Open Access

Epithelial cell adhesion molecule‑targeting designed ankyrin repeat protein‑toxin fusion Ec1‑LoPE exhibits potent cytotoxic action in prostate cancer cells

  • Authors:
    • Tianqi Xu
    • Yongsheng Liu
    • Alexey Schulga
    • Elena Konovalova
    • Sergey M. Deyev
    • Vladimir Tolmachev
    • Anzhelika Vorobyeva
  • View Affiliations

  • Published online on: March 18, 2022     https://doi.org/10.3892/or.2022.8305
  • Article Number: 94
  • Copyright: © Xu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Targeted anticancer therapeutics offer the advantage of reducing cytotoxic side effects to normal cells by directing the cytotoxic payload selectively to cancer cells. Designed ankyrin repeat proteins (DARPins) are promising non‑immunoglobulin‑based scaffold proteins for payload delivery to cancer‑associated molecular targets. Epithelial cell adhesion molecule (EpCAM) is overexpressed in 40‑60% of prostate cancers (PCs) and is associated with metastasis, increased risk of PC recurrence and resistance to treatment. Here, we investigated the use of DARPin Ec1 for targeted delivery of Pseudomonas exotoxin A variant (LoPE) with low immunogenicity and low non‑specific toxicity to EpCAM‑expressing prostate cancer cells. Ec1‑LoPE fusion protein was radiolabeled with tricarbonyl technetium‑99m and its binding specificity, binding kinetics, cellular processing, internalization and cytotoxicity were evaluated in PC‑3 and DU145 cell lines. Ec1‑LoPE showed EpCAM‑specific binding to EpCAM‑expressing prostate cancer cells. Rapid internalization mediated potent cytotoxic effect with picomolar IC50 values in both studied cell lines. Taken together, these data support further evaluation of Ec1‑LoPE in a therapeutic setting in a prostate cancer model in vivo.

Introduction

Efficient treatment of metastatic prostate cancer (PC) remains a current clinical need (1). Initial androgen deprivation therapy provides stabilization of disease for several years; however, the development of resistance occurs in the large majority of patients with time (2). Insensitivity to existing treatments leads to disease progression, which necessitates the identification of novel molecular targets and targeted therapeutic approaches.

Prostate-specific membrane antigen (PSMA)-targeted therapy using radioligands, such as [177Lu]Lu-PSMA-617, has demonstrated promising results in clinical trials and improved outcomes for patients with metastatic castration-resistant PC (3). However, patients with low PSMA expression are not eligible for this therapy (4); some patients do not respond to it and some develop resistance despite initially good response (5).

Epithelial cell adhesion molecule (EpCAM) is a potential therapeutic target in PC. EpCAM is a type I transmembrane glycoprotein, which participates in cell adhesion and proliferation, and was reported to be involved in oncogenic signaling by engagement of elements of the Wnt pathway (6). Upregulated EpCAM expression is detected in early stages of PC and is further induced in high-grade tumors and metastatic lesions (7,8). Intense EpCAM overexpression is found in 40–60% of prostate tumor samples (9) and is associated with metastasis and increased risk of PC recurrence (10,11). Due to its involvement in mechanisms regulating resistance to treatment, EpCAM has been suggested as a therapeutic target to sensitize PC cells to chemotherapy and radiotherapy (12). The EpCAM protein surface level was found to be up to 16-fold higher in PC cells compared to non-cancerous prostate cells and up to 4-fold higher in malignant compared to benign prostate tumors (7), which creates a potential therapeutic window for targeted cytotoxic delivery.

A number of therapeutic agents targeting EpCAM have been evaluated in preclinical and clinical trials for different types of cancer (13). They include monoclonal antibodies (mAbs), their fragments, as well as their conjugates with cytotoxic drugs and immunotoxins (14,15). The only EpCAM-targeting agent that has received approval in the European Union is the rat-mouse bispecific anti-EpCAM/anti-CD3 antibody catumaxomab for the treatment of patients with malignant ascites by intraperitoneal infusion (16). While being more effective than conventional treatment, it has shown limited efficacy in solid tumors, which could arise from several factors, such as heterogeneity of EpCAM expression in patients, limited penetration in the tumor due to the size of the targeting agent and limited cytotoxic effect.

One way to increase the potency of targeted therapy is conjugation of a cytotoxic payload, such as drugs, plant or bacterial toxins, to a targeting molecule for selective eradication of malignant cells. The limited therapeutic effect observed for EpCAM-targeting mAbs promoted the development of targeted immunotoxins (17). Several of them have reached clinical trials, such as the mouse mAb MOC31 and its scFv fragment (4D5MOCB) conjugated to Pseudomonas aeruginosa exotoxin A (PE) (VB4-845, oportuzumab monatox) (1821) and a Fab fragment conjugated with the plant-derived ribosome-inactivating protein de-bouganin (VB6-845, citatuzumab bogatox) (22). Still, there is a number of physiological barriers hampering the efficient tumor delivery of cytotoxic payloads using bulky immunoglobulins, and the use of smaller targeting vectors should be beneficial (17).

