [CrossRef]
  67. Burgess, A.W.; Cho, H.S.; Eigenbrot, C.; Ferguson, K.M.; Garrett, T.P.; Leahy, D.J.; Lemmon, M.A.; Sliwkowski, M.X.; Ward, C.W.; Yokoyama, S. An open-and-shut case? Recent insights into the activation of EGF/ErbB receptors. Mol. Cell 2003, 12, 541–552. [Google Scholar] [CrossRef]
  68. Kovacs, E.; Zorn, J.A.; Huang, Y.; Barros, T.; Kuriyan, J. A structural perspective on the regulation of the epidermal growth factor receptor. Annu. Rev. Biochem. 2015, 84, 739–764. [Google Scholar] [CrossRef]
  69. Yamashita, S.; Kojima, M.; Onda, N.; Yoshida, T.; Shibutani, M. Trastuzumab-based near-infrared photoimmunotherapy in xenograft mouse of breast cancer. Cancer Med. 2023, 12, 4579–4589. [Google Scholar] [CrossRef]
  70. Hirata, H.; Kuwatani, M.; Nakajima, K.; Kodama, Y.; Yoshikawa, Y.; Ogawa, M.; Sakamoto, N. Near-infrared photoimmunotherapy (NIR-PIT) on cholangiocarcinoma using a novel catheter device with light emitting diodes. Cancer Sci. 2021, 112, 828–838. [Google Scholar] [CrossRef]
  71. Hartmans, E.; Linssen, M.D.; Sikkens, C.; Levens, A.; Witjes, M.J.H.; van Dam, G.M.; Nagengast, W.B. Tyrosine kinase inhibitor induced growth factor receptor upregulation enhances the efficacy of near-infrared targeted photodynamic therapy in esophageal adenocarcinoma cell lines. Oncotarget 2017, 8, 29846–29856. [Google Scholar] [CrossRef]
  72. Liu, Y.; Ren, S.; Xie, L.; Cui, C.; Xing, Y.; Liu, C.; Cao, B.; Yang, F.; Li, Y.; Chen, X.; et al. Mutation of N-linked glycosylation at Asn548 in CD133 decreases its ability to promote hepatoma cell growth. Oncotarget 2015, 6, 20650–20660. [Google Scholar] [CrossRef]
  73. Corbeil, D.; Marzesco, A.M.; Wilsch-Brauninger, M.; Huttner, W.B. The intriguing links between prominin-1 (CD133), cholesterol-based membrane microdomains, remodeling of apical plasma membrane protrusions, extracellular membrane particles, and (neuro)epithelial cell differentiation. FEBS Lett. 2010, 584, 1659–1664. [Google Scholar] [CrossRef] [PubMed]
  74. Karbanova, J.; Lorico, A.; Bornhauser, M.; Corbeil, D.; Fargeas, C.A. Prominin-1/CD133: Lipid Raft Association, Detergent Resistance, and Immunodetection. Stem Cells Transl. Med. 2018, 7, 155–160. [Google Scholar] [CrossRef] [PubMed]
  75. Miraglia, S.; Godfrey, W.; Yin, A.H.; Atkins, K.; Warnke, R.; Holden, J.T.; Bray, R.A.; Waller, E.K.; Buck, D.W. A novel five-transmembrane hematopoietic stem cell antigen: Isolation, characterization, and molecular cloning. Blood 1997, 90, 5013–5021. [Google Scholar] [CrossRef]
  76. Shmelkov, S.V.; St Clair, R.; Lyden, D.; Rafii, S. AC133/CD133/Prominin-1. Int. J. Biochem. Cell Biol. 2005, 37, 715–719. [Google Scholar] [CrossRef]
  77. Swaminathan, S.K.; Roger, E.; Toti, U.; Niu, L.; Ohlfest, J.R.; Panyam, J. CD133-targeted paclitaxel delivery inhibits local tumor recurrence in a mouse model of breast cancer. J. Control Release 2013, 171, 280–287. [Google Scholar] [CrossRef]
  78. Weng, D.; Jin, X.; Qin, S.; Lan, X.; Chen, C.; Sun, X.; She, X.; Dong, C.; An, R. Radioimmunotherapy for CD133(+) colonic cancer stem cells inhibits tumor development in nude mice. Oncotarget 2017, 8, 44004–44014. [Google Scholar] [CrossRef]
