Next Article in Journal
Multi-Algorithm Analysis Reveals Pyroptosis-Linked Genes as Pancreatic Cancer Biomarkers
Previous Article in Journal
PIK Your Poison: The Effects of Combining PI3K and CDK Inhibitors against Metastatic Cutaneous Squamous Cell Carcinoma In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Promising Diagnostic and Therapeutic Approaches Based on VHHs for Cancer Management

by
Ying Cong
1,
Nick Devoogdt
2,
Philippe Lambin
1,3,
Ludwig J. Dubois
1,* and
Ala Yaromina
1,*
1
The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6211 LK Maastricht, The Netherlands
2
Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, 1090 Brussels, Belgium
3
Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
*
Authors to whom correspondence should be addressed.
Cancers 2024, 16(2), 371; https://doi.org/10.3390/cancers16020371
Submission received: 29 November 2023 / Revised: 4 January 2024 / Accepted: 4 January 2024 / Published: 15 January 2024
(This article belongs to the Section Clinical Research of Cancer)

Abstract

:

Simple Summary

With around 15 kD molecular weight, VHHs are considered the smallest antigen-binding fragments. Based on their unique properties, VHHs have broad application prospects such as bio-sensing, molecular imaging, drug delivery, disease treatments, and diagnosis. In this review, we discuss VHH applications for cancer treatments and diagnosis. Meanwhile, an overview of VHH-based agents in clinical trials is provided.

Abstract

The discovery of the distinctive structure of heavy chain-only antibodies in species belonging to the Camelidae family has elicited significant interest in their variable antigen binding domain (VHH) and gained attention for various applications, such as cancer diagnosis and treatment. This article presents an overview of the characteristics, advantages, and disadvantages of VHHs as compared to conventional antibodies, and their usage in diverse applications. The singular properties of VHHs are explained, and several strategies that can augment their utility are outlined. The preclinical studies illustrating the diagnostic and therapeutic efficacy of distinct VHHs in diverse formats against solid cancers are summarized, and an overview of the clinical trials assessing VHH-based agents in oncology is provided. These investigations demonstrate the enormous potential of VHHs for medical research and healthcare.

1. Introduction

In 1993, a different type of antibody called heavy chain only antibody (HCAb) was discovered in camelids, which had another structure compared with conventional antibodies (Figure 1) [1]. Another type of antibody lacking light chains is also found in cartilaginous fish [2,3]. HCAbs only contain two heavy chains, each one bearing beside two constant regions CH2 and CH3 also a variable region. This variable region is also called the variable domain of heavy chain of heavy-chain-only antibody (VHH), can be recombinantly produced and has a molecular weight of only about 15 kD, which is 10% of a conventional antibody and half of a single-chain variable fragment (scFv). Therefore, VHHs are also called single domain antibodies (sdAb) or nanobodies.
While conventional antibodies have been used in anti-cancer therapies for many years, VHH-based therapies have more recently started to be explored. The unique features of VHH, such as high affinity and lower immunogenicity, may provide important differences in anti-cancer therapies. To date, preclinical studies and early-stage clinical trials have demonstrated the promising potential of VHH for cancer management. In this review, we provide an overview of VHH characteristics and the general route of VHH development. Furthermore, we summarize the applications of VHH in anti-cancer therapies and provide an overview of registered clinical trials testing VHH-based therapeutic and diagnostic agents for oncological diseases.

2. General Characteristics of VHH

In VHH, there are only three complementarity-determining regions (CDRs) that make contact with the antigen, as compared to six in conventional antibodies (3 in VH and 3 in VL). To compensate for this and to ensure sufficient contact interface, the amino chain in CDR3 (and to a lesser extent CDR1) is longer in VHH than its counterpart in conventional antibodies [4,5]. As a result, the diversity and specificity of CDR3 is improved. Additionally, CDR3 in VHH can make up an exposed ring structure, which can behave like a ‘finger’ to stick into the ‘pocket’ of antigen, where a conventional antibody rather interacts with flat surfaces [6,7,8,9]. In addition, an extra cysteine residue in CDR3 can form a disulfide bond with an extra cysteine residue in either CDR1 or in the framework region 2 (FR2), which increases the stability of the VHH structure, while decreasing the energy requested for binding with antigens [4,5].

2.1. Biochemical and Biophysical Characteristics

The unique biochemical and biophysical characteristics of VHH are their stability, in addition to high solubility, thermo- and proteolytic-resistance [10]. FR2 contains many hydrophilic amino acid residues, which determines the high solubility and dispersibility of VHH in water. VHH has also good conformational and thermal stability, which allows VHH to be stable at body temperature during a week while still having sufficient binding ability [11]. Additionally, VHH is also stable at extreme pH conditions, ensuring that VHH maintains its bioactivity in the stomach or the intestine [12,13,14,15]. The stability of VHH makes it possible to design VHH treatments following various administration methods, including intravenous injection, inhalation, and oral and intranasal administration. Altogether, the stable biochemical and biophysical properties nurture the increasing applications of VHH.

2.2. Low Immunogenicity

Relatively little scientific evidence is publicly available to make strong claims about the immunogenicity of VHHs. Nevertheless, the structure of VHH has a high similarity with the human VH structure, in particular, it has a high homology (86–94%) with the human VH3 family [16,17]. The relatively small size of VHH (125 amino acids) implies a low number of potentially immunogenic epitopes. The high solubility of VHHs reduces the likeliness to form highly immunogenic aggregates. Moreover, unmodified VHHs show fast blood clearance, which minimizes capture by antigen-presenting cells. All these features favor low immunogenicity. In a phase I clinical study of 68Ga-HER2-VHH, 20 patients were free of anti-drug antibodies (ADA) after one injection [18]. A phase I study of ALX-0141, a trimeric format consisting of two identical anti-RANKL humanized VHHs and one anti-albumin humanized VHH, demonstrated that after a single subcutaneous injection in 42 healthy volunteers, no ADAs were found [19]. In summary, the immunogenicity of VHH in humans seems to be relatively low, which prevents one of the main causes of adverse effects, i.e., allergy. Humanization of VHH is possible and can further reduce its immunogenicity [20,21,22].

2.3. High Tissue Penetration and Fast Blood Clearance

Theoretically, the passive diffusion rate of a molecule in tissue is inversely proportional to its molecular size. Therefore, compared with conventional antibody (150 kD), the monovalent VHH (15 kD) has a fast blood extravasation and a better tissue penetration to reach its target. This results in a more homogeneous distribution, for example in solid tumors, compared to conventional antibodies [23,24,25,26]. It has been shown that VHHs can cross the blood-brain barrier (BBB) of healthy rats [27], providing improved ability for the diagnosis and treatment of brain cancer, where in addition the BBB is likely disrupted [28,29,30,31,32]. Due to the small molecular weight, VHH is usually cleared quickly through the kidneys [33,34,35,36]. As a result, the half-life time of VHH in the human body is mostly less than an hour [37,38], a feature helpful to prevent toxicity. Although the short half-life of VHH is advantageous for diagnostic applications, it may limit its therapeutic efficacy [39,40,41]. Methods such as PEGylation, fusion or binding with albumin, multimerization, or fusion with antibody Fc can help to extend the half-life [42,43,44,45,46,47]. For example, a bivalent anti-VEGF VHH showed a 1.8-fold longer half-life in comparison with the monovalent VHH [42]. PEGylation resulted in a 12-fold longer half-life of an anti-CEA/CD3 bispecific VHH [45].

3. The Generation of VHH

There are several steps in the identification of a functional VHH.
  • The construction of a VHH gene library
The first step in the generation of antigen-specific VHHs is to construct a VHH-encoding cDNA library. Antigen-specific VHHs can be retrieved either from an immune VHH library, a naive VHH library, or a synthetic VHH library. The immune library is the most frequently used method, exploiting the in vivo affinity maturation of the VHHs. To construct an immune library, camelid animals are immunized with a targeted antigen. The targeted antigen is injected into camelids multiple times, e.g., 7 times every two weeks [48]. 5~7 days after the immunization lymphocytes are then extracted from the peripheral blood, which contains B cells producing the matured HCAbs. Lymphocyte RNA is extracted and reverse transcribed into cDNA [49]. The bulk of cDNA is used as a template for PCR with VHH-specific primers [50]. Subsequently, the amplified VHH DNA fragments are cloned into bacterial vectors to construct the library [51].
2.
The selection of specific VHH
Phage display is the dominant technique to screen libraries for functional VHHs. Here, the VHH immune libraries are cloned in phagemids in fusion with bacteriophage gene III, and then VHHs are displayed on the tip of the M13 bacteriophage by infection with helper phages providing phage structural components in trans. The target antigen is fixed directly or indirectly on a solid carrier, to allow the phage library to interact with the antigen. While the phages of interest bind with the fixed antigen, unbounded or unspecific phages are removed by washing. Subsequently, a specific acid (e.g., 0.1 M pH 2.2 HCl-Glycine) or base (e.g., 0.1 M triethylamine) is used to release the phage from its antigen. Generally, after 2–5 selection rounds of panning, the VHHs with good specificity and affinity can be selected [52].
3.
The production of VHH
Due to its simple structure and good water solubility, VHH can be mass-produced by using relatively low-cost expression systems such as bacterial or yeast expression systems [51,53,54,55,56,57]. After verifying the nucleic acid sequence of the selected VHH, an expression vector is constructed and transferred into bacteria (e.g., E. coli BL21) or yeast (e.g., Pichia pastoris GS115 or X33) for the production of VHH. In the bacteria expression process, successfully transformed colonies are picked for culture expansion and induction of VHH production. For example, if a Lac-operon is present in the expression vector, IPTG (Isopropyl-β-d-thiogalactoside) is used to induce gene expression when the bacteria reach the exponential growth phase. Most often VHHs are transported to the periplasm and extracted from there, although also cytoplasmic expression is being pursued [58]. In the yeast expression process, successfully transformed yeast colonies are picked for culture expansion following induction, with VHHs most often ending up in the conditioned medium due to secretion. The VHH protein in the supernatant, cell lysates, or periplasmic extracts can be purified by methods such as affinity chromatography, ion exchange chromatography, or molecular sieve chromatography. The production of VHHs in mammalian cell lines is rarely reported because of the high cost of this expression system.

4. Applications of VHH

4.1. Molecular Imaging

Molecular imaging is one of the important methods in cancer research, which allows the noninvasive study of tumors and their microenvironment, as well as tracing or monitoring cancer progression and therapy effectiveness. Generally, a molecular targeted imaging agent contains a targeting moiety and a radioisotope for nuclear imaging or a fluorescent moiety for optical imaging [59]. To target cancer cells more precisely, the imaging agent is required to have higher specificity and sensitivity, with lower toxicity and fewer adverse effects.
Conventional antibodies have been frequently tested in cancer molecular imaging studies [60]. However, the application of conventional antibody-based molecular imaging is rather limited by its large size and relatively long plasma half-life. This requires the use of radionuclides with a long physical half-life, resulting in more radiation exposure and additional measures for radiation protection [61]. With their smaller molecular weight, VHHs have a deeper penetration into the central part of solid tumors to detect target expression [62,63]. In addition, the short half-life and fast blood clearance limit non-specific VHH presence resulting in an improved signal-to-noise ratio and fewer adverse effects. Therefore, the VHH-based imaging technique is currently heavily investigated for cancer diagnosis, to target proteins or receptors which are overexpressed in tumor cells or the tumor microenvironment during cancer progression.
Positron emission tomography (PET) and single photon emission computed tomography (SPECT) are the two main nuclear imaging techniques for cancer diagnosis. To improve the accuracy and specificity and to reduce potential side effects of radiation exposure in PET/SPECT imaging, the VHH has been used as an ‘immune carrier’ in the construction of radioactive probes. In the very first study, Gainkam et al. investigated the uptake of the 99mTc labeled anti-EGFR VHH and showed that tumor uptake was correlated with tumor burden, concluding that the VHH can be used to track the tumor response to therapy [34]. Table 1 identifies several preclinical studies on VHH-radionuclide conjugates for imaging. Clinically, in a phase II study, 68Ga labeled anti-HER2 VHH was used to assess HER2 expression in primary breast cancer and metastatic lesions [64]. This clinical study showed that the 68Ga-HER2-nanobody could be safely administered, with favorable biodistribution and high accumulation in HER2-positive primary breast cancer and its metastases. In another recent phase I study, a 68Ga-labeled VHH against the macrophage mannose receptor CD206 was tested in a phase I PET safety study to track tumor-associated macrophages in the tumor stroma [65]. Other clinical studies are summarized in Table 7.
Besides being conjugated with a radionuclide, VHH can also be conjugated with a fluorescent moiety for molecular imaging [85]. For example, Debie et al. extensively evaluated fluorescently labeled anti-HER2 VHH for image-guided surgery and demonstrated a significant reduction of residual tumor lesions as compared to conventional surgery [86,87]. Van Brussel et al. developed a CAIX-specific VHH conjugated to IRDye800CW, that could visualize tumors compared to the background 2–3 h after the anti-CAIX VHH injection [88]. Furthermore, the VHH can also be used in magnetic resonance imaging (MRI) by conjugation with MRI contrast agents, such as gadolinium or paramagnetic nanoparticles [89,90].

4.2. VHHs in Anti-Cancer Therapies

The unique features that have been discussed above make VHHs promising replacements for monoclonal antibodies or other types of antibody fragments. Figure 2 generally describes different applications of VHHs in anti-cancer therapies.

4.2.1. Radioimmunotherapy

Radioimmunotherapy (RIT) is a “targeted” radionuclide therapy coupling radioisotopes with therapeutic properties to antibodies. These antibodies bind to specific targets in the tumors and deliver their cargo, i.e., the radionuclide, at that site, resulting in minimal toxicity to normal tissue [91]. 90Y-ibritumomab tiuxetan (Zevalin®, Bayer) and 131I-tositumomab (Bexxar®, GSK) are so far the only two RITs approved for clinical use to treat hematological malignancies [92,93]. Both are designed based on anti-CD20 monoclonal antibodies to treat CD20-positive non-Hodgkin’s lymphoma (NHL). The application of RIT for solid tumor treatment is, however, still a challenge. In addition to the large size of conventional antibodies, the leakage of tumor blood vessels and the complexity of the matrix in solid tumors make it difficult for RIT to reach all tumor cells. Therefore, alternative carriers of therapeutic radioisotopes, such as VHHs and peptides have been investigated for cancer RIT, especially for the treatment of solid tumors.
Table 2 summarizes the preclinical studies on the application of VHH in RIT of solid tumors. For example, D’Huyvetter et al. treated mice bearing HER2-positive SKOV3 ovarian adenocarcinoma xenografts with the 177Lu-labeled anti-HER2 VHH and demonstrated that the tumor growth was almost completely inhibited. Moreover, the event-free survival was significantly longer for the treated group compared to the control group that received vehicle treatment, while no evidence of renal inflammation or necrosis was observed [94]. Similarly, anti-FAP VHHs that are conjugated to 131I or 225Ac were able to limit tumor growth and enhance the survival of mice bearing FAP-expressing tumors [95]. To target the TME, Xu et al. generated the anti-FAPα (fibroblast activation protein-α) VHH-Fc fusion labeled with 177Lu and showed therapeutic efficacy in HT1080 fibrosarcoma xenografts in mice [96]. Anti-MMR VHH labeled with 177Lu was designed to target the stroma of solid tumors. The 177Lu-labeled anti-MMR VHH significantly delayed tumor growth, which outcompeted the effects of currently used therapies, such as immune checkpoint inhibitors, anti-angiogenic therapy (anti-VEGFR2 mAbs), and chemotherapies (doxorubicin, paclitaxel) [97].