Designed ankyrin repeat proteins (DARPins) are a type of engineered scaffold proteins with promising tumor-targeting properties (23). Their use for tumor targeting offers a number of advantages in comparison with the traditional IgG scaffold. The small size (14–18 kDa) could enable more efficient interstitial transport, faster extravasation and deeper penetration of DARPins into the tumors compared to therapeutics based on the IgG scaffold (150 kDa) (24). Their high affinity ensures a strong binding to malignant cells and a good retention in tumors. Rapid excretion of DARPins from blood provides high imaging contrast already several hours after injection. DARPins have shown promising results in preclinical studies for imaging of human epidermal growth factor receptor 2 (HER2) (2528) and EpCAM expression (2932) and could be used as companion diagnostic agents during targeted therapy. The clinical trial evaluating anti-HER2 DARPin G3 labeled with technetium-99m showed its capacity of specific targeting HER2-positive breast cancers and demonstrated the safety of this protein scaffold in humans, with no toxicity or side effects observed (33).

Another advantage of DARPin scaffold over IgG includes its robustness, high solubility and thermodynamic stability. Conjugates of DARPins with protein-based toxins can be genetically engineered and produced as single protein fusions in large amounts with lower manufacturing costs (13,23,34).

In the present study, we investigated a fusion protein consisting of an EpCAM-targeting DARPin Ec1 and a re-engineered Pseudomonas exotoxin A variant (LoPE) bacterial toxin for cytotoxic therapy of prostate cancer. The N-terminal DARPin Ec1 (18 kDa) binds to EpCAM with picomolar affinity [KD 68 pM (35)], while the C-terminal LoPE toxin (25 kDa) irreversibly inhibits eukaryotic elongation factor eEF2 that leads to inhibition of protein synthesis in the cell (36). LoPE is the deimmunized C-terminal catalytic subunit of bacterial Pseudomonas aeruginosa Exotoxin A (PE toxin) with lower immunogenicity and general toxicity in vivo in comparison with previous versions of PE toxin (37).

The aim of the present study was to characterize the functional parameters of Ec1-LoPE, such as binding specificity and kinetics of binding to living cells, cellular processing, internalization and cytotoxicity in PC cell lines and to evaluate its potential for targeted therapy of PC in vivo.

Materials and methods

Cells

The human prostate cancer cell lines PC-3 and DU145 overexpressing EpCAM were purchased from the American Type Culture Collection (ATCC; LGC Promochem). The cells were cultured in Roswell Park Memorial Institute (RPMI)-1640 medium with L-glutamine (L0500, Biowest) supplemented with 10% fetal bovine serum (FBS) (F7524, Sigma-Aldrich; Merck KGaA) and penicillin-streptomycin solution (L0022, Biowest) at 37°C and 5% CO2 atmosphere, unless stated otherwise. Cells were detached using trypsin-ethylenediaminetetraacetic acid (EDTA) solution (25200056, Thermo Fisher Scientific, Inc.).

Targeting protein

Ec1-LoPE (Fig. 1) was produced and characterized as described previously (38). To confirm the identity, the molecular weight of the protein was determined using electrospray ionization mass spectrometry (Impact II instrument, Bruker Corp.). The spectrometer worked on line with Dionex UltiMate 3000 ultra-high performance liquid chromatography (UHPLC) system (Thermo Fisher Scientific, Inc.) equipped with a ProSwift RP-4H column (1×50 mm, Thermo Fisher Scientific, Inc.). The chromatographic system used two solvents: Solvent A (3% acetonitrile, 0.1% formic acid in water) and solvent B (95% acetonitrile, 0.1% formic acid in water), and the flow rate was 200 µl/min. The following gradient profile was used: 4% solvent B for 2 min, 4–90% solvent B within 6 min, 90% solvent B for 2 min, 90–4% solvent B within 1 min followed by 4% solvent B for 4 min. The found molecular weight (43,061 kDa) was in excellent agreement with the calculated molecular weight (43,061 kDa) (Fig. 2A). According to HPLC analysis, the purity of Ec1-LoPE was >99% (Fig. 2B).

Radiolabeling

Ec1-LoPE having (His)6-tag at the C-terminus was radiolabeled site-specifically with [99mTc][Tc(CO)3(H2O)3]+ as described previously (38). Briefly, technetium-99m pertechnetate [99mTc]TcO4 was obtained by eluting a commercial 99Mo/99mTc generator (Mallinckrodt) with sterile 0.9% NaCl. To generate the [99mTc][Tc(CO)3(H2O)3]+, approximately 500 µl of [99mTc]TcO4 eluate containing 3–4 GBq activity was added to a CRS kit vial (PSI) and incubated at 100°C for 30 min followed by cooling down at room temperature for 10 min. Solution of [99mTc][Tc(CO)3(H2O)3]+ (100 MBq, 15 µl) was mixed with 30 µl of 0.1 M HCl and added to 30 µg of Ec1-LoPE [25 µl in phosphate-buffered saline (PBS)]. The reaction mixture was incubated for 60 min at 40°C. A pre-purification challenge was performed by adding a 1,000-fold molar excess of histidine (110 µg in 11 µl of PBS) to the reaction mixture and incubating at 40°C for 10 min to remove any loosely-bound [99mTc]Tc(CO)3(H2O)3]+. The radiolabeled compound was separated from free [99mTc]Tc(CO)3(H2O)3]+ by passage through a NAP-5 size-exclusion column (GE Healthcare) pre-equilibrated and eluted with PBS. Radiochemical yield and purity of [99mTc]Tc(CO)3-Ec1-LoPE were measured using instant thin-layer chromatography (iTLC) silica gel strips eluted in PBS. In this system, the radiolabeled compound stays at the application point; all forms of free radionuclide move with the solvent front. The activity distribution along the iTLC strip was measured with a Storage Phosphor System (CR-35 BIO Plus, Elysia-Raytest) and analyzed with AIDA Image Analysis software (Elysia-Raytest).