  79. Damek-Poprawa, M.; Volgina, A.; Korostoff, J.; Sollecito, T.P.; Brose, M.S.; O’Malley, B.W., Jr.; Akintoye, S.O.; DiRienzo, J.M. Targeted inhibition of CD133+ cells in oral cancer cell lines. J. Dent. Res. 2011, 90, 638–645. [Google Scholar] [CrossRef]
  80. Stamenkovic, I.; Amiot, M.; Pesando, J.M.; Seed, B. A lymphocyte molecule implicated in lymph node homing is a member of the cartilage link protein family. Cell 1989, 56, 1057–1062. [Google Scholar] [CrossRef]
  81. Weng, X.; Maxwell-Warburton, S.; Hasib, A.; Ma, L.; Kang, L. The membrane receptor CD44: Novel insights into metabolism. Trends Endocrinol. Metab. 2022, 33, 318–332. [Google Scholar] [CrossRef]
  82. Ponta, H.; Sherman, L.; Herrlich, P.A. CD44: From adhesion molecules to signalling regulators. Nat. Rev. Mol. Cell Biol. 2003, 4, 33–45. [Google Scholar] [CrossRef]
  83. Kasper, M.; Gunthert, U.; Dall, P.; Kayser, K.; Schuh, D.; Haroske, G.; Muller, M. Distinct expression patterns of CD44 isoforms during human lung development and in pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 1995, 13, 648–656. [Google Scholar] [CrossRef] [PubMed]
  84. Thapa, R.; Wilson, G.D. The Importance of CD44 as a Stem Cell Biomarker and Therapeutic Target in Cancer. Stem Cells Int. 2016, 2016, 2087204. [Google Scholar] [CrossRef] [PubMed]
  85. Zoller, M. CD44: Can a cancer-initiating cell profit from an abundantly expressed molecule? Nat. Rev. Cancer 2011, 11, 254–267. [Google Scholar] [CrossRef] [PubMed]
  86. Lusche, D.F.; Wessels, D.J.; Reis, R.J.; Forrest, C.C.; Thumann, A.R.; Soll, D.R. New monoclonal antibodies that recognize an unglycosylated, conserved, extracellular region of CD44 in vitro and in vivo, and can block tumorigenesis. PLoS ONE 2021, 16, e0250175. [Google Scholar] [CrossRef]
  87. Li, L.; Hao, X.; Qin, J.; Tang, W.; He, F.; Smith, A.; Zhang, M.; Simeone, D.M.; Qiao, X.T.; Chen, Z.N.; et al. Antibody against CD44s inhibits pancreatic tumor initiation and postradiation recurrence in mice. Gastroenterology 2014, 146, 1108–1118. [Google Scholar] [CrossRef]
  88. Mikecz, K.; Brennan, F.R.; Kim, J.H.; Glant, T.T. Anti-CD44 treatment abrogates tissue oedema and leukocyte infiltration in murine arthritis. Nat. Med. 1995, 1, 558–563. [Google Scholar] [CrossRef]
  89. Uchino, M.; Kojima, H.; Wada, K.; Imada, M.; Onoda, F.; Satofuka, H.; Utsugi, T.; Murakami, Y. Nuclear beta-catenin and CD44 upregulation characterize invasive cell populations in non-aggressive MCF-7 breast cancer cells. BMC Cancer 2010, 10, 414. [Google Scholar] [CrossRef]
  90. Nagaya, T.; Friedman, J.; Maruoka, Y.; Ogata, F.; Okuyama, S.; Clavijo, P.E.; Choyke, P.L.; Allen, C.; Kobayashi, H. Host Immunity Following Near-Infrared Photoimmunotherapy Is Enhanced with PD-1 Checkpoint Blockade to Eradicate Established Antigenic Tumors. Cancer Immunol. Res. 2019, 7, 401–413. [Google Scholar] [CrossRef]
  91. Wakiyama, H.; Furusawa, A.; Okada, R.; Inagaki, F.; Kato, T.; Maruoka, Y.; Choyke, P.L.; Kobayashi, H. Increased Immunogenicity of a Minimally Immunogenic Tumor after Cancer-Targeting Near Infrared Photoimmunotherapy. Cancers 2020, 12, 3747. [Google Scholar] [CrossRef]
  92. Maruoka, Y.; Furusawa, A.; Okada, R.; Inagaki, F.; Fujimura, D.; Wakiyama, H.; Kato, T.; Nagaya, T.; Choyke, P.L.; Kobayashi, H. Near-Infrared Photoimmunotherapy Combined with CTLA4 Checkpoint Blockade in Syngeneic Mouse Cancer Models. Vaccines 2020, 8, 528. [Google Scholar] [CrossRef]