4.2.2. Photodynamic Therapy

VHH can be conjugated to a photosensitizer (PS), which is a light-activatable compound that generates cytotoxic reactive oxygen species upon activation with a certain light wavelength [104]. These VHH-PS conjugates can also be used as tracers or probes to monitor tumor progression using optical imaging after photodynamic therapy (PDT). IRDye700DX, a phthalocyanine dye, is one of the most promising photosensitizers, whose chemical structure is similar to hematoporphyrin. Its maximum absorption wavelength is in the infrared wavelength region and therefore easier to be transmitted through human tissue. Additionally, IRDye has low absorption of light with a wavelength of 400–600 nm, which reduces the skin photosensitivity reaction. Therefore, many studies have used IRDye700DX as PS to couple with VHH for PDT. For example, Renard et al. generated anti-EGFR VHH conjugated with 111In-DPTA (diethylenetriaminepentaacetic acid) and IRDye700DX PS in a site-specific way, which homed to A431 xenografts in vivo and resulted in light-induced toxicity via cellular internalization [105]. Deken et al. investigated anti-HER2 VHH conjugated with IRDye700DX PS that induced significant regression of trastuzumab-resistant HER2-expressing breast tumors after a single treatment session with selective cytotoxicity [106]. Table 3 summarizes cancer targets investigated using VHH-PS conjugates in preclinical studies.

4.2.3. VHH as Immune Checkpoint Inhibitor

Several conventional antibodies have been used as immune checkpoint inhibitors (ICIs) for anti-cancer treatments in the past years. However, due to the complex structure of these antibodies and the tumor heterogeneity, antibody-based ICIs face many challenges such as insufficient tumor penetration and immune-related adverse effects [112], highlighting the need for optimization of antibody-based ICIs.
Due to the smaller molecular weight, VHHs have a higher penetration potential in solid tumors. Petit et al. showed that T cell-mediated targeted delivery of an anti-PD-L1 VHH outperformed an anti-PD-L1 conventional antibody in inhibiting tumor growth, related to its higher tumor penetration in MC38 tumor-bearing mice [113]. Table 4 summarizes the studies evaluating VHH-based ICIs. Although the VHHs lack ADCC and CDC due to the absence of Fc fragment, the VHH still has anti-cancer efficacy by inhibiting the bioactivity of immune checkpoints to attenuate immune suppression in tumors [114]. Therefore, fusing VHH with an Fc fragment can also compensate the ADCC and CDC in anti-cancer therapies [115]. Ma et al. demonstrated that a tetravalent anti-PD-L1 VHH-Fc fusion had a higher inhibitory effect on tumor growth compared to an equimolar dose of an anti-PD-L1 monoclonal antibody (Tecentriq analog) in MC38 xenograft-bearing mice [116]. Clinically, envafolimab (KN035), anti-PD-L1 VHH-Fc fusion formulated for subcutaneous injection, has demonstrated a favorable safety and pharmacokinetic profile, with promising antitumor activity in patients with advanced solid tumors in Phase I–II trials [117,118,119].

4.2.4. Targeting Tumor-Specific Antigens

Tumor-specific antigens (TSAs) are important targets for cancer treatments and diagnosis. However, TSAs are not exclusively expressed in cancer cells. Normal cells may express TSAs at low levels, while cancer cells have high expression of TSAs during tumor proliferation. As a consequence, molecules targeting TSAs should have high specificity and affinity, which can be achieved with VHH.
VHHs have been designed to bind tumor-specific targets TSAs such as HER2, EGFR, and VEGFR, over the past years and they are reported to have anti-tumor efficacy. Table 5 summarizes the preclinical studies evaluating the therapeutic efficacy of various anti-TSA VHHs. As the small molecular weight of VHH allows better intratumor penetration, it leads to high VHH clearance resulting in a shorter plasma half-life during the treatment. In vivo studies showed the half-life of VHH binding with its target within 1–2 h [128,129]. In addition, VHHs are not capable of promoting ADCC or CDC effects during the treatment, which should also be considered. These two limitations can be overcome by modification of the VHHs. For example, Sadeghi et al. generated an anti-VEGF bivalent VHH that showed a 1.8-fold longer half-life compared to its monovalent form [42]. An et al. coupled ABD035 to anti-GPC3 VHH labeled with 68Ga, which reached a peak tumor uptake at 6 h post-injection combined with a reduced kidney accumulation at 1 h post-injection [78]. Another method is coupling VHHs with an Fc fragment to obtain the VHH-conjugates with the ability to promote ADCC or CDC [130].

4.2.5. VHH-Drug Conjugates (VHH-DC)

Antibody-drug conjugates (ADCs) are immune-conjugates formed with a monoclonal antibody as a ‘delivery carrier’ and a cytotoxic/immune-promoting drug as ‘cargo’, which are conjugated via a chemical linker [144]. Highly specific targeting antibodies are used to overcome the toxicity and adverse effects of traditional chemotherapeutic drugs. In addition, the antibody may also have additive or synergistic anti-cancer effects with the drug, when both components of ADC exhibit anti-tumor effects [145]. Nowadays, third-generation ADCs have been developed and investigated. With novel techniques for ADC generation, ADCs with high targeting and therapeutic efficacy, relatively low immunogenicity, consistent drug-to-antibody ratio (DAR) and low toxicity are becoming available [146]. As more ADCs have been entering clinical trials, more challenges for ADC development have been raised. During the synthesis of ADC, the chemical modification leads to the instability of monoclonal antibodies. In addition, the high molecular weight makes it difficult for ADC to enter the inner hypoxic part of solid tumors, which limits the application of ADCs in cancer treatments. Using VHHs as a ‘carrier’ of cytotoxic drugs is a novel alternative to overcome the disadvantages of conventional monoclonal antibodies. Table 6 summarizes recent VHH-drug conjugates studies. Indeed, Wu et al. demonstrated that the anti-5T4 VHH-SN38 (irinotecan analog) conjugate was detected at 85 μm from the periphery of the patient-derived organoids (PDOs), while the anti-5T4 conventional antibody-SN38 conjugate mainly located around the periphery of PDOs [147]. Drug resistance and downregulation of tumor antigens are challenging for ADCs as well [148]. Espelin et al. used anti-HER2 VHH conjugated with doxorubicin (MM-302) in combination with trastuzumab to demonstrate synergistic anti-tumor activity both in vitro and in vivo, supporting its translation into clinical trials [149]. Therefore, other strategies are being investigated including utilization of novel tumor antigens, antibody formats, linkers, payloads, etc.

4.2.6. VHH-Based CAR-T

The chimeric antigen receptor (CAR) is an engineered synthetic receptor that can redirect lymphocytes to recognize and eliminate specific cells, which express the target antigens [153]. T cells are most commonly used to carry CARs, but CARs can also be applied to other types of immune cells, such as NK cells, dendritic cells, macrophages, etc. [154,155]. Hitherto, six CAR-T therapies have been approved by the FDA to treat relapsed/refractory multiple myeloma (RRMM), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), follicular lymphoma (FL) and Precursor B-cell lymphoblastic leukemia, of which anti-CD19 CAR-T products’ antigen recognition domain is based on the same scFv (Kymriah®, Yescarta®, Tecartus®, and Breyanzi®), whereas the antigen recognition domain of B-cell maturation antigen (BCMA, Carvykti®, Legend Biotech and Janssen Biotech) is based on VHH [156]. The peptide linker of scFv-based CAR-T, however, showed immunogenicity as neutralizing antibodies were generated against the linker [157]. Additionally, T cell exhaustion is another factor that limits the application of CAR-T, which occurs independently of the binding with the target antigen but is caused by the unstable structure of scFv. The exposed hydrophobic residues in the scFv variable domain and poor VH or VL folding stability can result in CAR aggregation. The CAR aggregation on the surface of CAR-T can lead in turn to the activation of effector cells and cytotoxic signaling cascade, which contribute to T cell exhaustion [158,159]. Xie et al. generated VHH-based CAR-T cells, which were designed to target PD-L1, CD47, or EIIIB+ fibronectin splice variant to target the TME, and each VHH-based CAR-T could reduce solid tumor growth and improve survival in immunocompetent tumor-bearing mice [160,161]. Rajabzadeh et al. designed an anti-MUC1 CAR-T which could increase secretion of Th1 cytokines and cytotoxic activity that could inhibit cancer cell viability [162]. In summary, VHH-based CAR-T is promising as an approach to treating solid tumors [163].

4.3. Other Applications of VHHs

VHHs can also be applied in other novel anti-cancer treatments. VHH, which binds and neutralizes cytokines or growth factors such as TNFα or VEGF, respectively, can attenuate tumor growth by inhibiting metastasis or angiogenesis [13,164,165,166]. Alternatively, VHHs conjugated with proteins (e.g., cucurmosin, pseudomonas exotoxin), cytokines (e.g., IFNγ, IL-2, TNFα) or peptides show promising potential of inhibiting tumor growth [167,168,169,170,171]. Yin et al. investigated an anti-PD-L1 VHH and used it for modified liposomes co-delivery of simevastatin/gefitinib that could remodel the TME of EGFR-T790M-mutated NSCLC and overcome the drug resistance [172]. Furthermore, VHHs can be used to construct cancer vaccinations. For example, conjugating anti-CD11b VHH with H-2Db-restricted immunodominant E7 epitope (E749-57), which exists in high-risk human papillomavirus-associated cancers, induced stronger CD8+ T-cell responses against HPV positive tumors than E749-57 alone [173]. The tumor vaccine that contains anti-CD47 VHH could induce suppression of tumor progression and improve the long-term survival of tumor-bearing mice by remodeling the TME [174]. Altogether, VHHs have great potential to develop novel anti-cancer therapies with higher efficacy and lower toxicity, which warrants further investigations.

5. An Overview of Ongoing Clinical Trials of VHHs in Cancer Treatments

With the failure of many promising anti-cancer therapies in clinical trials and understanding the progression of cancer, more strict safety and efficacy requirements have been raised for the investigation of cancer treatments. Since the discovery of VHHs in 1993, VHH have become a promising agent for the diagnosis and treatment of cancer, viral infection [41,175,176], and other diseases due to its unique structure and physicochemical properties. The CAR-T, in which anti-BCMA VHHs serve as antigen receptors on T cells (Carvykti®, Legend Biotech, and Janssen Biotech), has been approved by the FDA for the treatment of advanced multiple myeloma in 2022. Meanwhile, many clinical trials evaluating the diagnostic and therapeutic potential of VHH-based agents in solid and hematological malignancies are ongoing. Table 7 summarizes some of these clinical trials. Data was collected on ClinicalTrial.gov, on 24 August 2023 (https://www.clinicaltrials.gov/ct2/home, accessed on 24 August 2023, Keywords: VHH, nanobody, sdAb, cancer, tumor, malignancy).

6. Discussion and Future Directions

Since its discovery in 1993, VHHs have become one of the most promising approaches for the treatment and diagnosis of various diseases, including cancer, autoimmune diseases, respiratory diseases, and blood systemic diseases. The first VHH-based drug Caplacizumab was approved in 2018 for the treatment of thrombotic thrombocytopenic purpura and thrombosis. In 2022, the first VHH-based CAR-T treatment Carvykti has been approved by the FDA to treat multiple myeloma. Up until now, VHH alone as a therapeutical agent has not been approved for anti-cancer treatments.
As the smallest known antibody fragment that can recognize antigen, the VHHs retain high affinity and specificity, whilst having unique chemical and physical features compared to conventional antibodies. The unique molecular structure of VHHs makes it possible to bind some epitopes that conventional antibodies are not able to access, such as the active structure in the middle of the protein cleft. These unique features may behave like a ‘double-edged sword’ in the therapeutic application of VHHs. For example, the single domain character of VHH makes almost every amino acid residue crucial for antigen-antibody interaction increasing the difficulty of VHH humanization. Additionally, fast blood clearance can reduce the toxicity of the treatment, but it may compromise treatment efficacy as well. In addition, although the lack of Fc fragments minimize immunogenicity, the absence of ADCC or CDC limits the application of VHHs in anti-cancer treatments, which are mainly promoted by Fc fragments. Therefore, related technologies should be applied in the VHHs development process to overcome these drawbacks of VHHs. In addition, in preclinical studies evaluating the efficacy of VHHs, the cancer models should be chosen carefully, and the data should be interpreted with caution, since in some studies VHH imaging agents recognizing human antigen were tested in human tumor xenografts in mice, which cannot provide information on the binding of VHH in normal tissues of human origin. Similarly, VHH with immune-modulating properties targeting human antigens should be investigated in humanized mouse models.

7. Conclusions

According to the database of clinical trials investigating VHH-based anti-cancer agents, most VHHs in ongoing clinical trials are still in their early stages. Most of these anti-cancer targets cover PD-1, PD-L1, CTLA4, HER2, EGFR, and BCMA. The VHH studies gathered in this review indicate that many promising novel VHH-based anti-cancer treatments and diagnostic tools have been investigated. With the progressive understanding of cancer biology, more potential anti-cancer targets have been identified. Together with straightforward selection methods, efficient and cost-effective expression, and purification technological processes, VHHs represent a promising opportunity in anti-cancer treatment and cancer diagnosis.

Author Contributions

Writing, Y.C.; review and editing, N.D., P.L., L.J.D. and A.Y. All authors have read and agreed to the published version of the manuscript.

Funding

Authors acknowledge financial support from the China Scholarship Council (CSC, grant number: 202006100044 for Y.C.).

Conflicts of Interest

Lambin reports, within and outside the submitted work, grants/sponsored research agreements from Varian medical, Oncoradiomics, ptTheragnostic/DNAmito, Health Innovation Ventures. He received an advisor/presenter fee and/or reimbursement of travel costs/external grant writing fee and/or in-kind manpower contribution from Oncoradiomics, BHV, Merck, Varian, Elekta, ptTheragnostic and Convert pharmaceuticals. Lambin has shares in the company Oncoradiomics SA, Convert Pharmaceuticals SA and The Medical Cloud Company SPRL and is co-inventor of two issued patents with royalties on radiomics (PCT/NL2014/050248, PCT/NL2014/050728) licensed to Oncoradiomics and one issue patent on mtDNA (PCT/EP2014/059089) licensed to ptTheragnostic/DNAmito, three non-patented invention (softwares) licensed to ptTheragnos-tic/DNAmito, Oncoradiomics and Health Innovation Ventures and three non-issues, non-licensed patents on Deep Learning-Radiomics and LSRT (N2024482, N2024889, N2024889). Dubois reports, outside the submitted work, shares in the company Convert Pharmaceuticals SA and is co-inventor of a non-issue patent on LSRT (N2024889). Devoogdt has several patents and patent applications related to VHH products and technologies. He is co-founder and consultant of the companies ABSCINT and PRECIRIX that utilize VHHs in their technology program. His research group has received research support from the companies Boehringer Ingelheim, Complix, 121BIO, Agenus, Telix Pharma, Roche, Confo Therapeutics, Exevir and Esobiotec.