In vitro stability test

The evaluation of radiolabel stability of [99mTc]Tc(CO)3-Ec1-LoPE was performed by incubating it with a 1,000-fold molar excess of histidine in PBS at room temperature for 4 h. The control samples were incubated in PBS. Samples were analyzed using radio-iTLC in PBS. The values were normalized to the starting radiochemical purity taken as 100%.

In vitro specificity

The binding specificity of [99mTc]Tc(CO)3-Ec1-LoPE to EpCAM-overexpressing cells PC-3 and DU145 was performed by using in vitro saturation assay (38). Briefly, the cells were seeded in a 6-well plate at a density of 5×105 cells per well one day before the experiment and allowed for attachment overnight. A set of three wells was used for each group. The next day, blocking was performed by incubating one group of cells with 200 nM of non-radiolabeled DARPin Ec1 in 0.5 ml of culture medium at room temperature for 15 min to saturate EpCAM receptors. The same volume of culture medium only was added to another group of cells. After 15 min, 0.5 ml of [99mTc]Tc(CO)3-Ec1-LoPE was added to each well to a final concentration of 2 nM and incubated at 37°C for 1 h. After incubation, medium was discarded, cells were washed once with 1 ml of PBS. The cells were then incubated with 0.5 ml of trypsin solution at 37°C to allow for detachment and resuspended with 0.5 ml of culture medium. The cell suspension was collected, counted for cell number, followed by a wash with 1 ml PBS, which was also collected. Three standard samples containing 0.5 ml of [99mTc]Tc(CO)3-Ec1-LoPE were collected. The activity in standard samples and fractions containing cells was measured using an automatic gamma spectrometer (2480 Wizard, Perkin Elmer), percentage of cell-associated activity per 1×106 cells was calculated.

Cellular processing

Cellular processing and internalization of [99mTc]Tc(CO)3-Ec1-LoPE by PC-3 and DU145 cells were evaluated during continuous incubation using acid wash method (39). Briefly, cells were seeded one day before the experiment in 3-cm Petri dishes at a density of 7×105 cells per dish. At the day of the experiment, the radiolabeled compound (2 nM, 1 ml) was added to each dish and the dishes were incubated at 37°C in a humidified incubator. Three standard samples containing 1 ml of [99mTc]Tc(CO)3-Ec1-LoPE were collected and used as total added activity. At 1, 2, 4 and 6 h a set of three dishes was removed, medium was discarded, cells were washed once with 1 ml of ice-cold PBS and discarded. To collect the membrane-bound activity, the cells were incubated with 1 ml of 0.2 M glycine buffer containing 4 M urea (pH 2.0) for 5 min on ice, washed with 1 ml of the same buffer and with 1 ml of PBS. These steps were performed on ice to prevent internalization. To collect the internalized activity, the cells were incubated with 1 ml of 1 M NaOH at 37°C for 30 min to lyse the cells. The cell lysate was collected, followed by washing with 1 ml of PBS. The activity in the standards, membrane-bound and internalized fractions was measured using a gamma spectrometer and percentage cell-associated activity was calculated.

Affinity

The binding affinity of [99mTc]Tc(CO)3-Ec1-LoPE to DU145 cells was measured using LigandTracer Yellow instrument (Ridgeview Diagnostics) as described previously (38). The experiment was performed in duplicate at room temperature to prevent internalization. In brief, 2×106 cells were seeded to a local area of an 89-mm Petri dish one day before the experiment. The binding kinetics was measured by adding increasing concentrations of the radiolabeled compound (3 and 9 nM) every 1.5 h. Thereafter, the medium containing the radiolabeled compound was replaced with cell culture medium to measure the dissociation rate. The binding curve was analyzed using TraceDrawer Software (Ridgeview Instruments, version 1.9) and the equilibrium dissociation constant (KD) was calculated. The interaction heterogeneity was estimated using Interaction Map analysis (Ridgeview Diagnostics).

Cytotoxicity

The in vitro cytotoxicity assay was performed using PC3 and DU-145 cells. Cells were seeded in a 96-well plate at a density of 5,000 cells per well in 100 µl culture medium and allowed to attach overnight. On the experimental day, the medium was removed, serial dilutions of Ec1-LoPE in culture medium were added for each concentration (n=4-5) and the plate was incubated in a humidified incubator. After 72 h, the medium was replaced, and cell viability was evaluated with the Cell Counting Kit-8 (CCK-8; Sigma-Aldrich; Merck KGaA) according to the manufacturer's protocol. Briefly, 10 µl of CCK-8 kit was added to each well containing 100 µl culture medium, followed by incubation at 37°C for 1 to 2 h. The absorbance at 450 nm was measured using a microplate reader. The value from the wells containing medium only was taken as the background; the value from the wells containing cells with medium (no Ec1-LoPE) was taken as 100% viability control. The data were analyzed by GraphPad Prism (version 9.0.2 for Windows, GraphPad Software, Inc.) using a log(inhibitor) vs. response-variable slope (four parameters) model to obtain a half-maximal inhibitory concentration (IC50) value. To test the specificity of Ec1-LoPE cytotoxic action, one group of cells was incubated with a 100-fold molar excess of DARPin Ec1 for 10 min before the addition of Ec1-LoPE (10 nM) to saturate binding sites on EpCAM and prevent binding of Ec1-LoPE. The cell viability was evaluated as described above.