  93. Maruoka, Y.; Furusawa, A.; Okada, R.; Inagaki, F.; Wakiyama, H.; Kato, T.; Nagaya, T.; Choyke, P.L.; Kobayashi, H. Interleukin-15 after Near-Infrared Photoimmunotherapy (NIR-PIT) Enhances T Cell Response against Syngeneic Mouse Tumors. Cancers 2020, 12, 2575. [Google Scholar] [CrossRef]
  94. Nagaya, T.; Nakamura, Y.; Okuyama, S.; Ogata, F.; Maruoka, Y.; Choyke, P.L.; Allen, C.; Kobayashi, H. Syngeneic Mouse Models of Oral Cancer Are Effectively Targeted by Anti-CD44-Based NIR-PIT. Mol. Cancer Res. 2017, 15, 1667–1677. [Google Scholar] [CrossRef] [PubMed]
  95. Stanners, C.P.; DeMarte, L.; Rojas, M.; Gold, P.; Fuks, A. Opposite functions for two classes of genes of the human carcinoembryonic antigen family. Tumour Biol. 1995, 16, 23–31. [Google Scholar] [CrossRef] [PubMed]
  96. Gold, P.; Freedman, S.O. Demonstration of Tumor-Specific Antigens in Human Colonic Carcinomata by Immunological Tolerance and Absorption Techniques. J. Exp. Med. 1965, 121, 439–462. [Google Scholar] [CrossRef] [PubMed]
  97. 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]
  98. Berinstein, N.L. Carcinoembryonic antigen as a target for therapeutic anticancer vaccines: A review. J. Clin. Oncol. 2002, 20, 2197–2207. [Google Scholar] [CrossRef]
  99. Abdul-Wahid, A.; Huang, E.H.; Cydzik, M.; Bolewska-Pedyczak, E.; Gariepy, J. The carcinoembryonic antigen IgV-like N domain plays a critical role in the implantation of metastatic tumor cells. Mol. Oncol. 2014, 8, 337–350. [Google Scholar] [CrossRef]
  100. Hefta, S.A.; Hefta, L.J.; Lee, T.D.; Paxton, R.J.; Shively, J.E. Carcinoembryonic antigen is anchored to membranes by covalent attachment to a glycosylphosphatidylinositol moiety: Identification of the ethanolamine linkage site. Proc. Natl. Acad. Sci. USA 1988, 85, 4648–4652. [Google Scholar] [CrossRef]
  101. Hammarstrom, S. The carcinoembryonic antigen (CEA) family: Structures, suggested functions and expression in normal and malignant tissues. Semin. Cancer Biol. 1999, 9, 67–81. [Google Scholar] [CrossRef]
  102. Molina, R.; Barak, V.; van Dalen, A.; Duffy, M.J.; Einarsson, R.; Gion, M.; Goike, H.; Lamerz, R.; Nap, M.; Soletormos, G.; et al. Tumor markers in breast cancer- European Group on Tumor Markers recommendations. Tumour Biol. 2005, 26, 281–293. [Google Scholar] [CrossRef] [PubMed]
  103. Gansauge, S.; Gansauge, F.; Beger, H.G. Molecular oncology in pancreatic cancer. J. Mol. Med. 1996, 74, 313–320. [Google Scholar] [CrossRef] [PubMed]
  104. Grunnet, M.; Sorensen, J.B. Carcinoembryonic antigen (CEA) as tumor marker in lung cancer. Lung Cancer 2012, 76, 138–143. [Google Scholar] [CrossRef]
  105. Juweid, M.; Sharkey, R.M.; Behr, T.; Swayne, L.C.; Rubin, A.D.; Herskovic, T.; Hanley, D.; Markowitz, A.; Dunn, R.; Siegel, J.; et al. Improved detection of medullary thyroid cancer with radiolabeled antibodies to carcinoembryonic antigen. J. Clin. Oncol. 1996, 14, 1209–1217. [Google Scholar] [CrossRef] [PubMed]
  106. Goldenberg, D.M.; DeLand, F.; Kim, E.; Bennett, S.; Primus, F.J.; van Nagell, J.R., Jr.; Estes, N.; DeSimone, P.; Rayburn, P. Use of radiolabeled antibodies to carcinoembryonic antigen for the detection and localization of diverse cancers by external photoscanning. N. Engl. J. Med. 1978, 298, 1384–1386. [Google Scholar] [CrossRef] [PubMed]