References

  1. Hamers-Casterman, C.; Atarhouch, T.; Muyldermans, S.; Robinson, G.; Hamers, C.; Songa, E.B.; Bendahman, N.; Hamers, R. Naturally occurring antibodies devoid of light chains. Nature 1993, 363, 446–448. [Google Scholar] [CrossRef] [PubMed]
  2. Streltsov, V.A.; Carmichael, J.A.; Nuttall, S.D. Structure of a shark IgNAR antibody variable domain and modeling of an early-developmental isotype. Protein Sci. 2005, 14, 2901–2909. [Google Scholar] [CrossRef]
  3. Greenberg, A.S.; Avila, D.; Hughes, M.; Hughes, A.; McKinney, E.C.; Flajnik, M.F. A new antigen receptor gene family that undergoes rearrangement and extensive somatic diversification in sharks. Nature 1995, 374, 168–173. [Google Scholar] [CrossRef]
  4. Vu, K.B.; Ghahroudi, M.A.; Wyns, L.; Muyldermans, S. Comparison of llama VH sequences from conventional and heavy chain antibodies. Mol. Immunol. 1997, 34, 1121–1131. [Google Scholar] [CrossRef] [PubMed]
  5. Desmyter, A.; Transue, T.R.; Ghahroudi, M.A.; Thi, M.H.; Poortmans, F.; Hamers, R.; Muyldermans, S.; Wyns, L. Crystal structure of a camel single-domain VH antibody fragment in complex with lysozyme. Nat. Struct. Biol. 1996, 3, 803–811. [Google Scholar] [CrossRef] [PubMed]
  6. De Groof, T.W.M.; Bobkov, V.; Heukers, R.; Smit, M.J. Nanobodies: New avenues for imaging, stabilizing and modulating GPCRs. Mol. Cell. Endocrinol. 2019, 484, 15–24. [Google Scholar] [CrossRef] [PubMed]
  7. De Groof, T.W.M.; Mashayekhi, V.; Fan, T.S.; Bergkamp, N.D.; Sastre Torano, J.; van Senten, J.R.; Heukers, R.; Smit, M.J.; Oliveira, S. Nanobody-Targeted Photodynamic Therapy Selectively Kills Viral GPCR-Expressing Glioblastoma Cells. Mol. Pharm. 2019, 16, 3145–3156. [Google Scholar] [CrossRef] [PubMed]
  8. Rasmussen, S.G.; Choi, H.J.; Fung, J.J.; Pardon, E.; Casarosa, P.; Chae, P.S.; Devree, B.T.; Rosenbaum, D.M.; Thian, F.S.; Kobilka, T.S.; et al. Structure of a nanobody-stabilized active state of the beta(2) adrenoceptor. Nature 2011, 469, 175–180. [Google Scholar] [CrossRef]
  9. Rasmussen, B.; Fletcher, I.R.; Brocks, J.J.; Kilburn, M.R. Reassessing the first appearance of eukaryotes and cyanobacteria. Nature 2008, 455, 1101–1104. [Google Scholar] [CrossRef]
  10. Salvador, J.P.; Vilaplana, L.; Marco, M.P. Nanobody: Outstanding features for diagnostic and therapeutic applications. Anal. Bioanal. Chem. 2019, 411, 1703–1713. [Google Scholar] [CrossRef]
  11. Kunz, P.; Zinner, K.; Mucke, N.; Bartoschik, T.; Muyldermans, S.; Hoheisel, J.D. The structural basis of nanobody unfolding reversibility and thermoresistance. Sci. Rep. 2018, 8, 7934. [Google Scholar] [CrossRef] [PubMed]
  12. Vosjan, M.J.; Perk, L.R.; Roovers, R.C.; Visser, G.W.; Stigter-van Walsum, M.; van Bergen En Henegouwen, P.M.; van Dongen, G.A. Facile labelling of an anti-epidermal growth factor receptor Nanobody with 68Ga via a novel bifunctional desferal chelate for immuno-PET. Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 753–763. [Google Scholar] [CrossRef]
  13. Ebrahimizadeh, W.; Mousavi Gargari, S.L.; Javidan, Z.; Rajabibazl, M. Production of Novel VHH Nanobody Inhibiting Angiogenesis by Targeting Binding Site of VEGF. Appl. Biochem. Biotechnol. 2015, 176, 1985–1995. [Google Scholar] [CrossRef]
  14. Dumoulin, M.; Conrath, K.; Van Meirhaeghe, A.; Meersman, F.; Heremans, K.; Frenken, L.G.; Muyldermans, S.; Wyns, L.; Matagne, A. Single-domain antibody fragments with high conformational stability. Protein Sci. 2002, 11, 500–515. [Google Scholar] [CrossRef] [PubMed]
  15. Goldman, E.R.; Liu, J.L.; Zabetakis, D.; Anderson, G.P. Enhancing Stability of Camelid and Shark Single Domain Antibodies: An Overview. Front. Immunol. 2017, 8, 865. [Google Scholar] [CrossRef]
  16. Arbabi-Ghahroudi, M. Camelid Single-Domain Antibodies: Promises and Challenges as Lifesaving Treatments. Int. J. Mol. Sci. 2022, 23, 5009. [Google Scholar] [CrossRef] [PubMed]
  17. Achour, I.; Cavelier, P.; Tichit, M.; Bouchier, C.; Lafaye, P.; Rougeon, F. Tetrameric and homodimeric camelid IgGs originate from the same IgH locus. J. Immunol. 2008, 181, 2001–2009. [Google Scholar] [CrossRef] [PubMed]
  18. Keyaerts, M.; Xavier, C.; Heemskerk, J.; Devoogdt, N.; Everaert, H.; Ackaert, C.; Vanhoeij, M.; Duhoux, F.P.; Gevaert, T.; Simon, P.; et al. Phase I Study of 68Ga-HER2-Nanobody for PET/CT Assessment of HER2 Expression in Breast Carcinoma. J. Nucl. Med. 2016, 57, 27–33. [Google Scholar] [CrossRef]
  19. Schoen, P.; Jacobs, S.; Verschueren, K.; Ottevaere, I.; Sobry, S.; Holz, J.-B. Anti-RANKL nanobody ALX-0141 shows sustained biomarker inhibition in a phase I study in healthy postmenopausal women. In Bone Abstracts; Bioscientifica: Bristol, UK, 2013. [Google Scholar]
  20. Li, Y.; Zhou, C.; Li, J.; Liu, J.; Lin, L.; Li, L.; Cao, D.; Li, Q.; Wang, Z. Single domain based bispecific antibody, Muc1-Bi-1, and its humanized form, Muc1-Bi-2, induce potent cancer cell killing in muc1 positive tumor cells. PLoS ONE 2018, 13, e0191024. [Google Scholar] [CrossRef]
  21. Sulea, T. Humanization of Camelid Single-Domain Antibodies. In Methods in Molecular Biology; Springer: Berlin/Heidelberg, Germany, 2022; pp. 299–312. [Google Scholar]
  22. Ackaert, C.; Smiejkowska, N.; Xavier, C.; Sterckx, Y.G.J.; Denies, S.; Stijlemans, B.; Elkrim, Y.; Devoogdt, N.; Caveliers, V.; Lahoutte, T.; et al. Immunogenicity Risk Profile of Nanobodies. Front. Immunol. 2021, 12, 632687. [Google Scholar] [CrossRef]
  23. Lwin, T.M.; Hernot, S.; Hollandsworth, H.; Amirfakhri, S.; Filemoni, F.; Debie, P.; Hoffman, R.M.; Bouvet, M. Tumor-specific near-infrared nanobody probe rapidly labels tumors in an orthotopic mouse model of pancreatic cancer. Surgery 2020, 168, 85–91. [Google Scholar] [CrossRef]
  24. Debie, P.; Lafont, C.; Defrise, M.; Hansen, I.; van Willigen, D.M.; van Leeuwen, F.W.B.; Gijsbers, R.; D’Huyvetter, M.; Devoogdt, N.; Lahoutte, T.; et al. Size and affinity kinetics of nanobodies influence targeting and penetration of solid tumours. J. Control. Release 2020, 317, 34–42. [Google Scholar] [CrossRef]
  25. Xavier, C.; Blykers, A.; Vaneycken, I.; D’Huyvetter, M.; Heemskerk, J.; Lahoutte, T.; Devoogdt, N.; Caveliers, V. (18)F-nanobody for PET imaging of HER2 overexpressing tumors. Nucl. Med. Biol. 2016, 43, 247–252. [Google Scholar] [CrossRef]
  26. Broos, K.; Keyaerts, M.; Lecocq, Q.; Renmans, D.; Nguyen, T.; Escors, D.; Liston, A.; Raes, G.; Breckpot, K.; Devoogdt, N. Non-invasive assessment of murine PD-L1 levels in syngeneic tumor models by nuclear imaging with nanobody tracers. Oncotarget 2017, 8, 41932–41946. [Google Scholar] [CrossRef]
  27. Caljon, G.; Caveliers, V.; Lahoutte, T.; Stijlemans, B.; Ghassabeh, G.H.; Van Den Abbeele, J.; Smolders, I.; De Baetselier, P.; Michotte, Y.; Muyldermans, S.; et al. Using microdialysis to analyse the passage of monovalent nanobodies through the blood-brain barrier. Br. J. Pharmacol. 2012, 165, 2341–2353. [Google Scholar] [CrossRef] [PubMed]
  28. Ruiz-López, E.; Schuhmacher, A.J. Transportation of Single-Domain Antibodies through the Blood-Brain Barrier. Biomolecules 2021, 11, 1131. [Google Scholar] [CrossRef] [PubMed]
  29. Jovcevska, I.; Muyldermans, S. The Therapeutic Potential of Nanobodies. BioDrugs 2020, 34, 11–26. [Google Scholar] [CrossRef] [PubMed]
  30. Lesniak, W.G.; Chu, C.; Jablonska, A.; Behnam Azad, B.; Zwaenepoel, O.; Zawadzki, M.; Lisok, A.; Pomper, M.G.; Walczak, P.; Gettemans, J.; et al. PET imaging of distinct brain uptake of a nanobody and similarly-sized PAMAM dendrimers after intra-arterial administration. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 1940–1951. [Google Scholar] [CrossRef]
  31. Custers, M.L.; Wouters, Y.; Jaspers, T.; De Bundel, D.; Dewilde, M.; Van Eeckhaut, A.; Smolders, I. Applicability of cerebral open flow microperfusion and microdialysis to quantify a brain-penetrating nanobody in mice. Anal. Chim. Acta 2021, 1178, 338803. [Google Scholar] [CrossRef]
  32. Heukers, R.; Fan, T.S.; de Wit, R.H.; van Senten, J.R.; De Groof, T.W.M.; Bebelman, M.P.; Lagerweij, T.; Vieira, J.; de Munnik, S.M.; Smits-de Vries, L.; et al. The constitutive activity of the virally encoded chemokine receptor US28 accelerates glioblastoma growth. Oncogene 2018, 37, 4110–4121. [Google Scholar] [CrossRef]
  33. Chakravarty, R.; Goel, S.; Cai, W. Nanobody: The “magic bullet” for molecular imaging? Theranostics 2014, 4, 386–398. [Google Scholar] [CrossRef]
  34. Gainkam, L.O.; Keyaerts, M.; Caveliers, V.; Devoogdt, N.; Vanhove, C.; Van Grunsven, L.; Muyldermans, S.; Lahoutte, T. Correlation between epidermal growth factor receptor-specific nanobody uptake and tumor burden: A tool for noninvasive monitoring of tumor response to therapy. Mol. Imaging Biol. 2011, 13, 940–948. [Google Scholar] [CrossRef] [PubMed]
  35. Vaneycken, I.; D’Huyvetter, M.; Hernot, S.; De Vos, J.; Xavier, C.; Devoogdt, N.; Caveliers, V.; Lahoutte, T. Immuno-imaging using nanobodies. Curr. Opin. Biotechnol. 2011, 22, 877–881. [Google Scholar] [CrossRef] [PubMed]
  36. Vaneycken, I.; Devoogdt, N.; Van Gassen, N.; Vincke, C.; Xavier, C.; Wernery, U.; Muyldermans, S.; Lahoutte, T.; Caveliers, V. Preclinical screening of anti-HER2 nanobodies for molecular imaging of breast cancer. FASEB J. 2011, 25, 2433–2446. [Google Scholar] [CrossRef] [PubMed]
  37. Coppieters, K.; Dreier, T.; Silence, K.; de Haard, H.; Lauwereys, M.; Casteels, P.; Beirnaert, E.; Jonckheere, H.; Van de Wiele, C.; Staelens, L.; et al. Formatted anti-tumor necrosis factor alpha VHH proteins derived from camelids show superior potency and targeting to inflamed joints in a murine model of collagen-induced arthritis. Arthritis Rheum. 2006, 54, 1856–1866. [Google Scholar] [CrossRef]
  38. Iqbal, U.; Trojahn, U.; Albaghdadi, H.; Zhang, J.; O’Connor-McCourt, M.; Stanimirovic, D.; Tomanek, B.; Sutherland, G.; Abulrob, A. Kinetic analysis of novel mono- and multivalent VHH-fragments and their application for molecular imaging of brain tumours. Br. J. Pharmacol. 2010, 160, 1016–1028. [Google Scholar] [CrossRef]
  39. Altunay, B.; Morgenroth, A.; Beheshti, M.; Vogg, A.; Wong, N.C.L.; Ting, H.H.; Biersack, H.J.; Stickeler, E.; Mottaghy, F.M. HER2-directed antibodies, affibodies and nanobodies as drug-delivery vehicles in breast cancer with a specific focus on radioimmunotherapy and radioimmunoimaging. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 1371–1389. [Google Scholar] [CrossRef] [PubMed]
  40. Bao, G.; Tang, M.; Zhao, J.; Zhu, X. Nanobody: A promising toolkit for molecular imaging and disease therapy. EJNMMI Res. 2021, 11, 6. [Google Scholar] [CrossRef]
  41. Sheikhi, A.; Hojjat-Farsangi, M. An immunotherapeutic method for COVID-19 patients: A soluble ACE2-Anti-CD16 VHH to block SARS-CoV-2 Spike protein. Hum. Vaccines Immunother. 2021, 17, 92–97. [Google Scholar] [CrossRef]
  42. Sadeghi, A.; Behdani, M.; Muyldermans, S.; Habibi-Anbouhi, M.; Kazemi-Lomedasht, F. Development of a mono-specific anti-VEGF bivalent nanobody with extended plasma half-life for treatment of pathologic neovascularization. Drug Test. Anal. 2020, 12, 92–100. [Google Scholar] [CrossRef] [PubMed]
  43. Hoefman, S.; Ottevaere, I.; Baumeister, J.; Sargentini-Maier, M.L. Pre-clinical intravenous serum pharmacokinetics of albumin binding and non-half-life extended Nanobodies®. Antibodies 2015, 4, 141–156. [Google Scholar] [CrossRef]
  44. Sleep, D.; Cameron, J.; Evans, L.R. Albumin as a versatile platform for drug half-life extension. BBA-Gen. Subj. 2013, 1830, 5526–5534. [Google Scholar] [CrossRef]
  45. Pan, H.; Liu, J.; Deng, W.; Xing, J.; Li, Q.; Wang, Z. Site-specific PEGylation of an anti-CEA/CD3 bispecific antibody improves its antitumor efficacy. Int. J. Nanomed. 2018, 13, 3189–3201. [Google Scholar] [CrossRef]
  46. Kontermann, R.E. Strategies for extended serum half-life of protein therapeutics. Curr. Opin. Biotechnol. 2011, 22, 868–876. [Google Scholar] [CrossRef] [PubMed]