Statistics

Data were analyzed using GraphPad Prism (version 9.0.2 for Windows, GraphPad Software, Inc.). Unpaired 2-tailed t-test was performed to determine significant differences (P<0.05).

Results

Radiolabeling and stability

Ec1-LoPE was successfully labeled with [99mTc]Tc(CO)3(H2O)3]+ with a radiochemical yield of 51±13% (n=7) and radiochemical purity over 99% (n=7) after purification using size-exclusion column (Table I). Radiolabeled [99mTc]Tc(CO)3-Ec1-LoPE demonstrated high stability during incubation with a 1,000-fold molar excess of histidine for up to 4 h at room temperature (Table I).

Table I.

Radiolabeling of EpCAM-targeting Ec1-LoPE with [99mTc]Tc(CO)3 and in vitro stability under a 1,000-fold molar excess of histidine compared to PBS (control).

Table I.

Radiolabeling of EpCAM-targeting Ec1-LoPE with [99mTc]Tc(CO)3 and in vitro stability under a 1,000-fold molar excess of histidine compared to PBS (control).

Protein-associated activity (%) (n=2)

1 h4 h


Radiochemical yield (%)Radiochemical purity (%)x1,000 HistidinePBS (control)x1,000 HistidinePBS (control)
[99mTc]Tc(CO)3-51±1399±0100±0100±0100±0100±0
Ec1-LoPE(n=7)(n=7)

[i] Stability analysis was performed in duplicate. Values are normalized to the starting radiochemical purity taken as 100%.

In vitro binding specificity

The specificity of [99mTc]Tc(CO)3-Ec1-LoPE binding to EpCAM-expressing PC-3 and DU145 cells was studied by saturating the EpCAM receptors with a large excess of unlabeled DARPin Ec1. A significant (P<0.001) reduction in cell-associated activity was observed in the groups where EpCAM was pre-saturated, indicating specific binding of [99mTc]Tc(CO)3-Ec1-LoPE (Fig. 3). Cell-associated activity was higher for PC-3 cells compared to DU145 cells.

Cellular processing

The binding of [99mTc]Tc-labeled Ec1-LoPE to both PC-3 and DU145 cells was rapid, and cell-associated activity was increased with incubation time. The internalized fraction reached 25±2% in the PC-3 cells and 30±0% in the DU145 cells after 6 h of incubation (Fig. 4).

Binding kinetics

The binding of [99mTc]Tc(CO)3-Ec1-LoPE to living DU145 cells was characterized by rapid association, while the dissociation included a rapid and a slow phase (Fig. 5). The Interaction Map analysis indicated the presence of two interactions, one with affinity in the low nanomolar range (KD1=2.1±0.3 nM) and another one approximately 20 times weaker (KD2=45±21 nM) (Table II).

Table II.

Equilibrium dissociation constants (KD) for the interaction between [99mTc]Tc-labeled Ec1-LoPE and EpCAM-expressing DU145 cells.

Table II.

Equilibrium dissociation constants (KD) for the interaction between [99mTc]Tc-labeled Ec1-LoPE and EpCAM-expressing DU145 cells.

KD1 KD2
[99mTc]Tc(CO)3-2.1±0.3 nM45±21 nM
Ec1-LoPE

[i] Analysis was performed in duplicate.

Cytotoxicity

The results of cytotoxicity of Ec1-LoPE on EpCAM-expressing PC-3 and DU145 cells are shown in Fig. 6. Ec1-LoPE demonstrated a dose-dependent cytotoxic effect with subnanomolar IC50 values, 36 pM for PC-3 cells and 56 pM for DU145 cells. Pre-incubation of cells with a large excess of DARPin Ec1 before the addition of Ec1-LoPE significantly (P<0.0001) reduced its cytotoxic action (Fig. 7), which suggested that the cytotoxic action of Ec1-LoPE was EpCAM-dependent.

Discussion

Treatment of metastasized prostate cancer (PC) with androgen deprivation therapy (ADT) provides an effective initial response in the majority of patients; however, it is not curative and tumors often develop resistance over the course of treatment. In the next phase of cancer progression into metastatic castration-resistant type, chemotherapy with docetaxel is added to ADT to increase the therapeutic efficacy (40). While providing a cytotoxic effect, conventional chemotherapeutic drugs do not discriminate between normal and cancer cells resulting in numerous side effects due to cumulative toxicity to normal organs.