  107. Campos-da-Paz, M.; Dorea, J.G.; Galdino, A.S.; Lacava, Z.G.M.; de Fatima Menezes Almeida Santos, M. 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]
  108. Shinmi, D.; Nakano, R.; Mitamura, K.; Suzuki-Imaizumi, M.; Iwano, J.; Isoda, Y.; Enokizono, J.; Shiraishi, Y.; Arakawa, E.; Tomizuka, K.; et al. Novel anticarcinoembryonic antigen antibody-drug conjugate has antitumor activity in the existence of soluble antigen. Cancer Med. 2017, 6, 798–808. [Google Scholar] [CrossRef]
  109. Dotan, E.; Cohen, S.J.; Starodub, A.N.; Lieu, C.H.; Messersmith, W.A.; Simpson, P.S.; Guarino, M.J.; Marshall, J.L.; Goldberg, R.M.; Hecht, J.R.; et al. Phase I/II Trial of Labetuzumab Govitecan (Anti-CEACAM5/SN-38 Antibody-Drug Conjugate) in Patients With Refractory or Relapsing Metastatic Colorectal Cancer. J. Clin. Oncol. 2017, 35, 3338–3346. [Google Scholar] [CrossRef]
  110. Conaghan, P.; Ashraf, S.; Tytherleigh, M.; Wilding, J.; Tchilian, E.; Bicknell, D.; Mortensen, N.J.; Bodmer, W. Targeted killing of colorectal cancer cell lines by a humanised IgG1 monoclonal antibody that binds to membrane-bound carcinoembryonic antigen. Br. J. Cancer 2008, 98, 1217–1225. [Google Scholar] [CrossRef]
  111. Durbin, H.; Young, S.; Stewart, L.M.; Wrba, F.; Rowan, A.J.; Snary, D.; Bodmer, W.F. An epitope on carcinoembryonic antigen defined by the clinically relevant antibody PR1A3. Proc. Natl. Acad. Sci. USA 1994, 91, 4313–4317. [Google Scholar] [CrossRef]
  112. Shirasu, N.; Yamada, H.; Shibaguchi, H.; Kuroki, M.; Kuroki, M. Potent and specific antitumor effect of CEA-targeted photoimmunotherapy. Int. J. Cancer 2014, 135, 2697–2710. [Google Scholar] [CrossRef]
  113. Maawy, A.A.; Hiroshima, Y.; Zhang, Y.; Garcia-Guzman, M.; Luiken, G.A.; Kobayashi, H.; Hoffman, R.M.; Bouvet, M. Photoimmunotherapy lowers recurrence after pancreatic cancer surgery in orthotopic nude mouse models. J. Surg. Res. 2015, 197, 5–11. [Google Scholar] [CrossRef] [PubMed]
  114. Ghosh, A.; Heston, W.D. Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancer. J. Cell Biochem. 2004, 91, 528–539. [Google Scholar] [CrossRef] [PubMed]
  115. Barinka, C.; Sacha, P.; Sklenar, J.; Man, P.; Bezouska, K.; Slusher, B.S.; Konvalinka, J. Identification of the N-glycosylation sites on glutamate carboxypeptidase II necessary for proteolytic activity. Protein Sci. 2004, 13, 1627–1635. [Google Scholar] [CrossRef] [PubMed]
  116. Ghosh, A.; Heston, W.D. Effect of carbohydrate moieties on the folate hydrolysis activity of the prostate specific membrane antigen. Prostate 2003, 57, 140–151. [Google Scholar] [CrossRef]
  117. Schulke, N.; Varlamova, O.A.; Donovan, G.P.; Ma, D.; Gardner, J.P.; Morrissey, D.M.; Arrigale, R.R.; Zhan, C.; Chodera, A.J.; Surowitz, K.G.; et al. The homodimer of prostate-specific membrane antigen is a functional target for cancer therapy. Proc. Natl. Acad. Sci. USA 2003, 100, 12590–12595. [Google Scholar] [CrossRef]
  118. Israeli, R.S.; Powell, C.T.; Fair, W.R.; Heston, W.D. Molecular cloning of a complementary DNA encoding a prostate-specific membrane antigen. Cancer Res. 1993, 53, 227–230. [Google Scholar]