  47. Xenaki, K.T.; Dorrestijn, B.; Muns, J.A.; Adamzek, K.; Doulkeridou, S.; Houthoff, H.; Oliveira, S.; van Bergen En Henegouwen, P.M. Homogeneous tumor targeting with a single dose of HER2-targeted albumin-binding domain-fused nanobody-drug conjugates results in long-lasting tumor remission in mice. Theranostics 2021, 11, 5525–5538. [Google Scholar] [CrossRef] [PubMed]
  48. Chow, K.M.; Whiteheart, S.W.; Smiley, J.R.; Sharma, S.; Boaz, K.; Coleman, M.J.; Maynard, A.; Hersh, L.B.; Vander Kooi, C.W. Immunization of Alpacas (Lama pacos) with Protein Antigens and Production of Antigen-specific Single Domain Antibodies. J. Vis. Exp. 2019, 143, e58471. [Google Scholar] [CrossRef]
  49. Marturano, A.; Hendrickx, M.L.V.; Falcinelli, E.; Sebastiano, M.; Guglielmini, G.; Hassanzadeh-Ghassabeh, G.; Muyldermans, S.; Declerck, P.J.; Gresele, P. Development of anti-matrix metalloproteinase-2 (MMP-2) nanobodies as potential therapeutic and diagnostic tools. Nanomedicine 2020, 24, 102103. [Google Scholar] [CrossRef]
  50. Jakobs, B.D.; Spannagel, L.; Purvanov, V.; Uetz-von Allmen, E.; Matti, C.; Legler, D.F. Engineering of Nanobodies Recognizing the Human Chemokine Receptor CCR7. Int. J. Mol. Sci. 2019, 20, 2597. [Google Scholar] [CrossRef]
  51. Baharlou, R.; Tajik, N.; Habibi-Anbouhi, M.; Shokrgozar, M.A.; Zarnani, A.H.; Shahhosseini, F.; Behdani, M. Generation and characterization of an anti-delta like ligand-4 Nanobody to induce non-productive angiogenesis. Anal. Biochem. 2018, 544, 34–41. [Google Scholar] [CrossRef]
  52. Ledsgaard, L.; Kilstrup, M.; Karatt-Vellatt, A.; McCafferty, J.; Laustsen, A.H. Basics of Antibody Phage Display Technology. Toxins 2018, 10, 236. [Google Scholar] [CrossRef]
  53. Evazalipour, M.; D’Huyvetter, M.; Tehrani, B.S.; Abolhassani, M.; Omidfar, K.; Abdoli, S.; Arezumand, R.; Morovvati, H.; Lahoutte, T.; Muyldermans, S.; et al. Generation and characterization of nanobodies targeting PSMA for molecular imaging of prostate cancer. Contrast Media Mol. Imaging 2014, 9, 211–220. [Google Scholar] [CrossRef]
  54. Ma, L.; Gu, K.; Zhang, C.H.; Chen, X.T.; Jiang, Y.; Melcher, K.; Zhang, J.; Wang, M.; Xu, H.E. Generation and characterization of a human nanobody against VEGFR-2. Acta Pharmacol. Sin. 2016, 37, 857–864. [Google Scholar] [CrossRef] [PubMed]
  55. Zhai, T.; Wang, C.; Xu, Y.; Huang, W.; Yuan, Z.; Wang, T.; Dai, S.; Peng, S.; Pang, T.; Jiang, W.; et al. Generation of a safe and efficacious llama single-domain antibody fragment (vHH) targeting the membrane-proximal region of 4-1BB for engineering therapeutic bispecific antibodies for cancer. J. Immunother. Cancer 2021, 9, e002131. [Google Scholar] [CrossRef]
  56. Chen, Q.; Zhou, Y.; Yu, J.; Liu, W.; Li, F.; Xian, M.; Nian, R.; Song, H.; Feng, D. An efficient constitutive expression system for Anti-CEACAM5 nanobody production in the yeast Pichia pastoris. Protein Expr. Purif. 2019, 155, 43–47. [Google Scholar] [CrossRef]
  57. Detalle, L.; Stohr, T.; Palomo, C.; Piedra, P.A.; Gilbert, B.E.; Mas, V.; Millar, A.; Power, U.F.; Stortelers, C.; Allosery, K.; et al. Generation and Characterization of ALX-0171, a Potent Novel Therapeutic Nanobody for the Treatment of Respiratory Syncytial Virus Infection. Antimicrob. Agents Chemother. 2016, 60, 6–13. [Google Scholar] [CrossRef]
  58. Djender, S.; Schneider, A.; Beugnet, A.; Crepin, R.; Desrumeaux, K.E.; Romani, C.; Moutel, S.; Perez, F.; de Marco, A. Bacterial cytoplasm as an effective cell compartment for producing functional VHH-based affinity reagents and Camelidae IgG-like recombinant antibodies. Microb. Cell Fact. 2014, 13, 140. [Google Scholar] [CrossRef] [PubMed]
  59. Hong, H.; Zhang, Y.; Sun, J.; Cai, W. Molecular imaging and therapy of cancer with radiolabeled nanoparticles. Nano Today 2009, 4, 399–413. [Google Scholar] [CrossRef] [PubMed]
  60. van Dongen, G.; Beaino, W.; Windhorst, A.D.; Zwezerijnen, G.J.C.; Oprea-Lager, D.E.; Hendrikse, N.H.; van Kuijk, C.; Boellaard, R.; Huisman, M.C.; Vugts, D.J. The Role of (89)Zr-Immuno-PET in Navigating and Derisking the Development of Biopharmaceuticals. J. Nucl. Med. 2021, 62, 438–445. [Google Scholar] [CrossRef]
  61. Hegi-Johnson, F.; Rudd, S.; Hicks, R.J.; De Ruysscher, D.; Trapani, J.A.; John, T.; Donnelly, P.; Blyth, B.; Hanna, G.; Everitt, S.; et al. Imaging immunity in patients with cancer using positron emission tomography. NPJ Precis. Oncol. 2022, 6, 24. [Google Scholar] [CrossRef]
  62. Kijanka, M.; Warnders, F.J.; El Khattabi, M.; Lub-de Hooge, M.; van Dam, G.M.; Ntziachristos, V.; de Vries, L.; Oliveira, S.; van Bergen En Henegouwen, P.M. Rapid optical imaging of human breast tumour xenografts using anti-HER2 VHHs site-directly conjugated to IRDye 800CW for image-guided surgery. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 1718–1729. [Google Scholar] [CrossRef]
  63. Terwisscha van Scheltinga, A.G.; van Dam, G.M.; Nagengast, W.B.; Ntziachristos, V.; Hollema, H.; Herek, J.L.; Schröder, C.P.; Kosterink, J.G.; Lub-de Hoog, M.N.; de Vries, E.G. Intraoperative near-infrared fluorescence tumor imaging with vascular endothelial growth factor and human epidermal growth factor receptor 2 targeting antibodies. J. Nucl. Med. 2011, 52, 1778–1785. [Google Scholar] [CrossRef] [PubMed]
  64. Gondry, O.C.V.; Xavier, C.; Raes, L.; Vanhoeij, M.; Verfaillie, G.; Fontaine, C.; Glorieus, K.; De Grève, J.; Joris, S.; Luyten, I.; et al. Phase II trial to assess the repeatability and tumor uptake of [68Ga]Ga-HER2-sdAb PET/CT in patients with Breast Carcinoma. J. Nucl. Med. 2023, in press.
  65. Gondry, O.; Xavier, C.; Raes, L.; Heemskerk, J.; Devoogdt, N.; Everaert, H.; Breckpot, K.; Lecocq, Q.; Decoster, L.; Fontaine, C.; et al. Phase I Study of [68Ga]Ga-Anti-CD206-sdAb for PET/CT Assessment of Protumorigenic Macrophage Presence in Solid Tumors (MMR Phase I). J. Nucl. Med. 2023, 64, 1378–1384. [Google Scholar] [CrossRef]
  66. Rashidian, M.; Ingram, J.R.; Dougan, M.; Dongre, A.; Whang, K.A.; LeGall, C.; Cragnolini, J.J.; Bierie, B.; Gostissa, M.; Gorman, J.; et al. Predicting the response to CTLA-4 blockade by longitudinal noninvasive monitoring of CD8 T cells. J. Exp. Med. 2017, 214, 2243–2255. [Google Scholar] [CrossRef]
  67. Sriraman, S.K.; Davies, C.W.; Gill, H.; Kiefer, J.R.; Yin, J.; Ogasawara, A.; Urrutia, A.; Javinal, V.; Lin, Z.; Seshasayee, D.; et al. Development of an (18)F-labeled anti-human CD8 VHH for same-day immunoPET imaging. Eur. J. Nucl. Med. Mol. Imaging 2023, 50, 679–691. [Google Scholar] [CrossRef] [PubMed]
  68. Chatalic, K.L.; Veldhoven-Zweistra, J.; Bolkestein, M.; Hoeben, S.; Koning, G.A.; Boerman, O.C.; de Jong, M.; van Weerden, W.M. A Novel (1)(1)(1)In-Labeled Anti-Prostate-Specific Membrane Antigen Nanobody for Targeted SPECT/CT Imaging of Prostate Cancer. J. Nucl. Med. 2015, 56, 1094–1099. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, Y.; Ding, Y.; Li, N.; Wang, S.; Zhou, S.; Li, R.; Yang, H.; Li, W.; Qu, J. Noninvasive Imaging of Tumor PD-L1 Expression Using [99mTc]Tc-Labeled KN035 with SPECT/CT. Mol. Pharm. 2023, 20, 690–700. [Google Scholar] [CrossRef] [PubMed]
  70. Lv, G.; Sun, X.; Qiu, L.; Sun, Y.; Li, K.; Liu, Q.; Zhao, Q.; Qin, S.; Lin, J. PET Imaging of Tumor PD-L1 Expression with a Highly Specific Nonblocking Single-Domain Antibody. J. Nucl. Med. 2020, 61, 117–122. [Google Scholar] [CrossRef]
  71. Liu, Q.; Jiang, L.; Li, K.; Li, H.; Lv, G.; Lin, J.; Qiu, L. Immuno-PET imaging of 68Ga-labeled nanobody Nb109 for dynamic monitoring the PD-L1 expression in cancers. Cancer Immunol. Immunother. 2021, 70, 1721–1733. [Google Scholar] [CrossRef]
  72. Movahedi, K.; Schoonooghe, S.; Laoui, D.; Houbracken, I.; Waelput, W.; Breckpot, K.; Bouwens, L.; Lahoutte, T.; De Baetselier, P.; Raes, G.; et al. Nanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophages. Cancer Res. 2012, 72, 4165–4177. [Google Scholar] [CrossRef]
  73. Xavier, C.; Blykers, A.; Laoui, D.; Bolli, E.; Vaneyken, I.; Bridoux, J.; Baudhuin, H.; Raes, G.; Everaert, H.; Movahedi, K.; et al. Clinical Translation of [68Ga]Ga-NOTA-anti-MMR-sdAb for PET/CT Imaging of Protumorigenic Macrophages. Mol. Imaging Biol. 2019, 21, 898–906. [Google Scholar] [CrossRef] [PubMed]
  74. Lecocq, Q.; Zeven, K.; De Vlaeminck, Y.; Martens, S.; Massa, S.; Goyvaerts, C.; Raes, G.; Keyaerts, M.; Breckpot, K.; Devoogdt, N. Noninvasive Imaging of the Immune Checkpoint LAG-3 Using Nanobodies, from Development to Pre-Clinical Use. Biomolecules 2019, 9, 548. [Google Scholar] [CrossRef] [PubMed]
  75. Lecocq, Q.; Awad, R.M.; De Vlaeminck, Y.; de Mey, W.; Ertveldt, T.; Goyvaerts, C.; Raes, G.; Thielemans, K.; Keyaerts, M.; Devoogdt, N.; et al. Single-Domain Antibody Nuclear Imaging Allows Noninvasive Quantification of LAG-3 Expression by Tumor-Infiltrating Leukocytes and Predicts Response of Immune Checkpoint Blockade. J. Nucl. Med. 2021, 62, 1638–1644. [Google Scholar] [CrossRef] [PubMed]
  76. Xavier, C.; Vaneycken, I.; D’Huyvetter, M.; Heemskerk, J.; Keyaerts, M.; Vincke, C.; Devoogdt, N.; Muyldermans, S.; Lahoutte, T.; Caveliers, V. Synthesis, preclinical validation, dosimetry, and toxicity of 68Ga-NOTA-anti-HER2 Nanobodies for iPET imaging of HER2 receptor expression in cancer. J. Nucl. Med. 2013, 54, 776–784. [Google Scholar] [CrossRef] [PubMed]
  77. Li, L.; Liu, T.; Shi, L.; Zhang, X.; Guo, X.; Hu, B.; Yao, M.; Zhu, H.; Yang, Z.; Jia, B.; et al. HER2-targeted dual radiotracer approach with clinical potential for noninvasive imaging of trastuzumab-resistance caused by epitope masking. Theranostics 2022, 12, 5551–5563. [Google Scholar] [CrossRef]
  78. An, S.; Zhang, D.; Zhang, Y.; Wang, C.; Shi, L.; Wei, W.; Huang, G.; Liu, J. GPC3-targeted immunoPET imaging of hepatocellular carcinomas. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 2682–2692. [Google Scholar] [CrossRef]
  79. Liu, T.; Wu, Y.; Shi, L.; Li, L.; Hu, B.; Wang, Y.; Gao, H.; Yu, X.; Zhang, X.; Zhao, H.; et al. Preclinical evaluation of [99mTc]Tc-labeled anti-EpCAM nanobody for EpCAM receptor expression imaging by immuno-SPECT/CT. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 1810–1821. [Google Scholar] [CrossRef]
  80. Piramoon, M.; Hosseinimehr, S.J.; Omidfar, K.; Noaparast, Z.; Abedi, S.M. 99m Tc-anti-epidermal growth factor receptor nanobody for tumor imaging. Chem. Biol. Drug Des. 2017, 89, 498–504. [Google Scholar] [CrossRef]
  81. Hu, G.; Zhu, W.; Liu, Y.; Wang, Y.; Zhang, Z.; Zhu, S.; Duan, W.; Zhou, P.; Fu, C.; Li, F.; et al. Development and comparison of three (89)Zr-labeled anti-CLDN18.2 antibodies to noninvasively evaluate CLDN18.2 expression in gastric cancer: A preclinical study. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 2634–2644. [Google Scholar] [CrossRef]
  82. Wang, H.; Meng, A.M.; Li, S.H.; Zhou, X.L. A nanobody targeting carcinoembryonic antigen as a promising molecular probe for non-small cell lung cancer. Mol. Med. Rep. 2017, 16, 625–630. [Google Scholar] [CrossRef]
  83. van Lith, S.A.M.; Huizing, F.J.; Franssen, G.M.; Hoeben, B.A.W.; Lok, J.; Doulkeridou, S.; Boerman, O.C.; Gotthardt, M.; van Bergen En Henegouwen, P.M.P.; Bussink, J.; et al. Novel VHH-Based Tracers with Variable Plasma Half-Lives for Imaging of CAIX-Expressing Hypoxic Tumor Cells. Mol. Pharm. 2022, 19, 3511–3520. [Google Scholar] [CrossRef]
  84. Jailkhani, N.; Ingram, J.R.; Rashidian, M.; Rickelt, S.; Tian, C.; Mak, H.; Jiang, Z.; Ploegh, H.L.; Hynes, R.O. Noninvasive imaging of tumor progression, metastasis, and fibrosis using a nanobody targeting the extracellular matrix. Proc. Natl. Acad. Sci. USA 2019, 116, 14181–14190. [Google Scholar] [CrossRef] [PubMed]
  85. Debie, P.; Declerck, N.B.; van Willigen, D.; Huygen, C.M.; De Sloovere, B.; Mateusiak, L.; Bridoux, J.; Puttemans, J.; Devoogdt, N.; van Leeuwen, F.W.B.; et al. The Design and Preclinical Evaluation of a Single-Label Bimodal Nanobody Tracer for Image-Guided Surgery. Biomolecules 2021, 11, 360. [Google Scholar] [CrossRef] [PubMed]