Targeted therapeutic agents offer the advantage of reducing cytotoxic side effects on normal cells by directing the payload selectively to cancer cells. The most investigated targeted cytotoxic agents are antibody-drug conjugates, with several of them being approved for clinical use (41). In comparison with monoclonal antibodies (mAbs), small engineered scaffold proteins, such as designed ankyrin repeat proteins (DARPins), might be more effective to overcome biological barriers and deliver a cytotoxic payload to solid tumors, as well as to provide more even distribution in tumors (24). Due to faster clearance from blood and reduced exposure to normal tissues, a larger therapeutic window may also be achieved. Fusion of a DARPin to a bacterial toxin by genetic engineering enables recombinant production of a therapeutic agent as a single protein, removing the need for additional conjugation and purification steps (34). Genetic engineering provides an opportunity for facile optimization of molecular design (e.g., addition of targeting modules or linkers) and biodistribution characteristics.

Examples of scaffold proteins carrying bacterial toxins as a payload include affibody molecules (42), ABD-derived affinity proteins (ADAPTs) (43) and DARPins (44) targeting human epidermal growth factor receptor 2 (HER2). A number of agents have been developed using DARPins targeting EpCAM (Ec1 or Ec4) carrying Pseudomonas aeruginosa exotoxin A (ETA”, PE40), which have shown potent antitumor activity in xenograft models of colon cancer, small cell lung carcinoma (45) and breast cancer (46). To address the problem of immunogenic response to the bacterial toxin, B-cell recognition epitopes on the PE toxin were removed (36,47). The new toxin variant termed LoPE was fused to HER2-targeting DARPin 9_29 and showed lower non-specific toxicity and lower immunogenicity in vivo than DARPin 9_29-PE40, while preserving a potent cytotoxic action on ovarian cancer xenografts in mice (37). We recently demonstrated that Ec1-LoPE fusion targeting EpCAM caused a significant inhibition of tumor growth, which was additionally potentiated by a combination with HER2-targeting treatment in breast cancer (48) and ovarian cancer (38) models in mice. Repeated administration of Ec1-LoPE was well-tolerated with no observable toxicities or weight loss in mice.

Contrary to the IgG scaffold, the use of small engineered scaffold proteins for the development of targeted anticancer therapeutics is very recent. Therefore, more studies are necessary to improve the understanding of their therapeutic effect in different cancer types. In this study, we used DARPin Ec1 as an EpCAM-targeting module with a molecular weight of approximately 8-fold smaller than an mAb. The addition of LoPE toxin provided Ec1-LoPE fusion protein (43 kDa), which is smaller than a Fab fragment. Overexpression of EpCAM in a large fraction of prostate cancers and in metastatic lesions makes it a possible target for therapy of disseminated PC. This is further supported by a correlation between EpCAM overexpression and increased risk of PC recurrence (10,11).

To obtain quantitative information concerning the functional characteristics of Ec1-LoPE in PC cell lines, we applied site-specific radiolabeling approach using tricarbonyl technetium-99m as a label. Tricarbonyl technetium-99m was attached to the (His)6-tag at the C-terminus of Ec1-LoPE to form a residualizing label, which stays inside the cells after internalization and lysosomal degradation of the protein. This labeling method should have a minimal impact on the binding properties of Ec1-LoPE, since the label and the binding site are spatially separated. Radiolabeling results were in agreement with the previously reported data (38) and the stability of the radiolabel was confirmed (Table I).

The binding of [99mTc]Tc(CO)3-Ec1-LoPE to EpCAM-expressing PC-3 and DU145 cells was specific as demonstrated by the saturation experiment. The level of cell-associated activity per cell number was higher for PC-3 cells than for DU145 cells. Massoner et al reported that PC-3 and DU145 cells have a similar level of EpCAM expression at both the mRNA and protein levels, while PC-3 cells were found to have a higher degree of EpCAM-positive cells than DU145 by flow cytometry (7), which might be an explanation for the differences in the level of the cell-associated activity.

Previous studies have shown that the binding of Ec1 to EpCAM-expressing cells triggers receptor-mediated endocytosis and enables intracellular delivery of cytotoxic agents (35,48). Cellular processing experiment showed that approximately a quarter of cell-bound activity (25±2%) of [99mTc]Tc(CO)3-Ec1-LoPE was internalized by 6 h in PC-3 cells and 30±0% in DU145 cells, which suggests a sufficiently high internalization rate. These results are in line with a previous study in ovarian carcinoma cell lines, where internalization of [99mTc]Tc(CO)3-Ec1-LoPE was also rapid, with 31±0% in OVCAR3 cells and 38±3% in SKOV3 cells by 6 h (38).

Analysis of [99mTc]Tc(CO)3-Ec1-LoPE binding kinetics to DU145 cells showed the presence of two interactions, a stronger (KD1=2.1±0.3 nM) and a weaker one (KD2=45±21 nM), both being in the nanomolar range. A similar pattern of two interactions was observed for [99mTc]Tc(CO)3-Ec1-LoPE binding to SKOV3 and OVCAR3 cells, with the first equilibrium dissociation constant (KD1) being in the subnanomolar range. Comparing these results to DARPin Ec1 alone having KD of approximately 0.3 nM to DU145 cells (average value for different radiolabel types) (32), it can be concluded that fusion of Ec1 with LoPE resulted in a slight decrease in affinity, which might be expected due to the addition of a bulky toxin moiety to the targeting Ec1 module.