  119. Rajasekaran, A.K.; Anilkumar, G.; Christiansen, J.J. Is prostate-specific membrane antigen a multifunctional protein? Am. J. Physiol. Cell Physiol. 2005, 288, C975–C981. [Google Scholar] [CrossRef]
  120. Feneley, M.R.; Jan, H.; Granowska, M.; Mather, S.J.; Ellison, D.; Glass, J.; Coptcoat, M.; Kirby, R.S.; Ogden, C.; Oliver, R.T.; et al. Imaging with prostate-specific membrane antigen (PSMA) in prostate cancer. Prostate Cancer Prostatic Dis. 2000, 3, 47–52. [Google Scholar] [CrossRef]
  121. Elgamal, A.A.; Troychak, M.J.; Murphy, G.P. ProstaScint scan may enhance identification of prostate cancer recurrences after prostatectomy, radiation, or hormone therapy: Analysis of 136 scans of 100 patients. Prostate 1998, 37, 261–269. [Google Scholar] [CrossRef]
  122. Kahn, D.; Austin, J.C.; Maguire, R.T.; Miller, S.J.; Gerstbrein, J.; Williams, R.D. A phase II study of [90Y] yttrium-capromab pendetide in the treatment of men with prostate cancer recurrence following radical prostatectomy. Cancer Biother. Radiopharm. 1999, 14, 99–111. [Google Scholar] [CrossRef] [PubMed]
  123. Troyer, J.K.; Beckett, M.L.; Wright, G.L., Jr. Location of prostate-specific membrane antigen in the LNCaP prostate carcinoma cell line. Prostate 1997, 30, 232–242. [Google Scholar] [CrossRef]
  124. Liu, H.; Moy, P.; Kim, S.; Xia, Y.; Rajasekaran, A.; Navarro, V.; Knudsen, B.; Bander, N.H. Monoclonal antibodies to the extracellular domain of prostate-specific membrane antigen also react with tumor vascular endothelium. Cancer Res. 1997, 57, 3629–3634. [Google Scholar]
  125. Hsieh, H.H.; Kuo, W.Y.; Lin, J.J.; Chen, H.S.; Hsu, H.J.; Wu, C.Y. Tumor-Targeting Ability of Novel Anti-Prostate-Specific Membrane Antigen Antibodies. ACS Omega 2022, 7, 31529–31537. [Google Scholar] [CrossRef]
  126. Nakajima, K.; Miyazaki, F.; Terada, K.; Takakura, H.; Suzuki, M.; Ogawa, M. Comparison of low-molecular-weight ligand and whole antibody in prostate-specific membrane antigen targeted near-infrared photoimmunotherapy. Int. J. Pharm. 2021, 609, 121135. [Google Scholar] [CrossRef]
  127. Rishi, A.K.; Joyce-Brady, M.; Fisher, J.; Dobbs, L.G.; Floros, J.; VanderSpek, J.; Brody, J.S.; Williams, M.C. Cloning, characterization, and development expression of a rat lung alveolar type I cell gene in embryonic endodermal and neural derivatives. Dev. Biol. 1995, 167, 294–306. [Google Scholar] [CrossRef]
  128. Breiteneder-Geleff, S.; Matsui, K.; Soleiman, A.; Meraner, P.; Poczewski, H.; Kalt, R.; Schaffner, G.; Kerjaschki, D. Podoplanin, novel 43-kd membrane protein of glomerular epithelial cells, is down-regulated in puromycin nephrosis. Am. J. Pathol. 1997, 151, 1141–1152. [Google Scholar]
  129. Breiteneder-Geleff, S.; Soleiman, A.; Kowalski, H.; Horvat, R.; Amann, G.; Kriehuber, E.; Diem, K.; Weninger, W.; Tschachler, E.; Alitalo, K.; et al. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: Podoplanin as a specific marker for lymphatic endothelium. Am. J. Pathol. 1999, 154, 385–394. [Google Scholar] [CrossRef]
  130. Kaneko, M.K.; Kato, Y.; Kitano, T.; Osawa, M. Conservation of a platelet activating domain of Aggrus/podoplanin as a platelet aggregation-inducing factor. Gene 2006, 378, 52–57. [Google Scholar] [CrossRef]