  86. Debie, P.; Devoogdt, N.; Hernot, S. Targeted Nanobody-Based Molecular Tracers for Nuclear Imaging and Image-Guided Surgery. Surgery 2020, 8, 12. [Google Scholar] [CrossRef] [PubMed]
  87. Debie, P.; Vanhoeij, M.; Poortmans, N.; Puttemans, J.; Gillis, K.; Devoogdt, N.; Lahoutte, T.; Hernot, S. Improved Debulking of Peritoneal Tumor Implants by Near-Infrared Fluorescent Nanobody Image Guidance in an Experimental Mouse Model. Mol. Imaging Biol. 2018, 20, 361–367. [Google Scholar] [CrossRef] [PubMed]
  88. van Brussel, A.S.; Adams, A.; Oliveira, S.; Dorresteijn, B.; El Khattabi, M.; Vermeulen, J.F.; van der Wall, E.; Mali, W.P.; Derksen, P.W.; van Diest, P.J.; et al. Hypoxia-Targeting Fluorescent Nanobodies for Optical Molecular Imaging of Pre-Invasive Breast Cancer. Mol. Imaging Biol. 2016, 18, 535–544. [Google Scholar] [CrossRef]
  89. Huang, H.; Wu, T.; Shi, H.; Wu, Y.; Yang, H.; Zhong, K.; Wang, Y.; Liu, Y. Modular design of nanobody-drug conjugates for targeted-delivery of platinum anticancer drugs with an MRI contrast agent. Chem. Commun. 2019, 55, 5175–5178. [Google Scholar] [CrossRef]
  90. Prantner, A.M.; Yin, C.; Kamat, K.; Sharma, K.; Lowenthal, A.C.; Madrid, P.B.; Scholler, N. Molecular Imaging of Mesothelin-Expressing Ovarian Cancer with a Human and Mouse Cross-Reactive Nanobody. Mol. Pharm. 2018, 15, 1403–1411. [Google Scholar] [CrossRef]
  91. Larson, S.M.; Carrasquillo, J.A.; Cheung, N.K.; Press, O.W. Radioimmunotherapy of human tumours. Nat. Rev. Cancer 2015, 15, 347–360. [Google Scholar] [CrossRef]
  92. Wiseman, G.A.; Kornmehl, E.; Leigh, B.; Erwin, W.D.; Podoloff, D.A.; Spies, S.; Sparks, R.B.; Stabin, M.G.; Witzig, T.; White, C.A. Radiation dosimetry results and safety correlations from 90Y-ibritumomab tiuxetan radioimmunotherapy for relapsed or refractory non-Hodgkin’s lymphoma: Combined data from 4 clinical trials. J. Nucl. Med. 2003, 44, 465–474. [Google Scholar]
  93. Rajendran, J.G.; Gopal, A.K.; Fisher, D.R.; Durack, L.D.; Gooley, T.A.; Press, O.W. Myeloablative 131I-tositumomab radioimmunotherapy in treating non-Hodgkin’s lymphoma: Comparison of dosimetry based on whole-body retention and dose to critical organ receiving the highest dose. J. Nucl. Med. 2008, 49, 837–844. [Google Scholar] [CrossRef] [PubMed]
  94. D’Huyvetter, M.; Vincke, C.; Xavier, C.; Aerts, A.; Impens, N.; Baatout, S.; De Raeve, H.; Muyldermans, S.; Caveliers, V.; Devoogdt, N.; et al. Targeted radionuclide therapy with A 177Lu-labeled anti-HER2 nanobody. Theranostics 2014, 4, 708–720. [Google Scholar] [CrossRef] [PubMed]
  95. Dekempeneer, Y.M.S.; Santens, F.; Navarro, L.; Berdal, M.; Lucero, M.M.; Pombo Antunes, A.R.; Van Ginderachter, J.A.; Lahoutte, T.; Devoogdt, N.; D’Huyvetter, M. Preclinical evaluation of a radiotheranostic single-domain antibody against Fibroblast Activation Protein alpha. J. Nucl. Med. 2023, in press.
  96. Xu, J.; Li, S.; Xu, S.; Dai, J.; Luo, Z.; Cui, J.; Cai, F.; Geng, C.; Wang, Z.; Tang, X. Screening and Preclinical Evaluation of Novel Radiolabeled Anti-Fibroblast Activation Protein-α Recombinant Antibodies. Cancer Biother. Radiopharm. 2023, 38, 726–737. [Google Scholar] [CrossRef] [PubMed]
  97. Bolli, E.; D’Huyvetter, M.; Murgaski, A.; Berus, D.; Stange, G.; Clappaert, E.J.; Arnouk, S.; Pombo Antunes, A.R.; Krasniqi, A.; Lahoutte, T.; et al. Stromal-targeting radioimmunotherapy mitigates the progression of therapy-resistant tumors. J. Control Release 2019, 314, 1–11. [Google Scholar] [CrossRef]
  98. D’Huyvetter, M.; De Vos, J.; Xavier, C.; Pruszynski, M.; Sterckx, Y.G.J.; Massa, S.; Raes, G.; Caveliers, V.; Zalutsky, M.R.; Lahoutte, T.; et al. (131)I-labeled Anti-HER2 Camelid sdAb as a Theranostic Tool in Cancer Treatment. Clin. Cancer Res. 2017, 23, 6616–6628. [Google Scholar] [CrossRef]
  99. Feng, Y.; Meshaw, R.; McDougald, D.; Zhou, Z.; Zhao, X.G.; Jannetti, S.A.; Reiman, R.E.; Pippen, E.; Marjoram, R.; Schaal, J.L.; et al. Evaluation of an (131)I-labeled HER2-specific single domain antibody fragment for the radiopharmaceutical therapy of HER2-expressing cancers. Sci. Rep. 2022, 12, 3020. [Google Scholar] [CrossRef]
  100. Pruszynski, M.; Koumarianou, E.; Vaidyanathan, G.; Revets, H.; Devoogdt, N.; Lahoutte, T.; Lyerly, H.K.; Zalutsky, M.R. Improved tumor targeting of anti-HER2 nanobody through N-succinimidyl 4-guanidinomethyl-3-iodobenzoate radiolabeling. J. Nucl. Med. 2014, 55, 650–656. [Google Scholar] [CrossRef]
  101. Choi, J.; Vaidyanathan, G.; Koumarianou, E.; Kang, C.M.; Zalutsky, M.R. Astatine-211 labeled anti-HER2 5F7 single domain antibody fragment conjugates: Radiolabeling and preliminary evaluation. Nucl. Med. Biol. 2018, 56, 10–20. [Google Scholar] [CrossRef]
  102. Feng, Y.; Meshaw, R.; Zhao, X.G.; Jannetti, S.; Vaidyanathan, G.; Zalutsky, M.R. Effective Treatment of Human Breast Carcinoma Xenografts with Single-Dose (211)At-Labeled Anti-HER2 Single-Domain Antibody Fragment. J. Nucl. Med. 2023, 64, 124–130. [Google Scholar] [CrossRef]
  103. Pruszynski, M.; D’Huyvetter, M.; Bruchertseifer, F.; Morgenstern, A.; Lahoutte, T. Evaluation of an Anti-HER2 Nanobody Labeled with (225)Ac for Targeted α-Particle Therapy of Cancer. Mol. Pharm. 2018, 15, 1457–1466. [Google Scholar] [CrossRef]
  104. Castano, A.P.; Mroz, P.; Hamblin, M.R. Photodynamic therapy and anti-tumour immunity. Nat. Rev. Cancer 2006, 6, 535–545. [Google Scholar] [CrossRef] [PubMed]
  105. Renard, E.; Collado Camps, E.; Canovas, C.; Kip, A.; Gotthardt, M.; Rijpkema, M.; Denat, F.; Goncalves, V.; van Lith, S.A.M. Site-Specific Dual-Labeling of a VHH with a Chelator and a Photosensitizer for Nuclear Imaging and Targeted Photodynamic Therapy of EGFR-Positive Tumors. Cancers 2021, 13, 428. [Google Scholar] [CrossRef]
  106. Deken, M.M.; Kijanka, M.M.; Beltran Hernandez, I.; Slooter, M.D.; de Bruijn, H.S.; van Diest, P.J.; van Bergen En Henegouwen, P.M.P.; Lowik, C.; Robinson, D.J.; Vahrmeijer, A.L.; et al. Nanobody-targeted photodynamic therapy induces significant tumor regression of trastuzumab-resistant HER2-positive breast cancer, after a single treatment session. J. Control Release 2020, 323, 269–281. [Google Scholar] [CrossRef] [PubMed]
  107. van Driel, P.; Boonstra, M.C.; Slooter, M.D.; Heukers, R.; Stammes, M.A.; Snoeks, T.J.A.; de Bruijn, H.S.; van Diest, P.J.; Vahrmeijer, A.L.; van Bergen En Henegouwen, P.M.P.; et al. EGFR targeted nanobody-photosensitizer conjugates for photodynamic therapy in a pre-clinical model of head and neck cancer. J. Control Release 2016, 229, 93–105. [Google Scholar] [CrossRef] [PubMed]
  108. van Driel, P.B.; van der Vorst, J.R.; Verbeek, F.P.; Oliveira, S.; Snoeks, T.J.; Keereweer, S.; Chan, B.; Boonstra, M.C.; Frangioni, J.V.; van Bergen en Henegouwen, P.M.; et al. Intraoperative fluorescence delineation of head and neck cancer with a fluorescent anti-epidermal growth factor receptor nanobody. Int. J. Cancer 2014, 134, 2663–2673. [Google Scholar] [CrossRef] [PubMed]
  109. Beltrán Hernández, I.; Angelier, M.L.; Del Buono D’Ondes, T.; Di Maggio, A.; Yu, Y.; Oliveira, S. The Potential of Nanobody-Targeted Photodynamic Therapy to Trigger Immune Responses. Cancers 2020, 12, 978. [Google Scholar] [CrossRef] [PubMed]
  110. Heukers, R.; Mashayekhi, V.; Ramirez-Escudero, M.; de Haard, H.; Verrips, T.C.; van Bergen En Henegouwen, P.M.P.; Oliveira, S. VHH-Photosensitizer Conjugates for Targeted Photodynamic Therapy of Met-Overexpressing Tumor Cells. Antibodies 2019, 8, 26. [Google Scholar] [CrossRef]
  111. Mashayekhi, V.; Xenaki, K.T.; van Bergen En Henegouwen, P.M.P.; Oliveira, S. Dual Targeting of Endothelial and Cancer Cells Potentiates In Vitro Nanobody-Targeted Photodynamic Therapy. Cancers 2020, 12, 2732. [Google Scholar] [CrossRef]
  112. de Miguel, M.; Calvo, E. Clinical Challenges of Immune Checkpoint Inhibitors. Cancer Cell 2020, 38, 326–333. [Google Scholar] [CrossRef]
  113. Petit, P.F.; Bombart, R.; Desimpel, P.H.; Naulaerts, S.; Thouvenel, L.; Collet, J.F.; Van den Eynde, B.J.; Zhu, J. T Cell-Mediated Targeted Delivery of Anti-PD-L1 Nanobody Overcomes Poor Antibody Penetration and Improves PD-L1 Blocking at the Tumor Site. Cancer Immunol. Res. 2022, 10, 713–727. [Google Scholar] [CrossRef] [PubMed]
  114. Sato, Y.; Casson, C.N.; Matsuda, A.; Kim, J.I.; Shi, J.Q.; Iwasaki, S.; Chen, S.; Modrell, B.; Chan, C.; Tavares, D.; et al. Fc-independent functions of anti-CTLA-4 antibodies contribute to anti-tumor efficacy. Cancer Immunol. Immunother. 2022, 71, 2421–2431. [Google Scholar] [CrossRef] [PubMed]
  115. Jiang, C.; Zhang, L.; Xu, X.; Qi, M.; Zhang, J.; He, S.; Tian, Q.; Song, S. Engineering a Smart Agent for Enhanced Immunotherapy Effect by Simultaneously Blocking PD-L1 and CTLA-4. Adv. Sci. 2021, 8, e2102500. [Google Scholar] [CrossRef]
  116. Ma, L.; Gai, J.; Qiao, P.; Li, Y.; Li, X.; Zhu, M.; Li, G.; Wan, Y. A novel bispecific nanobody with PD-L1/TIGIT dual immune checkpoint blockade. Biochem. Biophys. Res. Commun. 2020, 531, 144–151. [Google Scholar] [CrossRef] [PubMed]
  117. Papadopoulos, K.P.; Harb, W.; Peer, C.J.; Hua, Q.; Xu, S.; Lu, H.; Lu, N.; He, Y.; Xu, T.; Dong, R.; et al. First-in-Human Phase I Study of Envafolimab, a Novel Subcutaneous Single-Domain Anti-PD-L1 Antibody, in Patients with Advanced Solid Tumors. Oncologist 2021, 26, e1514–e1525. [Google Scholar] [CrossRef]
  118. Li, J.; Deng, Y.; Zhang, W.; Zhou, A.P.; Guo, W.; Yang, J.; Yuan, Y.; Zhu, L.; Qin, S.; Xiang, S.; et al. Subcutaneous envafolimab monotherapy in patients with advanced defective mismatch repair/microsatellite instability high solid tumors. J. Hematol. Oncol. 2021, 14, 95. [Google Scholar] [CrossRef]
  119. Shimizu, T.; Nakajima, T.E.; Yamamoto, N.; Yonemori, K.; Koyama, T.; Kondo, S.; Sunakawa, Y.; Izawa, N.; Horie, Y.; Xiang, S.; et al. Phase I study of envafolimab (KN035), a novel subcutaneous single-domain anti-PD-L1 monoclonal antibody, in Japanese patients with advanced solid tumors. Investig. New Drugs 2022, 40, 1021–1031. [Google Scholar] [CrossRef]
  120. Li, Y.; Wu, L.; Liu, Y.; Ma, S.; Huang, B.; Feng, X.; Wang, H. A novel multifunctional anti-PD-L1-CD16a-IL15 induces potent cancer cell killing in PD-L1-positive tumour cells. Transl. Oncol. 2022, 21, 101424. [Google Scholar] [CrossRef]
  121. Fang, T.; Li, R.; Li, Z.; Cho, J.; Guzman, J.S.; Kamm, R.D.; Ploegh, H.L. Remodeling of the Tumor Microenvironment by a Chemokine/Anti-PD-L1 Nanobody Fusion Protein. Mol. Pharm. 2019, 16, 2838–2844. [Google Scholar] [CrossRef]
  122. Awad, R.M.; Lecocq, Q.; Zeven, K.; Ertveldt, T.; De Beck, L.; Ceuppens, H.; Broos, K.; De Vlaeminck, Y.; Goyvaerts, C.; Verdonck, M.; et al. Formatting and gene-based delivery of a human PD-L1 single domain antibody for immune checkpoint blockade. Mol. Ther. Methods Clin. Dev. 2021, 22, 172–182. [Google Scholar] [CrossRef]
  123. Broos, K.; Lecocq, Q.; Xavier, C.; Bridoux, J.; Nguyen, T.T.; Corthals, J.; Schoonooghe, S.; Lion, E.; Raes, G.; Keyaerts, M.; et al. Evaluating a Single Domain Antibody Targeting Human PD-L1 as a Nuclear Imaging and Therapeutic Agent. Cancers 2019, 11, 872. [Google Scholar] [CrossRef]
  124. Broos, K.; Lecocq, Q.; Keersmaecker, B.; Raes, G.; Corthals, J.; Lion, E.; Thielemans, K.; Devoogdt, N.; Keyaerts, M.; Breckpot, K. Single Domain Antibody-Mediated Blockade of Programmed Death-Ligand 1 on Dendritic Cells Enhances CD8 T-cell Activation and Cytokine Production. Vaccines 2019, 7, 85. [Google Scholar] [CrossRef] [PubMed]
  125. Wen, B.; Zhao, L.; Wang, Y.; Qiu, C.; Xu, Z.; Huang, K.; Zhu, H.; Li, Z.; Li, H. Nanobodies targeting the interaction interface of programmed death receptor 1 (PD-1)/PD-1 ligand 1 (PD-1/PD-L1). Prep. Biochem. Biotechnol. 2020, 50, 252–259. [Google Scholar] [CrossRef]
  126. Noelia, S.-P.; Eva, M.; María Cristina, B.-B.; Sandra, H.-S.; Noelia, C.; Gualberto, G.-S.; Cristian, S.; Lucia, V. Long-Term Systemic Expression of a Novel PD-1 Blocking Nanobody from an AAV Vector Provides Antitumor Activity without Toxicity. Biomedicines 2020, 8, 562. [Google Scholar]