Ec1-LoPE demonstrated a potent cytotoxic effect with IC50 values in the low picomolar range in both PC-3 and DU145 cells. High efficiency of cell growth inhibition might be due to a combination of rapid internalization, efficient delivery and high sensitivity to LoPE toxin action. In our recent study, Ec1-LoPE efficiently inhibited tumor growth of SKOV3 ×enografts in mice, while having an IC50 value of 0.53 µM in SKOV3 cells in the cytotoxicity assay (38). It must be noted that SKOV3 cells were shown to be more resistant to the cytotoxic action of drug (49) and toxin conjugates (50) based on affibody molecules and ADAPTs (43,51) in comparison to other cell lines. Therefore, picomolar IC50 values of Ec1-LoPE in both PC-3 and DU145 cells suggest its potential for a therapeutic effect in PC models in vivo.

As the efficacy of targeted therapy depends on the presence of a sufficient amount of molecular target in tumors, it is important to select the patients with a high level of target expression. For the EpCAM-targeting mAb adecatumumab the probability of tumor progression was significantly lower in breast cancer patients who had high EpCAM expression and were treated with a high dose (14,52). High expression of EpCAM was also found to be a precondition for response of PC to adecatumumab (15). In addition to a conventional biopsy-based approach, evaluation of target expression could be performed by positron emission tomography (PET) or single photon emission tomography (SPECT) imaging. Radionuclide molecular imaging is a non-invasive method that provides whole-body information about target expression in real time and can be performed repeatedly to monitor changes in expression or receptor occupancy. The use of a pair of targeting agents, one for diagnostic imaging and another one for targeted therapy, forms a theranostic approach to patient treatment, where imaging is used for stratification of patients for targeted therapy. We recently reported on the preclinical development of Ec1-based radionuclide imaging probes, which efficiently visualize EpCAM expression in PC xenografts (32). Such probes may be used as companion diagnostics in the treatment of PC xenografts with Ec1-LoPE enabling selection of patients that would most likely benefit from such treatment.

In conclusion, DARPin Ec1 is an efficient targeting domain for LoPE toxin. Ec1-LoPE showed EpCAM-specific binding to EpCAM-expressing PC-3 and DU145 PC cells. Rapid internalization mediated potent cytotoxic effect in both studied cell lines. Taken together, these data support the further evaluation of Ec1-LoPE in a therapeutic setting in PC models in vivo.

Acknowledgements

The authors would like to thank Mr Haozhong Ding and Professor Torbjörn Gräslund for performing the LC-MS analysis of Ec1-LoPE. The authors would like to thank Ms. Kamila Seytova for proofreading the manuscript.

Funding

This research was funded by grants from the Swedish Prostate Cancer Federation (Prostatacancerförbundet) and the Swedish Cancer Society (Cancerfonden, 20 0181 P) to AV, by RFBR project 18-29-08030 in the part of Ec1-LoPE expression and purification.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

TX and YL performed the experiments and analyzed the data. AS and EK performed the production and purification of proteins. SMD participated in the molecular design, supervised the production, purification and characterization of protein, and coordinated the work. VT participated in the study design, labeling chemistry development, data treatment and interpretation, and coordinated the work. AV obtained funding, participated in the study design, labeling chemistry development, data treatment and interpretation, coordinated the work. TX and AV wrote the first version of the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work and all data are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Authors' information

ORCID nos.: TX 0000-0002-1826-4093; YL 0000-0001-5871-5779; AS 0000-0003-3425-0828; EK 0000-0002-4187-376X; SMD 0000-0002-3952-0631; VT 0000-0002-6122-1734; AV 0000-0002-4778-3909.

Glossary

Abbreviations

Abbreviations:

DARPin

designed ankyrin repeat protein

EpCAM

epithelial cell adhesion molecule

PBS

phosphate-buffered saline

KD

equilibrium dissociation constant

References

1 

Battaglia A, De Meerleer G, Tosco L, Moris L, Van den Broeck T, Devos G, Everaerts W and Joniau S: Novel insights into the management of oligometastatic prostate cancer: A comprehensive review. Eur Urol Oncol. 2:174–188. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Jacob A, Raj R, Allison DB and Myint ZW: Androgen receptor signaling in prostate cancer and therapeutic strategies. Cancers (Basel). 13:54172021. View Article : Google Scholar : PubMed/NCBI