  131. Suzuki, H.; Kaneko, M.K.; Kato, Y. Roles of Podoplanin in Malignant Progression of Tumor. Cells 2022, 11, 575. [Google Scholar] [CrossRef]
  132. Kato, Y.; Kaneko, M.K.; Kunita, A.; Ito, H.; Kameyama, A.; Ogasawara, S.; Matsuura, N.; Hasegawa, Y.; Suzuki-Inoue, K.; Inoue, O.; et al. Molecular analysis of the pathophysiological binding of the platelet aggregation-inducing factor podoplanin to the C-type lectin-like receptor CLEC-2. Cancer Sci. 2008, 99, 54–61. [Google Scholar] [CrossRef]
  133. Nakazawa, Y.; Takagi, S.; Sato, S.; Oh-hara, T.; Koike, S.; Takami, M.; Arai, H.; Fujita, N. Prevention of hematogenous metastasis by neutralizing mice and its chimeric anti-Aggrus/podoplanin antibodies. Cancer Sci. 2011, 102, 2051–2057. [Google Scholar] [CrossRef] [PubMed]
  134. Takagi, S.; Sato, S.; Oh-hara, T.; Takami, M.; Koike, S.; Mishima, Y.; Hatake, K.; Fujita, N. Platelets promote tumor growth and metastasis via direct interaction between Aggrus/podoplanin and CLEC-2. PLoS ONE 2013, 8, e73609. [Google Scholar] [CrossRef]
  135. Ukaji, T.; Takemoto, A.; Katayama, R.; Takeuchi, K.; Fujita, N. A safety study of newly generated anti-podoplanin-neutralizing antibody in cynomolgus monkey (Macaca fascicularis). Oncotarget 2018, 9, 33322–33336. [Google Scholar] [CrossRef] [PubMed]
  136. Shiina, S.; Ohno, M.; Ohka, F.; Kuramitsu, S.; Yamamichi, A.; Kato, A.; Motomura, K.; Tanahashi, K.; Yamamoto, T.; Watanabe, R.; et al. CAR T Cells Targeting Podoplanin Reduce Orthotopic Glioblastomas in Mouse Brains. Cancer Immunol. Res. 2016, 4, 259–268. [Google Scholar] [CrossRef] [PubMed]
  137. Kato, T.; Furusawa, A.; Okada, R.; Inagaki, F.; Wakiyama, H.; Furumoto, H.; Fukushima, H.; Okuyama, S.; Choyke, P.L.; Kobayashi, H. Near-Infrared Photoimmunotherapy Targeting Podoplanin-Expressing Cancer Cells and Cancer-Associated Fibroblasts. Mol. Cancer Ther. 2023, 22, 75–88. [Google Scholar] [CrossRef]
  138. Scott, D.W.; Patel, R.P. Endothelial heterogeneity and adhesion molecules N-glycosylation: Implications in leukocyte trafficking in inflammation. Glycobiology 2013, 23, 622–633. [Google Scholar] [CrossRef]
  139. Staunton, D.E.; Dustin, M.L.; Erickson, H.P.; Springer, T.A. The arrangement of the immunoglobulin-like domains of ICAM-1 and the binding sites for LFA-1 and rhinovirus. Cell 1990, 61, 243–254. [Google Scholar] [CrossRef]
  140. Takai, Y.; Miyoshi, J.; Ikeda, W.; Ogita, H. Nectins and nectin-like molecules: Roles in contact inhibition of cell movement and proliferation. Nat. Rev. Mol. Cell Biol. 2008, 9, 603–615. [Google Scholar] [CrossRef]
  141. Takai, Y.; Ikeda, W.; Ogita, H.; Rikitake, Y. The immunoglobulin-like cell adhesion molecule nectin and its associated protein afadin. Annu. Rev. Cell Dev. Biol. 2008, 24, 309–342. [Google Scholar] [CrossRef]
  142. Fabre, S.; Reymond, N.; Cocchi, F.; Menotti, L.; Dubreuil, P.; Campadelli-Fiume, G.; Lopez, M. Prominent role of the Ig-like V domain in trans-interactions of nectins. Nectin3 and nectin 4 bind to the predicted C-C′-C″-D beta-strands of the nectin1 V domain. J. Biol. Chem. 2002, 277, 27006–27013. [Google Scholar] [CrossRef] [PubMed]