  127. Wan, R.; Liu, A.; Hou, X.; Lai, Z.; Li, J.; Yang, N.; Tan, J.; Mo, F.; Hu, Z.; Yang, X.; et al. Screening and antitumor effect of an anti-CTLA-4 nanobody. Oncol. Rep. 2018, 39, 511–518. [Google Scholar] [CrossRef]
  128. Gainkam, L.O.; Huang, L.; Caveliers, V.; Keyaerts, M.; Hernot, S.; Vaneycken, I.; Vanhove, C.; Revets, H.; De Baetselier, P.; Lahoutte, T. Comparison of the biodistribution and tumor targeting of two 99mTc-labeled anti-EGFR nanobodies in mice, using pinhole SPECT/micro-CT. J. Nucl. Med. 2008, 49, 788–795. [Google Scholar] [CrossRef]
  129. Huang, L.; Gainkam, L.O.; Caveliers, V.; Vanhove, C.; Keyaerts, M.; De Baetselier, P.; Bossuyt, A.; Revets, H.; Lahoutte, T. SPECT imaging with 99mTc-labeled EGFR-specific nanobody for in vivo monitoring of EGFR expression. Mol. Imaging Biol. 2008, 10, 167–175. [Google Scholar] [CrossRef]
  130. Maryam, Q.; Mahdi, B.; Mohammad Ali, S.; Vahid, M.-K.; Homa, M.-K.; Mahdi, H.-A. Construction and expression of an anti-VEGFR2 Nanobody-Fc fusionbody in NS0 host cell. Protein Expr. Purif. 2016, 123, 19–25. [Google Scholar]
  131. Van Impe, K.; Bethuyne, J.; Cool, S.; Impens, F.; Ruano-Gallego, D.; De Wever, O.; Vanloo, B.; Van Troys, M.; Lambein, K.; Boucherie, C.; et al. A nanobody targeting the F-actin capping protein CapG restrains breast cancer metastasis. Breast Cancer Res. 2013, 15, R116. [Google Scholar] [CrossRef] [PubMed]
  132. Li, T.; Qi, S.; Unger, M.; Hou, Y.N.; Deng, Q.W.; Liu, J.; Lam, C.M.C.; Wang, X.W.; Xin, D.; Zhang, P.; et al. Immuno-targeting the multifunctional CD38 using nanobody. Sci. Rep. 2016, 6, 27055. [Google Scholar] [CrossRef]
  133. Ingram, J.R.; Blomberg, O.S.; Sockolosky, J.T.; Ali, L.; Schmidt, F.I.; Pishesha, N.; Espinosa, C.; Dougan, S.K.; Garcia, K.C.; Ploegh, H.L.; et al. Localized CD47 blockade enhances immunotherapy for murine melanoma. Proc. Natl. Acad. Sci. USA 2017, 114, 10184–10189. [Google Scholar] [CrossRef] [PubMed]
  134. Maussang, D.; Mujic-Delic, A.; Descamps, F.J.; Stortelers, C.; Vanlandschoot, P.; Stigter-van Walsum, M.; Vischer, H.F.; van Roy, M.; Vosjan, M.; Gonzalez-Pajuelo, M.; et al. Llama-derived single variable domains (nanobodies) directed against chemokine receptor CXCR7 reduce head and neck cancer cell growth in vivo. J. Biol. Chem. 2013, 288, 29562–29572. [Google Scholar] [CrossRef]
  135. Sadeghnezhad, G.; Romão, E.; Bernedo-Navarro, R.; Massa, S.; Khajeh, K.; Muyldermans, S.; Hassania, S. Identification of New DR5 Agonistic Nanobodies and Generation of Multivalent Nanobody Constructs for Cancer Treatment. Int. J. Mol. Sci. 2019, 20, 4818. [Google Scholar] [CrossRef]
  136. Zottel, A.; Jovčevska, I.; Šamec, N.; Mlakar, J.; Šribar, J.; Križaj, I.; Skoblar Vidmar, M.; Komel, R. Anti-vimentin, anti-TUFM, anti-NAP1L1 and anti-DPYSL2 nanobodies display cytotoxic effect and reduce glioblastoma cell migration. Ther. Adv. Med. Oncol. 2020, 12, 1758835920915302. [Google Scholar] [CrossRef] [PubMed]
  137. Tabtimmai, L.; Suphakun, P.; Srisook, P.; Kiriwan, D.; Phanthong, S.; Kiatwuthinon, P.; Chaicumpa, W.; Choowongkomon, K. Cell-penetrable nanobodies (transbodies) that inhibit the tyrosine kinase activity of EGFR leading to the impediment of human lung adenocarcinoma cell motility and survival. J. Cell. Biochem. 2019, 120, 18077–18087. [Google Scholar] [CrossRef]
  138. Chen, T.; Liu, X.; Hong, H.; Wei, H. Novel single-domain antibodies against the EGFR domain III epitope exhibit the anti-tumor effect. J. Transl. Med. 2020, 18, 376. [Google Scholar] [CrossRef]
  139. Lamtha, T.; Krobthong, S.; Yingchutrakul, Y.; Samutrtai, P.; Gerner, C.; Tabtimmai, L.; Choowongkomon, K. A novel nanobody as therapeutics target for EGFR-positive colorectal cancer therapy: Exploring the effects of the nanobody on SW480 cells using proteomics approach. Proteome Sci. 2022, 20, 9. [Google Scholar] [CrossRef] [PubMed]
  140. Su, Z.; Han, Y.; Sun, Q.; Wang, X.; Xu, T.; Xie, W.; Huang, X. Anti-MET VHH Pool Overcomes MET-Targeted Cancer Therapeutic Resistance. Mol. Cancer Ther. 2019, 18, 100–111. [Google Scholar] [CrossRef]
  141. Zhang, C.; Tang, Q.; Xia, H.; Xu, H.; Bi, F. Identification and characterization of inhibitory nanobody against p38delta. Biochem. Biophys. Res. Commun. 2022, 600, 60–66. [Google Scholar] [CrossRef]
  142. Zhang, N.; Guo, H.; Zheng, W.; Wang, T.; Ma, X. Design and screening of a chimeric survivin-specific nanobody and its anticancer activities in vitro. Anticancer. Drugs 2016, 27, 839–847. [Google Scholar] [CrossRef]
  143. Meltzer, M.; Eliash, N.; Azoulay, Z.; Hadad, U.; Papo, N. In vitro inhibition of cancer angiogenesis and migration by a nanobody that targets the orphan receptor Tie1. Cell. Mol. Life Sci. 2022, 79, 312. [Google Scholar] [CrossRef]
  144. Teicher, B.A. Antibody-drug conjugate targets. Curr. Cancer Drug Targets 2009, 9, 982–1004. [Google Scholar] [CrossRef]
  145. Jin, R.; Liu, L.; Xing, Y.; Meng, T.; Ma, L.; Pei, J.; Cong, Y.; Zhang, X.; Ren, Z.; Wang, X.; et al. Dual Mechanisms of Novel CD73-Targeted Antibody and Antibody-Drug Conjugate in Inhibiting Lung Tumor Growth and Promoting Antitumor Immune-Effector Function. Mol. Cancer Ther. 2020, 19, 2340–2352. [Google Scholar] [CrossRef] [PubMed]
  146. Goldenberg, D.M.; Sharkey, R.M. Sacituzumab govitecan, a novel, third-generation, antibody-drug conjugate (ADC) for cancer therapy. Expert. Opin. Biol. Ther. 2020, 20, 871–885. [Google Scholar] [CrossRef]
  147. Wu, Y.; Li, Q.; Kong, Y.; Wang, Z.; Lei, C.; Li, J.; Ding, L.; Wang, C.; Cheng, Y.; Wei, Y.; et al. A highly stable human single-domain antibody-drug conjugate exhibits superior penetration and treatment of solid tumors. Mol. Ther. 2022, 30, 2785–2799. [Google Scholar] [CrossRef]
  148. Dean, A.Q.; Luo, S.; Twomey, J.D.; Zhang, B. Targeting cancer with antibody-drug conjugates: Promises and challenges. MAbs 2021, 13, 1951427. [Google Scholar] [CrossRef] [PubMed]
  149. Espelin, C.W.; Leonard, S.C.; Geretti, E.; Wickham, T.J.; Hendriks, B.S. Dual HER2 Targeting with Trastuzumab and Liposomal-Encapsulated Doxorubicin (MM-302) Demonstrates Synergistic Antitumor Activity in Breast and Gastric Cancer. Cancer Res. 2016, 76, 1517–1527. [Google Scholar] [CrossRef]
  150. Rosenfeld, L.; Sananes, A.; Zur, Y.; Cohen, S.; Dhara, K.; Gelkop, S.; Ben Zeev, E.; Shahar, A.; Lobel, L.; Akabayov, B.; et al. Nanobodies Targeting Prostate-Specific Membrane Antigen for the Imaging and Therapy of Prostate Cancer. J. Med. Chem. 2020, 63, 7601–7615. [Google Scholar] [CrossRef] [PubMed]
  151. Shajari, S.; Farajollahi, M.M.; Behdani, M.; Tarighi, P. Production and Conjugation of Truncated Recombinant Diphtheria Toxin to VEGFR-2 Specific Nanobody and Evaluation of its Cytotoxic Effect on PC-3 Cell Line. Mol. Biotechnol. 2022, 64, 1218–1226. [Google Scholar] [CrossRef]
  152. Li, R.; Zhu, X.; Zhou, P.; Qiao, Y.; Li, Y.; Xu, Y.; Shi, X. Generation of a High-Affinity Nanobody Against CD147 for Tumor Targeting and Therapeutic Efficacy Through Conjugating Doxorubicin. Front. Immunol. 2022, 13, 852700. [Google Scholar] [CrossRef]
  153. Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef]
  154. Golubovskaya, V. CAR-T Cells Targeting Immune Checkpoint Pathway Players. Front. Biosci. 2022, 27, 121. [Google Scholar] [CrossRef] [PubMed]
  155. Titov, A.; Zmievskaya, E.; Ganeeva, I.; Valiullina, A.; Petukhov, A.; Rakhmatullina, A.; Miftakhova, R.; Fainshtein, M.; Rizvanov, A.; Bulatov, E. Adoptive Immunotherapy beyond CAR T-Cells. Cancers 2021, 13, 743. [Google Scholar] [CrossRef] [PubMed]
  156. Berdeja, J.G.; Madduri, D.; Usmani, S.Z.; Jakubowiak, A.; Agha, M.; Cohen, A.D.; Stewart, A.K.; Hari, P.; Htut, M.; Lesokhin, A.; et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): A phase 1b/2 open-label study. Lancet 2021, 398, 314–324. [Google Scholar] [CrossRef]
  157. Wagner, D.L.; Fritsche, E.; Pulsipher, M.A.; Ahmed, N.; Hamieh, M.; Hegde, M.; Ruella, M.; Savoldo, B.; Shah, N.N.; Turtle, C.J.; et al. Immunogenicity of CAR T cells in cancer therapy. Nat. Rev. Clin. Oncol. 2021, 18, 379–393. [Google Scholar] [CrossRef] [PubMed]
  158. Delgoffe, G.M.; Xu, C.; Mackall, C.L.; Green, M.R.; Gottschalk, S.; Speiser, D.E.; Zehn, D.; Beavis, P.A. The role of exhaustion in CAR T cell therapy. Cancer Cell 2021, 39, 885–888. [Google Scholar] [CrossRef]
  159. Lynn, R.C.; Weber, E.W.; Sotillo, E.; Gennert, D.; Xu, P.; Good, Z.; Anbunathan, H.; Lattin, J.; Jones, R.; Tieu, V.; et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 2019, 576, 293–300. [Google Scholar] [CrossRef] [PubMed]
  160. Xie, Y.J.; Dougan, M.; Jailkhani, N.; Ingram, J.; Fang, T.; Kummer, L.; Momin, N.; Pishesha, N.; Rickelt, S.; Hynes, R.O.; et al. Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc. Natl. Acad. Sci. USA 2019, 116, 7624–7631. [Google Scholar] [CrossRef]
  161. Xie, Y.J.; Dougan, M.; Ingram, J.R.; Pishesha, N.; Fang, T.; Momin, N.; Ploegh, H.L. Improved Antitumor Efficacy of Chimeric Antigen Receptor T Cells that Secrete Single-Domain Antibody Fragments. Cancer Immunol. Res. 2020, 8, 518–529. [Google Scholar] [CrossRef]
  162. Rajabzadeh, A.; Rahbarizadeh, F.; Ahmadvand, D.; Kabir Salmani, M.; Hamidieh, A.A. A VHH-Based Anti-MUC1 Chimeric Antigen Receptor for Specific Retargeting of Human Primary T Cells to MUC1-Positive Cancer Cells. Cell J. 2021, 22, 502–513. [Google Scholar] [CrossRef]
  163. Hanssens, H.; Meeus, F.; De Veirman, K.; Breckpot, K.; Devoogdt, N. The antigen-binding moiety in the driver’s seat of CARs. Med. Res. Rev. 2022, 42, 306–342. [Google Scholar] [CrossRef]
  164. Ji, X.; Peng, Z.; Li, X.; Yan, Z.; Yang, Y.; Qiao, Z.; Liu, Y. Neutralization of TNFα in tumor with a novel nanobody potentiates paclitaxel-therapy and inhibits metastasis in breast cancer. Cancer Lett. 2017, 386, 24–34. [Google Scholar] [CrossRef] [PubMed]
  165. Karami, E.; Sabatier, J.M.; Behdani, M.; Irani, S.; Kazemi-Lomedasht, F. A nanobody-derived mimotope against VEGF inhibits cancer angiogenesis. J. Enzym. Inhib. Med. Chem. 2020, 35, 1233–1239. [Google Scholar] [CrossRef]
  166. Farajpour, Z.; Rahbarizadeh, F.; Kazemi, B.; Ahmadvand, D. A nanobody directed to a functional epitope on VEGF, as a novel strategy for cancer treatment. Biochem. Biophys. Res. Commun. 2014, 446, 132–136. [Google Scholar] [CrossRef] [PubMed]
  167. Deng, C.; Xiong, J.; Gu, X.; Chen, X.; Wu, S.; Wang, Z.; Wang, D.; Tu, J.; Xie, J. Novel recombinant immunotoxin of EGFR specific nanobody fused with cucurmosin, construction and antitumor efficiency in vitro. Oncotarget 2017, 8, 38568–38580. [Google Scholar] [CrossRef]
  168. Mohammadlou, M.; Salehi, S.; Baharlou, R. Development of anti DLL4 Nanobody fused to truncated form of Pseudomonas exotoxin: As a novel immunotoxin to inhibit of cell proliferation and neovascularization. Anal. Biochem. 2022, 653, 114776. [Google Scholar] [CrossRef]
  169. Ji, F.; Sha, H.; Meng, F.; Zhu, A.; Ding, N.; Zhang, H.; Xu, H.; Qian, H.; Yu, L.; Liu, Q.; et al. Tumor-penetrating peptide fused EGFR single-domain antibody enhances radiation responses following EGFR inhibition in gastric cancer. Oncol. Rep. 2018, 40, 1583–1591. [Google Scholar] [CrossRef]
  170. Dougan, M.; Ingram, J.R.; Jeong, H.J.; Mosaheb, M.M.; Bruck, P.T.; Ali, L.; Pishesha, N.; Blomberg, O.; Tyler, P.M.; Servos, M.M.; et al. Targeting Cytokine Therapy to the Pancreatic Tumor Microenvironment Using PD-L1-Specific VHHs. Cancer Immunol. Res. 2018, 6, 389–401. [Google Scholar] [CrossRef] [PubMed]
  171. Ji, X.; Han, T.; Kang, N.; Huang, S.; Liu, Y. Preparation of RGD4C fused anti-TNFα nanobody and inhibitory activity on triple-negative breast cancer in vivo. Life Sci. 2020, 260, 118274. [Google Scholar] [CrossRef] [PubMed]
  172. Yin, W.; Yu, X.; Kang, X.; Zhao, Y.; Zhao, P.; Jin, H.; Fu, X.; Wan, Y.; Peng, C.; Huang, Y. Remodeling Tumor-Associated Macrophages and Neovascularization Overcomes EGFR(T790M) -Associated Drug Resistance by PD-L1 Nanobody-Mediated Codelivery. Small 2018, 14, e1802372. [Google Scholar] [CrossRef] [PubMed]