3 

Sadaghiani MS, Sheikhbahaei S, Werner RA, Pienta KJ, Pomper MG, Solnes LB, Gorin MA, Wang NY and Rowe SP: A systematic review and meta-analysis of the effectiveness and toxicities of lutetium-177-labeled prostate-specific membrane antigen-targeted radioligand therapy in metastatic castration-resistant prostate cancer. Eur Urol. 80:82–94. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Thang SP, Violet J, Sandhu S, Iravani A, Akhurst T, Kong G, Ravi Kumar A, Murphy DG, Williams SG, Hicks RJ and Hofman MS: Poor outcomes for patients with metastatic castration-resistant prostate cancer with low prostate-specific membrane antigen (PSMA) expression deemed ineligible for 177Lu-labelled PSMA radioligand therapy. Eur Urol Oncol. 2:670–676. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Rosar F, Hau F, Bartholomä M, Maus S, Stemler T, Linxweiler J, Ezziddin S and Khreish F: Molecular imaging and biochemical response assessment after a single cycle of [225Ac]Ac-PSMA-617/[177Lu]Lu-PSMA-617 tandem therapy in mCRPC patients who have progressed on [177Lu]Lu-PSMA-617 monotherapy. Theranostics. 11:4050–4060. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Munz M, Baeuerle PA and Gires O: The emerging role of EpCAM in cancer and stem cell signaling. Cancer Res. 69:5627–5629. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Massoner P, Thomm T, Mack B, Untergasser G, Martowicz A, Bobowski K, Klocker H, Gires O and Puhr M: EpCAM is overexpressed in local and metastatic prostate cancer, suppressed by chemotherapy and modulated by MET-associated miRNA-200c/205. Br J Cancer. 111:955–964. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Poczatek RB, Myers RB, Manne U, Oelschlager DK, Weiss HL, Bostwick DG and Grizzle WE: Ep-Cam levels in prostatic adenocarcinoma and prostatic intraepithelial neoplasia. J Urol. 162:1462–1466. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Went PT, Lugli A, Meier S, Bundi M, Mirlacher M, Sauter G and Dirnhofer S: Frequent EpCam protein expression in human carcinomas. Hum Pathol. 35:122–128. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Benko G, Spajić B, Krušlin B and Tomas D: Impact of the EpCAM expression on biochemical recurrence-free survival in clinically localized prostate cancer. Urol Oncol. 31:468–474. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Hu Y, Wu Q, Gao J, Zhang Y and Wang Y: A meta-analysis and the cancer genome atlas data of prostate cancer risk and prognosis using epithelial cell adhesion molecule (EpCAM) expression. BMC Urol. 19:672019. View Article : Google Scholar : PubMed/NCBI

12 

Ni J, Cozzi P, Hao J, Beretov J, Chang L, Duan W, Shigdar S, Delprado W, Graham P, Bucci J, et al: Epithelial cell adhesion molecule (EpCAM) is associated with prostate cancer metastasis and chemo/radioresistance via the PI3K/Akt/mTOR signaling pathway. Int J Biochem Cell Biol. 45:2736–2748. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Simon M, Stefan N, Plückthun A and Zangemeister-Wittke U: Epithelial cell adhesion molecule-targeted drug delivery for cancer therapy. Expert Opin Drug Deliv. 10:451–468. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Schmidt M, Scheulen ME, Dittrich C, Obrist P, Marschner N, Dirix L, Schmidt M, Rüttinger D, Schuler M, Reinhardt C and Awada A: An open-label, randomized phase II study of adecatumumab, a fully human anti-EpCAM antibody, as monotherapy in patients with metastatic breast cancer. Ann Oncol. 21:275–282. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Marschner N, Rüttinger D, Zugmaier G, Nemere G, Lehmann J, Obrist P, Baeuerle PA, Wolf A, Schmidt M, Abrahamsson PA, et al: Phase II study of the human anti-epithelial cell adhesion molecule antibody adecatumumab in prostate cancer patients with increasing serum levels of prostate-specific antigen after radical prostatectomy. Urol Int. 85:386–395. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Seimetz D, Lindhofer H and Bokemeyer C: Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM × anti-CD3) as a targeted cancer immunotherapy. Cancer Treat Rev. 36:458–467. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Macdonald J, Henri J, Roy K, Hays E, Bauer M, Veedu RN, Pouliot N and Shigdar S: EpCAM Immunotherapy versus specific targeted delivery of drugs. Cancers (Basel). 10:192018. View Article : Google Scholar : PubMed/NCBI

18 

Di Paolo C, Willuda J, Kubetzko S, Lauffer I, Tschudi D, Waibel R, Plückthun A, Stahel RA and Zangemeister-Wittke U: A recombinant immunotoxin derived from a humanized epithelial cell adhesion molecule-specific single-chain antibody fragment has potent and selective antitumor activity. Clin Cancer Res. 9:2837–2848. 2003.PubMed/NCBI

19 

MacDonald GC, Rasamoelisolo M, Entwistle J, Cuthbert W, Kowalski M, Spearman MA and Glover N: A phase I clinical study of intratumorally administered VB4-845, an anti-epithelial cell adhesion molecule recombinant fusion protein, in patients with squamous cell carcinoma of the head and neck. Med Oncol. 26:257–264. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Andersson Y, Engebraaten O, Juell S, Aamdal S, Brunsvig P, Fodstad Ø and Dueland S: Phase I trial of EpCAM-targeting immunotoxin MOC31PE, alone and in combination with cyclosporin. Br J Cancer. 113:1548–1555. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Frøysnes IS, Andersson Y, Larsen SG, Davidson B, Øien JT, Olsen KH, Giercksky KE, Julsrud L, Fodstad Ø, Dueland S and Flatmark K: Novel treatment with intraperitoneal MOC31PE immunotoxin in colorectal peritoneal metastasis: results from the ImmunoPeCa phase 1 trial. Ann Surg Oncol. 24:1916–1922. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Cizeau J, Grenkow DM, Brown JG, Entwistle J and MacDonald GC: Engineering and biological characterization of VB6-845, an anti-EpCAM immunotoxin containing a T-cell epitope-depleted variant of the plant toxin bouganin. J Immunother. 32:574–584. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Plückthun A: Designed ankyrin repeat proteins (DARPins): Binding proteins for research, diagnostics, and therapy. Annu Rev Pharmacol Toxicol. 55:489–511. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Thurber GM, Schmidt MM and Wittrup KD: Antibody tumor penetration: Transport opposed by systemic and antigen-mediated clearance. Adv Drug Deliv Rev. 60:1421–1434. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Goldstein R, Sosabowski J, Livanos M, Leyton J, Vigor K, Bhavsar G, Nagy-Davidescu G, Rashid M, Miranda E, Yeung J, et al: Development of the designed ankyrin repeat protein (DARPin) G3 for HER2 molecular imaging. Eur J Nucl Med Mol Imaging. 42:288–301. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Deyev S, Vorobyeva A, Schulga A, Proshkina G, Güler R, Löfblom J, Mitran B, Garousi J, Altai M, Buijs J, et al: Comparative evaluation of two DARPin variants: Effect of affinity, size, and label on tumor targeting properties. Mol Pharm. 16:995–1008. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Vorobyeva A, Sсhulga A, Konovalova E, Güler R, Mitran B, Garousi J, Rinne S, Löfblom J, Orlova A, Deyev S and Tolmachev V: Comparison of tumor-targeting properties of directly and indirectly radioiodinated designed ankyrin repeat protein (DARPin) G3 variants for molecular imaging of HER2. Int J Oncol. 54:1209–1220. 2019.PubMed/NCBI