  143. Reymond, N.; Fabre, S.; Lecocq, E.; Adelaide, J.; Dubreuil, P.; Lopez, M. Nectin4/PRR4, a new afadin-associated member of the nectin family that trans-interacts with nectin1/PRR1 through V domain interaction. J. Biol. Chem. 2001, 276, 43205–43215. [Google Scholar] [CrossRef]
  144. Kanzaki, N.; Ogita, H.; Komura, H.; Ozaki, M.; Sakamoto, Y.; Majima, T.; Ijuin, T.; Takenawa, T.; Takai, Y. Involvement of the nectin-afadin complex in PDGF-induced cell survival. J. Cell Sci. 2008, 121, 2008–2017. [Google Scholar] [CrossRef] [PubMed]
  145. Green, L.L. Antibody engineering via genetic engineering of the mouse: XenoMouse strains are a vehicle for the facile generation of therapeutic human monoclonal antibodies. J. Immunol. Methods 1999, 231, 11–23. [Google Scholar] [CrossRef] [PubMed]
  146. Challita-Eid, P.M.; Satpayev, D.; Yang, P.; An, Z.; Morrison, K.; Shostak, Y.; Raitano, A.; Nadell, R.; Liu, W.; Lortie, D.R.; et al. Enfortumab Vedotin Antibody-Drug Conjugate Targeting Nectin-4 Is a Highly Potent Therapeutic Agent in Multiple Preclinical Cancer Models. Cancer Res. 2016, 76, 3003–3013. [Google Scholar] [CrossRef]
  147. Maguire, W.F.; Lee, D.; Weinstock, C.; Gao, X.; Bulik, C.C.; Agrawal, S.; Chang, E.; Hamed, S.S.; Bloomquist, E.W.; Tang, S.; et al. FDA Approval Summary: Enfortumab Vedotin plus Pembrolizumab for Cisplatin-Ineligible Locally Advanced or Metastatic Urothelial Carcinoma. Clin. Cancer Res. 2024, 30, 2011–2016. [Google Scholar] [CrossRef]
Figure 1. Overview of NIR-PIT. (A) NIR-PIT utilizes a photoabsorber (IR700)-conjugated mAb and NIR light. Monotherapy with NIR-PIT agents and NIR light is minimally cytotoxic. Cytotoxicity is induced specifically in agent-bound cells upon light irradiation. (B) Representative images of the highly selective cytotoxicity of NIR-PIT. Both targeting protein-expressing and non-expressing cells were co-cultured and treated with NIR-PIT agents. After NIR irradiation, cytotoxic effects were only induced in agent-bound cells (arrowhead).
Figure 1. Overview of NIR-PIT. (A) NIR-PIT utilizes a photoabsorber (IR700)-conjugated mAb and NIR light. Monotherapy with NIR-PIT agents and NIR light is minimally cytotoxic. Cytotoxicity is induced specifically in agent-bound cells upon light irradiation. (B) Representative images of the highly selective cytotoxicity of NIR-PIT. Both targeting protein-expressing and non-expressing cells were co-cultured and treated with NIR-PIT agents. After NIR irradiation, cytotoxic effects were only induced in agent-bound cells (arrowhead).
Pharmaceuticals 18 01419 g001
Figure 2. Schematics of membrane proteins. (A) Type I transmembrane, (B) type II transmembrane, (C) multipass transmembrane, and (D) GPI-anchored membrane protein.
Figure 2. Schematics of membrane proteins. (A) Type I transmembrane, (B) type II transmembrane, (C) multipass transmembrane, and (D) GPI-anchored membrane protein.
Pharmaceuticals 18 01419 g002
Figure 3. Schematics of the antibody type. (A) The structures of different types of monoclonal antibodies. (B) Antibody fragment types. CDR, complementarity-determining region; scFV, single-chain antibody variable fragment.
Figure 3. Schematics of the antibody type. (A) The structures of different types of monoclonal antibodies. (B) Antibody fragment types. CDR, complementarity-determining region; scFV, single-chain antibody variable fragment.