  173. Woodham, A.W.; Cheloha, R.W.; Ling, J.; Rashidian, M.; Kolifrath, S.C.; Mesyngier, M.; Duarte, J.N.; Bader, J.M.; Skeate, J.G.; Da Silva, D.M.; et al. Nanobody-Antigen Conjugates Elicit HPV-Specific Antitumor Immune Responses. Cancer Immunol. Res. 2018, 6, 870–880. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, B.; Shu, Y.; Hu, S.; Qi, Z.; Chen, Y.; Ma, J.; Wang, Y.; Cheng, P. In Situ Tumor Vaccine Expressing Anti-CD47 Antibody Enhances Antitumor Immunity. Front. Oncol. 2022, 12, 897561. [Google Scholar] [CrossRef]
  175. Chen, F.; Liu, Z.; Jiang, F. Prospects of Neutralizing Nanobodies Against SARS-CoV-2. Front. Immunol. 2021, 12, 690742. [Google Scholar] [CrossRef]
  176. Güttler, T.; Aksu, M.; Dickmanns, A.; Stegmann, K.M.; Gregor, K.; Rees, R.; Taxer, W.; Rymarenko, O.; Schünemann, J.; Dienemann, C.; et al. Neutralization of SARS-CoV-2 by highly potent, hyperthermostable, and mutation-tolerant nanobodies. EMBO J. 2021, 40, e107985. [Google Scholar] [CrossRef] [PubMed]
  177. Zhao, L.; Liu, C.; Xing, Y.; He, J.; O’Doherty, J.; Huang, W.; Zhao, J. Development of a 99mTc-Labeled Single-Domain Antibody for SPECT/CT Assessment of HER2 Expression in Breast Cancer. Mol. Pharm. 2021, 18, 3616–3622. [Google Scholar] [CrossRef]
  178. Wong, N.C.; Cai, Y.; Meszaros, L.K.; Biersack, H.J.; Cook, G.J.; Ting, H.H.; Mottaghy, F.M. Preclinical development and characterisation of 99mTc-NM-01 for SPECT/CT imaging of human PD-L1. Am. J. Nucl. Med. Mol. Imaging 2021, 11, 154–166. [Google Scholar]
  179. Xing, Y.; Chand, G.; Liu, C.; Cook, G.J.R.; O’Doherty, J.; Zhao, L.; Wong, N.C.L.; Meszaros, L.K.; Ting, H.H.; Zhao, J. Early Phase I Study of a 99mTc-Labeled Anti-Programmed Death Ligand-1 (PD-L1) Single-Domain Antibody in SPECT/CT Assessment of PD-L1 Expression in Non-Small Cell Lung Cancer. J. Nucl. Med. 2019, 60, 1213–1220. [Google Scholar] [CrossRef] [PubMed]
  180. Hu, B.; Liu, T.; Li, L.; Shi, L.; Yao, M.; Li, C.; Ma, X.; Zhu, H.; Jia, B.; Wang, F. IgG-Binding Nanobody Capable of Prolonging Nanobody-Based Radiotracer Plasma Half-Life and Enhancing the Efficacy of Tumor-Targeted Radionuclide Therapy. Bioconjug. Chem. 2022, 33, 1328–1339. [Google Scholar] [CrossRef]
  181. Alauddin, M.M.; Khawli, L.A. Advances in Immuno-PET for the Detection of Cancer and Assessment of Response to Therapy. Curr. Med. Chem. 2021, 28, 647–672. [Google Scholar] [CrossRef]
  182. Varasteh, Z.; Braeuer, M.; Mohanta, S.; Steinsiek, A.L.; Habenicht, A.; Omidvari, N.; Topping, G.J.; Rischpler, C.; Weber, W.A.; Sager, H.B.; et al. In vivo Visualization of M2 Macrophages in the Myocardium After Myocardial Infarction (MI) Using (68) Ga-NOTA-Anti-MMR Nb: Targeting Mannose Receptor (MR, CD206) on M2 Macrophages. Front. Cardiovasc. Med. 2022, 9, 889963. [Google Scholar] [CrossRef]
  183. Varasteh, Z.; Mohanta, S.; Li, Y.; López Armbruster, N.; Braeuer, M.; Nekolla, S.G.; Habenicht, A.; Sager, H.B.; Raes, G.; Weber, W. Targeting mannose receptor expression on macrophages in atherosclerotic plaques of apolipoprotein E-knockout mice using 68Ga-NOTA-anti-MMR nanobody: Non-invasive imaging of atherosclerotic plaques. EJNMMI Res. 2019, 9, 5. [Google Scholar] [CrossRef] [PubMed]
  184. Fares, J.; Kanojia, D.; Rashidi, A.; Ahmed, A.U.; Balyasnikova, I.V.; Lesniak, M.S. Diagnostic Clinical Trials in Breast Cancer Brain Metastases: Barriers and Innovations. Clin. Breast Cancer 2019, 19, 383–391. [Google Scholar] [CrossRef] [PubMed]
  185. D’Huyvetter, M.; Vos, J.; Caveliers, V.; Vaneycken, I.; Heemskerk, J.; Duhoux, F.P.; Fontaine, C.; Vanhoeij, M.; Windhorst, A.D.; Aa, F.V.; et al. Phase I Trial of 131I-GMIB-Anti-HER2-VHH1, a New Promising Candidate for HER2-Targeted Radionuclide Therapy in Breast Cancer Patients. J. Nucl. Med. 2021, 62, 1097–1105. [Google Scholar] [CrossRef] [PubMed]
  186. Chocarro, L.; Arasanz, H.; Fernández-Rubio, L.; Blanco, E.; Echaide, M.; Bocanegra, A.; Teijeira, L.; Garnica, M.; Morilla, I.; Martínez-Aguillo, M.; et al. CAR-T Cells for the Treatment of Lung Cancer. Life 2022, 12, 561. [Google Scholar] [CrossRef] [PubMed]
  187. Lisi, L.; Lacal, P.M.; Martire, M.; Navarra, P.; Graziani, G. Clinical experience with CTLA-4 blockade for cancer immunotherapy: From the monospecific monoclonal antibody ipilimumab to probodies and bispecific molecules targeting the tumor microenvironment. Pharmacol. Res. 2022, 175, 105997. [Google Scholar] [CrossRef] [PubMed]
  188. Ma, B.T.-w.; Dai, V.; Greenberg, J.; Ballman, K.V.; Li, L.; Garcia, C.A.; Scheff, R.J.; Saxena, A.; Giaccone, G. Phase II study of KN046 in patients with thymic carcinoma who failed immune checkpoint inhibitors. J. Clin. Oncol. 2022, 40. [Google Scholar] [CrossRef]
  189. Zhang, F. Structural Biology of anti-PD-L1 and anti-CTLA4 Antibodies in Immune Therapies for Cancer Treatments. Ph.D. Thesis, Shanghai Jiaotong University, Shanghai, China, 2018. [Google Scholar]
  190. Papadopoulos, K.P.; Harb, W.; Lu, N.; Ma, X.; He, Y.; Yuan, L.; Fu, M.; Lin, Y.; Xu, W.; Wang, X.; et al. Phase I study of KN035, a novel fusion Anti-PD-L1 antibody administered subcutaneously in patients with advanced solid tumors in the USA. Ann. Oncol. 2018, 29, viii405. [Google Scholar] [CrossRef]
  191. Zhang, F.; Wei, H.; Wang, X.; Bai, Y.; Wang, P.; Wu, J.; Jiang, X.; Wang, Y.; Cai, H.; Xu, T.; et al. Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade. Cell Discov. 2017, 3, 17004. [Google Scholar] [CrossRef]
  192. Hong, D.; SciMentum, S.; Tanyi, J.; MacMullen, L.; Jalbert, L.; Muzithras, V.; Zikaras, K.; Cao, L.; O’Cearbhaill, R.; Quintás-Cardama, A. 959O Gavocabtagene autoleucel (gavo-cel, TC-210) dose escalation in refractory mesothelin-expressing solid tumors. Ann. Oncol. 2021, 32, S830. [Google Scholar] [CrossRef]
  193. Markham, A. Envafolimab: First Approval. Drugs 2022, 82, 235–240. [Google Scholar] [CrossRef]
  194. Zhao, L.; Liu, C.; Xing, Y.; He, J.; O’Doherty, J.; Huang, W.; Zhao, J. First-in-Human Study of a 99mTc-Labeled Single-Domain Antibody for SPECT/CT Assessment of HER2 Expression in Breast Cancer. 2021. Available online: https://www.researchsquare.com/article/rs-356098/v1 (accessed on 15 April 2022).
  195. D’Angelo, S.P.; Robinson, S.I.; Lam, J.; Adams, B.J.; Freddo, J.L.; Theuer, C.P.; Maki, R.G. ENVASARC: A pivotal trial of envafolimab, and envafolimab in combination with ipilimumab, in patients with advanced or metastatic undifferentiated pleomorphic sarcoma or myxofibrosarcoma who have progressed on prior chemotherapy. J. Clin. Oncol. 2021, 39, TPS11581. [Google Scholar] [CrossRef]
  196. Wang, Y.; Wang, C.; Huang, M.; Qin, S.; Zhao, J.; Sang, S.; Zheng, M.; Bian, Y.; Huang, C.; Zhang, H.; et al. Pilot study of a novel nanobody (68) Ga-NODAGA-SNA006 for instant PET imaging of CD8(+) T cells. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 4394–4405. [Google Scholar] [CrossRef] [PubMed]
  197. Fang, Y.L.; Yao, G.F.; Zhong, W.X.; Duan, W.W.; Zhou, Z.X.; Wen, X.F.; Chen, Y.L.; Fang, J.J.; Wang, Y.W.; Jiang, W.Q.; et al. DR30303, a SMART-VHHBody powered anti-CLDN18.2 VHH-Fc with enhanced ADCC activity for the treatment of gastric and pancreatic cancers. Cancer Res. 2022, 82, 2857. [Google Scholar] [CrossRef]
Figure 1. General structural comparison between conventional antibodies, heavy chain-only antibodies, single-chain Fv, and VHH. Abbreviations: Fab—Fragment of Antigen Binding; Fc—Fragment Crystallizable; CH—Constant domain of the Heavy chain; single-chain Fv; VH—Variable domain of the Heavy chain; CL—Constant domain of the Light chain; VL—Variable domain of the Light chain; VHH—Variable domain of the Heavy chain of Heavy-chain antibody.
Figure 1. General structural comparison between conventional antibodies, heavy chain-only antibodies, single-chain Fv, and VHH. Abbreviations: Fab—Fragment of Antigen Binding; Fc—Fragment Crystallizable; CH—Constant domain of the Heavy chain; single-chain Fv; VH—Variable domain of the Heavy chain; CL—Constant domain of the Light chain; VL—Variable domain of the Light chain; VHH—Variable domain of the Heavy chain of Heavy-chain antibody.
Cancers 16 00371 g001
Figure 2. A brief description of VHHs in anti-cancer therapies. TAA—Tumor Associated Antigen, PS—photosensitizer, IC—Immune Checkpoint.
Figure 2. A brief description of VHHs in anti-cancer therapies. TAA—Tumor Associated Antigen, PS—photosensitizer, IC—Immune Checkpoint.
Cancers 16 00371 g002
Table 1. Preclinical studies using VHH-radionuclide conjugates for imaging.
Table 1. Preclinical studies using VHH-radionuclide conjugates for imaging.
TargetConjugationCancer ModelsMain FindingsReference
CD889ZrBrCaThe CD8+ T cells in solid tumors were monitored by 89Zr-labeled anti-CD8-VHH, which signal positively corresponded with ICI treatment response.[66]
CD818FALLImaging with the 18F-VHH enabled rapid visualization of CD8+ T cells within 1 h, while no visible tumor uptake was observed with the control VHH.[67]
PSMA111InCRPCRenal uptake was efficiently reduced by co-injection of gelofusine and lysine. Replacing the c-myc-his tag with the cysteine reduced renal uptake without loss of targeting.[68]
PD-L199mTcTC-1 (immortalized murine lung epithelial cell)VHH accumulation correlated with the levels of PD-L1 in tumors, even if PD-L1 expression was low.[26]
PD-L199mTcNSCLC[99mTc]Tc-HYNIC-KN035 displayed a high PD-L1 specificity both in vitro and in vivo, that was positively correlated with the expression of PD-L1.[69]
PD-L168GaSKCM, BrCa68Ga-NOTA-Nb109 specifically accumulated in tumors with a maximum uptake of 5 ± 0.35% injected dose/g at 1 h.[70]
PD-L168GaGBM, CRC, NSCLCTumor-to-muscle ratio (TMR) reached its peak at 40 min post-injection. The heart uptake was almost fully cleared at 35 min post-injection.[71]
MMR99mTcTS/A (murine mammary adenocarcinoma), 3LL-R (Lewis Lung carcinoma)Anti-MMR VHH targeted pro-angiogenic MMR-expressing TAMs with tumor uptake correlating with the amount of TAMs in the tumor.[72]
MMR68Ga3LL-RTMR was determined while no treatment-related toxicologically relevant changes or acute immunological reactions were observed. The tolerated dose was established to be >1.68 mg/kg body weight. The dosimetry levels for humans were calculated by using the data in mice.[73]
LAG-399mTcTC-1 (immortalized murine lung epithelial cell)The tumor uptake of VHHs 3132 and 3206 targeting LAG-3 was comparable with high contrast at 1 h post-injection.[74]
LAG-399mTcMC38 (murine CRC), MO4 (murine melanoma), and TC-1The radiolabeled anti-LAG-3 VHH detected LAG-3 expressing TILs 1 h post tracer injection.[75]
HER268GaHER2+ cancerA high tumor-to-organ ratio was measured at 1 h post-injection with increased uptake upon increasing the injected dose.[76]
HER218FOvCaThe tumor-to-organ ratio at 1 h post-injection showed excellent specificity.[25]
HER299mTcBrCaThe tumor had significant radiotracer uptake at 0.5 h after injection.[77]
Glypican-368Ga, 18FHCCThe fusion of VHH to an albumin-binding domain increased the tumor uptake and decreased kidney accumulation of the radiotracer (1 h to 6 h).[78]
EpCAM99mTcEpCAM driven cancerThe uptake value in tumors was increased about two times from 0.5 h till 12 h after injection, while it could clearly image tumor-draining lymph nodes.[79]
EGFR99mTcEGFR+ cancer, A431 (epidermoid carcinoma)VHH uptake correlated with tumor burden and tumor response to EGFR inhibitor (erlotinib).[34]
EGFR99mTcA431In vivo, the study demonstrated that OA-cb6 labeled with 99mTc showed an approximately 2.7-fold tumor-muscle ratio at 4 h post-injection.[80]
CLDN18.289ZrSTADThe VHH had good tumor uptake to evaluate the expression of CLDN18.2 in gastric cancer for patient selection.[81]
CEACAM599mTcNSCLCThe high ratio of the signal in the tumor compared with the background confirmed that the VHH can be used as a molecular probe for imaging CEACAM5-expressing tumors.[82]
CAIX111InHNSCCThe anti-CAIX VHH targeted hypoxia regions in solid tumors.[83]
* EDB of FN64Cupan-cancerTargeted the extracellular matrix to image tumor progression, metastasis, and fibrosis.[84]
* Alternatively spliced EIIIB domain of fibronectin.
Table 2. VHH application in radioimmunotherapy.
Table 2. VHH application in radioimmunotherapy.
TargetsConjugatesCancer ModelsReferences
HER2131I (β/γ)HER2+ cancer[98,99]
125I, 131I-SGMIB (β/γ)BrCa[100]
177Lu (β)OvCa[94]
211At (α)HER2+ cancer[101,102]
225Ac (α)SKOV3, BrCa[103]
211At (α)BrCa[102]
FAPα *89Zr (γ), 177Lu(β)FAPα+ cancer[96]
131I-SGMIB (β/γ)FAPα+ cancer[95]
225Ac (α)
MMR177Lu (β), 111In(γ)TS/A[97]
* The anti-FAPα VHH is fused with the Fc fragment.
Table 3. Preclinical studies investigating VHH-PS conjugates.
Table 3. Preclinical studies investigating VHH-PS conjugates.
TargetsConjugatesCancer ModelsMain FindingsReferences
HER2IRDye700DXSK-BR-3 (HER2+, sensitive), HCC1954, JIMT1, HCC1419 (HER2+, resistant), MCF7 (HER2 low), MDA-MB-231 (HER2−)Anti-HER2 VHH-PS could potently and selectively induce cell death in HER2-positive cells regardless of its sensitivity to trastuzumab.[106]
EGFRIRDye700DXA431The PS was conjugated with 111In-VHH in a site-specific way, which resulted in light-induced toxicity via cellular internalization.[105]
EGFRIRDye700DXCell lines with different EGFR expressionThe anti-EGFR VHH-PS led to approx. 90% tumor necrosis and almost no toxicity in healthy tissue 24 h after PDT.[107,108]
EGFRIRDye700DXA431, SCC-U8VHH-PS induced the release of DAMPs (HSP70, ATP) and the pro-inflammatory cytokines of moDCs by incubating it with a conditioned medium, which stimulates the immune system.[109]
METIRDye700DXMKN45The anti-MET VHH-PS had a nanomolar affinity and led to cell death at nanomolar concentration with illumination.[110]
US28IRDye700DXU251-iUS28The anti-US28 VHH-PS was the first example using GPCR as a target for VHH-directed PDT, which selectively killed US28-expressing glioblastoma cells.[7,32]
EGFR/VEGFR2IRDye700DXOSCCThe dual-targeting VHH-PS showed improved efficacy in co-culture of endothelial and cancer cells.[111]
Table 4. Preclinical studies using VHH-based ICIs.
Table 4. Preclinical studies using VHH-based ICIs.
TargetsCancer ModelsMain FindingsReferences
PD-L1+CD16a+IL15PD-L1+ cancerThe fusion promoted cell growth in vitro, while it attenuated tumor growth in vivo.[120]
PD-L1PaCaThe VHH-CCL21 fusion could target PD-L1 positive TME and promote recruiting effector cells.[121]
PD-L1MC38VHH outperformed conventional antibodies in inhibiting tumor growth due to VHH’s higher tumor penetration in the MC38 tumor.[113]
PD-L1PD-L1+ cancerMonovalent, bivalent, and trivalent agents enhanced TCR signaling in PD-L1 positive cancer cells, to result in CD8+ T cell activation and cytokine production to attenuate cancer progression.[122,123,124]
PD-L1+TIGITMC38The multivalent bispecific VHH could synergistically enhance T cell activity by inhibiting tumor growth in vitro.[116]
PD-1A549, BxPC3The VHH could block the PD-1/PD-L1 interaction.[125]
PD-1MC38Long-term systemic expression of VHH by AAV vector provided anti-tumor activity without toxicity.[126]
CTLA-4MelanomaThe anti-CTLA4 VHH delayed melanoma growth and prolonged the survival time in mice.[127]
CTLA-4MC38, H22The half-life-extended version of VHH exhibited therapeutic efficacy in a Fc independent manner.[114]
4-1BB+PD-L1CT-26-huPD-L1, MC-38-huPD-L1The bispecific VHH showed anti-tumor efficacy with negligible hepatotoxicity.[55]
Table 5. Preclinical studies evaluating the therapeutic efficacy of anti-TAAs VHHs in solid cancer.
Table 5. Preclinical studies evaluating the therapeutic efficacy of anti-TAAs VHHs in solid cancer.
TargetCancer ModelsMain FindingsReference
CapGMDA-MB-231Anti-CapG VHH prevented the formation of lung metastasis.[131]
CD38MelanomaAnti-CD38 VHH Pseudomonas exotoxin A (PE38) showed highly selective cytotoxicity. The effectiveness could be increased by retinoid acid.[132]
CD47MelanomaAnti-CTLA4 VHH synergized with other immune therapies when CD47 in TME was near-completely blocked.[133]
CEACAM5/CD3LS174T, SKOV3The in vivo half-life of the bispecific VHH was increased 12-fold via the PEGylation strategy, accompanied by more potent tumor inhibition.[45]
CXCR7HNSCCThe anti-CXCR7 VHH inhibited tumor growth by reducing the secretion of CXCL1 in vitro and inhibiting angiogenesis in vivo.[134]
DLL4MKN, HEK293The DLL4 could bind on the surface of MKN cells, and gastric carcinoma tissue and inhibit the maturation of capillary-like structures in HUVECs.[51]
DR5Hela, Colo205Multivalent anti-DR5 VHHs had higher apoptotic capacity than the monovalent form that could mimic the activity of the natural TRAIL ligand.[135]
DPYSL2, TUFM, Vimentin, NAP1-L1GBMThe anti-TUFM VHH showed a cytotoxic effect on GBM CSCs, while other VHHs were shown to target mature GBM cells.[136]
EGFRLUADVHH was linked with the cell-penetrating peptide nonaarginine. The VHH inhibited intracellular signaling by binding EGFR resulting in reduced cell migration.[137]
EGFRA549, DU145, MCF-7The anti-EGFR extracellular domain III VHH showed an anti-tumor effect both in vitro and in vivo.[138]
EGFRSW480VHH could inhibit cancer cell viability by altering proteins involved in the DNA-damage checkpoint process.[139]
METHepG2, SK-HEP-1, HCC827, NIH3T3Anti-MET VHH pool that acts against the whole ectodomain of MET could overcome MET targeted treatment resistance by promoting MET degradation and blocking the kinase activity of MET. The anti-MET VHH treatment could suppress cancer proliferation, viability, and colony formation in vitro and tumorigenesis in vivo.[140]
p38δHelaThe VHH inhibited the target kinase activity and tumor growth.[141]
SurvivinHepG2The VHH targeted survivin and blocked the signaling pathway resulting in apoptosis.[142]
Tie1U87MGTargeting Tie1 with specific VHH triggered Tie1-dependent inhibition of RTK phosphorylation and angiogenesis in endothelial cells and suppressed GBM viability and migration.[143]
Table 6. Studies evaluating the therapeutic potential of VHH-drug conjugates.
Table 6. Studies evaluating the therapeutic potential of VHH-drug conjugates.
TargetCargoCancer ModelsMain FindingsReference
EGFRMal-PtA375, A431The VHH-DC could be specifically internalized into EGFR-positive cancer cells, resulting in higher therapeutic effects and lower side effects compared with cisplatin alone.[89]
PSMADoxorubicinPC3-PIP, PC3-flu *An in vivo study showed that a 42-fold lower amount of VHH-DC could result in similar tumor growth inhibition compared with commercial doxorubicin treatment.[150]
HER2DoxorubicinBT474-M3, NCI-N87VHH-DC could simultaneously bind with the HER2 target on cancer cells with trastuzumab, which results in synergistic antitumor activity.[149]
HER2Auristatin FBT474, MDA-MB-231VHH-DC-albumin fusion overcame the rapid renal clearance, which resulted in long-lasting tumor remission.[47]
VEGFR2Diphtheria ToxinPC3Coupling toxin with immune “carrier” resulted in cancer cell growth inhibition, while toxin alone was ineffective.[151]
CD147DoxorubicinHela, 4T1, U87, 293T(low), SMMC-7721In vitro studies showed the VHH-DC could inhibit tumor cell proliferation and induce cell apoptosis. The VHH-DC had a synergistic effect in inhibiting the growth of tumors in vivo, as compared with the treatment of doxorubicin or VHH monotherapy.[152]
5T4SN38BxPC-3, Huh-7N501-SN38 showed deeper tumor penetration, higher tumor uptake, and faster accumulation at the tumor site than conventional ADC and exhibited effective antitumor activity both in vitro and in vivo.[147]
* PC3-PIP and PC3-flu are PSMA positive or PSMA negative PC3, respectively.
Table 7. Overview of registered clinical trials of VHHs-based agents in the treatment of solid cancer.
Table 7. Overview of registered clinical trials of VHHs-based agents in the treatment of solid cancer.
AgentTargetCancer TypeStudy IdentifierPhaseStatusPrimary PurposeRelated Publication
99mTc-NM-02HER2Breast cancerNCT04040686Early Phase IRecruitingDiagnostic[177]
99mTc-NM-01PD-L1Non-Small Cell Lung CancerNCT02978196Early Phase IRecruitingDiagnostic[178,179]
99mTc-MIRC208HER2HER2 positive cancerNCT04591652Not ApplicableRecruitingDiagnostic[180]
89Zr-KN035PD-L1PD-L1 positive solid tumorNCT04977128Not ApplicableRecruitingDiagnostic[181]
68Ga-THP-APN09PD-L1Lung cancer
Melanoma
NCT05156515Not ApplicableRecruitingDiagnostic[181]
68Ga-NOTA-Anti-MMR-VHH2MMRBreast cancer
Head and Neck cancer
Melanoma (skin)
NCT04168528Phase I/IIaRecruitingDiagnostic[73,182,183]
68Ga-NOTA-Anti-MMR-VHH2MMRBreast cancer
Pancreatic cancer
Salivary gland cancer
Gastric cancer
Endometrial cancer
Uterine cancer
Non-Small Cell Lung Cancer
Biliary tract cancer
Cholangiocarcinoma
Colorectal cancer
Urothelial carcinoma
Prostate cancer
NCT03924466Phase IIRecruitingDiagnostic[73,182,183]
68Ga-NOTA-Anti-HER2 VHH1HER2Breast cancerNCT03924466Phase IIRecruitingDiagnostic[18,64,184]
68Ga-NOTA-Anti-HER2 VHH1HER2Breast cancerNCT03331601Phase IIRecruitingDiagnostic[18,64,184]
99mTc-NM01PD-L1Non-Small Cell Lung CancerNCT04992715Phase IIRecruitingDiagnostic[178,179]
131I-SGMIB Anti-HER2 VHH1HER2Breast cancerNCT02683083Phase ICompletedDiagnostic[185]
68Ga-ACN376CLDN18.2Solid tumorNCT05436093Not ApplicableRecruitingScreening
αPD1-MSLN-
CAR-T Cells
PD-1Solid tumorNCT05373147Early Phase IRecruitingTreatment[186]
αPD1-MSLN-
CAR-T Cells
PD-1Colorectal cancer
Ovarian cancer
NCT04503980Early Phase IRecruitingTreatment[186]
αPD1-MSLN-
CAR-T Cells
PD-1Non-small-cell Lung Cancer
Mesothelioma
NCT04489862Early Phase IRecruitingTreatment[186]
αPD1-MSLN-
CAR-T Cells
PD-1Colorectal cancerNCT05089266Phase INot yet recruitingTreatment[186]
KN046+AxitinibPD-L1/CTLA4 BispecificAdvanced Non-small Cell Lung cancerNCT05420220Phase IINot yet recruitingTreatment[187]
KN046PD-L1/CTLA4 BispecificThymic carcinomaNCT04469725Phase IIRecruitingTreatment[188]
KN044CTLA4Advanced solid tumorNCT04126590Phase IRecruitingTreatment[189]
KN035PD-L1Solid tumorNCT03101488Phase ICompletedTreatment[190,191]
KN035PD-L1Advanced or metastatic solid tumorNCT03248843Phase ICompletedTreatment[190,191]
JS014
(fusion with IL-21)
+ Pembrolizumab
Human Serum AlbuminMalignant neoplasm Experimental solid tumor
Adult lymphoma
NCT05296772Phase IActive, not recruitingTreatment
Gavocabtagene
autoleucel
(gavo-cel; TC-210)
MesothelinMesotheliomaNCT03907852Phase I Phase IIRecruitingTreatment[192]
Envofolimab (KN035)+Gemcitabine and CisplatinPD-L1Biliary tract cancerNCT04910386Phase IINot yet recruitingTreatment[193]
99mTc-NM-02,
188Re-NM-02
HER2Breast cancerNCT04674722Early
Phase I
RecruitingTreatment[177,194]
Envafolimab (+Ipilimumab)PD-L1Pleomorphic sarcoma MyxofibrosarcomaNCT04480502Phase IIRecruitingTreatment[195]
68Ga-NODAGA-SNA006CD8αSolid tumorsNCT05126927Early
Phase I
RecruitingDiagnostic[196]
DR30303-IgG1FcCLDN18.2Malignant neoplasm of the digestive systemNCT05639153Phase IRecruitingTreatment[197]
[99mTc]-NM-01PD-L1Non-small cell lung cancer, malignant melanomaNCT04436406Not ApplicableRecruitingDiagnostic
68Ga-PD-L2PD-L2Colorectal cancer, Lung cancerNCT05803746Not ApplicableRecruitingDiagnostic
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

Cong, Y.; Devoogdt, N.; Lambin, P.; Dubois, L.J.; Yaromina, A. Promising Diagnostic and Therapeutic Approaches Based on VHHs for Cancer Management. Cancers 2024, 16, 371. https://doi.org/10.3390/cancers16020371

AMA Style

Cong Y, Devoogdt N, Lambin P, Dubois LJ, Yaromina A. Promising Diagnostic and Therapeutic Approaches Based on VHHs for Cancer Management. Cancers. 2024; 16(2):371. https://doi.org/10.3390/cancers16020371

Chicago/Turabian Style

Cong, Ying, Nick Devoogdt, Philippe Lambin, Ludwig J. Dubois, and Ala Yaromina. 2024. "Promising Diagnostic and Therapeutic Approaches Based on VHHs for Cancer Management" Cancers 16, no. 2: 371. https://doi.org/10.3390/cancers16020371

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