Pharmaceuticals 18 01419 g003
Table 1. Clinical trials of NIR-PIT.
Table 1. Clinical trials of NIR-PIT.
Clinical TrialPatientsNIR-PITResultsRef.
Phase I/IIaMale: 24, Female: 61−4 cyclesCR: 4 (13.3%), PR: 9 (30%), SD: 11 (36.7%), DC: 24 (80%), PD: 6 (20%)[7]
Phase IFemale: 31 cyclesCR: 0 (0%), PR: 2 (66.7%), SD: 0 (0%), DC: 2 (66.7%), PD: 1 (33.3%)[5]
Table 2. Target molecules for photoimmunotherapy targeting cancer cells [5,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40].
Table 2. Target molecules for photoimmunotherapy targeting cancer cells [5,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40].
Protein TypeTarget MoleculeIR700 CarrierCancerRefs.
Type 1 single-pass transmembrane proteinCadherin-17ARB102Pancreatic cancer[13]
CD44IM7Breast cancer, colon cancer, oral cancer, lung cancer[14]
CD146YY146Melanoma[15]
c-KIT12A8Gastrointestinal stromal tumors[16]
DLL3RovalpituzumabLung cancer[17]
EGFRCetuximab, panitumumabHead and neck cancer, bladder cancer, breast cancer, lung cancer, liver cancer[5,18,19,20]
GPA33A33ScFvColon cancer[21]
HER2Trastuzumab, pertuzumabBreast cancer, gastric cancer, ovarian cancer[22,23,24]
ICAM1R65-D6Breast cancer[25]
Nectin-4Enfortumab biosimilarBladder cancer[26]
PodoplaninNZ-1Mesothelioma[27]
TROP2HuT6-16-2Pancreatic cancer[28]
Type 2 single-pass transmembrane proteinPSMAYPSMA-1Prostate cancer[29]
Multi-pass transmemnrane proteinCD20RituximabB cell lymphoma[30]
CD29KMI6Melanoma[31]
CD47B6H12Bladder cancer[32]
CD133AC133Glioblastoma[33]
GPI-anchored proteinCEAChimeric anti-CEA mAb, M5APancreatic cancer, colon cancer[34,35,36]
Glypican-1MiltuximabProstate cancer, bladder cancer, brain cancer, ovary cancer[37]
Glypican-3YP7, HN3A431/G1 cells (epidermoid cancer cells stably expressing human GPC3)[38,39]
MesothelinhYP218A431/H9 cells (epidermoid cancer cells stably expressing human mesothelin)[40]
Table 3. IgG isotype for photoimmunotherapy agents, PMID: [49,50,51,52].
Table 3. IgG isotype for photoimmunotherapy agents, PMID: [49,50,51,52].
TargetAntibodyTypeIsotypeRef.
CEALabetuzumabHumanizedIgG1[49]
CD44v6BivatuzumabHumanizedIgG1[50]
EGFRCetuximabChimericIgG1[51]
NecitumumabFully humanIgG1[51]
NimotuzumabHumanizedIgG1[51]
PanitumumabFully humanIgG2[51]
HER2PertuzumabHumanizedIgG1[51]
TrastuzumabHumanizedIgG1[51]
ICAM-1BersanlimabFully humanIgG1[52]
Nectin-4EnfortumabFully humanIgG1[51]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Suzuki, M.; Hanaoka, H. An Overview of Target Membrane Proteins for Near-Infrared Photoimmunotherapy. Pharmaceuticals 2025, 18, 1419. https://doi.org/10.3390/ph18091419

AMA Style

Suzuki M, Hanaoka H. An Overview of Target Membrane Proteins for Near-Infrared Photoimmunotherapy. Pharmaceuticals. 2025; 18(9):1419. https://doi.org/10.3390/ph18091419

Chicago/Turabian Style

Suzuki, Motofumi, and Hirofumi Hanaoka. 2025. "An Overview of Target Membrane Proteins for Near-Infrared Photoimmunotherapy" Pharmaceuticals 18, no. 9: 1419. https://doi.org/10.3390/ph18091419

APA Style

Suzuki, M., & Hanaoka, H. (2025). An Overview of Target Membrane Proteins for Near-Infrared Photoimmunotherapy. Pharmaceuticals, 18(9), 1419. https://doi.org/10.3390/ph18091419

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