28 

Vorobyeva A, Schulga A, Rinne SS, Günther T, Orlova A, Deyev S and Tolmachev V: Indirect radioiodination of DARPin G3 using N-succinimidyl-para-iodobenzoate improves the contrast of HER2 molecular imaging. Int J Mol Sci. 20:30472019. View Article : Google Scholar : PubMed/NCBI

29 

Deyev SM, Vorobyeva A, Schulga A, Abouzayed A, Günther T, Garousi J, Konovalova E, Ding H, Gräslund T, Orlova A and Tolmachev V: Effect of a radiolabel biochemical nature on tumor-targeting properties of EpCAM-binding engineered scaffold protein DARPin Ec1. Int J Biol Macromol. 145:216–225. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Vorobyeva A, Konovalova E, Xu T, Schulga A, Altai M, Garousi J, Rinne SS, Orlova A, Tolmachev V and Deyev S: Feasibility of imaging EpCAM expression in ovarian cancer using radiolabeled DARPin Ec1. Int J Mol Sci. 21:33102020. View Article : Google Scholar : PubMed/NCBI

31 

Vorobyeva A, Bezverkhniaia E, Konovalova E, Schulga A, Garousi J, Vorontsova O, Abouzayed A, Orlova A, Deyev S and Tolmachev V: Radionuclide molecular imaging of EpCAM expression in triple-negative breast cancer using the scaffold protein DARPin Ec1. Molecules. 25:47192020. View Article : Google Scholar : PubMed/NCBI

32 

Deyev SM, Xu T, Liu Y, Schulga A, Konovalova E, Garousi J, Rinne SS, Larkina M, Ding H, Gräslund T, et al: Influence of the position and composition of radiometals and radioiodine labels on imaging of Epcam expression in prostate cancer model using the DARPin Ec1. Cancers (Basel). 13:35892021. View Article : Google Scholar : PubMed/NCBI

33 

Bragina O, Chernov V, Schulga A, Konovalova E, Garbukov E, Vorobyeva A, Orlova A, Tashireva L, Sorensen J, Zelchan R, et al: Phase I trial of 99mTc-(HE)3-G3, a DARPin-based probe for imaging of HER2 expression in breast cancer. J Nucl Med. Aug 12–2021.(Epub ahead of print). View Article : Google Scholar

34 

Shilova O, Shramova E, Proshkina G and Deyev S: Natural and designed toxins for precise therapy: Modern approaches in experimental oncology. Int J Mol Sci. 22:49752021. View Article : Google Scholar : PubMed/NCBI

35 

Stefan N, Martin-Killias P, Wyss-Stoeckle S, Honegger A, Zangemeister-Wittke U and Plückthun A: DARPins recognizing the tumor-associated antigen EpCAM selected by phage and ribosome display and engineered for multivalency. J Mol Biol. 413:826–843. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Liu W, Onda M, Lee B, Kreitman RJ, Hassan R, Xiang L and Pastan I: Recombinant immunotoxin engineered for low immunogenicity and antigenicity by identifying and silencing human B-cell epitopes. Proc Natl Acad Sci USA. 109:11782–11787. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Sokolova EA, Shilova ON, Kiseleva DV, Schulga AA, Balalaeva IV and Deyev SM: HER2-specific targeted toxin DARPin-LoPE: Immunogenicity and antitumor effect on intraperitoneal ovarian cancer xenograft model. Int J Mol Sci. 20:23992019. View Article : Google Scholar : PubMed/NCBI

38 

Xu T, Vorobyeva A, Schulga A, Konovalova E, Vorontsova O, Ding H, Gräslund T, Tashireva LA, Orlova A, Tolmachev V and Deyev SM: Imaging-guided therapy simultaneously targeting HER2 and EpCAM with trastuzumab and EpCAM-directed toxin provides additive effect in ovarian cancer model. Cancers (Basel). 13:39392021. View Article : Google Scholar : PubMed/NCBI

39 

Wållberg H and Orlova A: Slow internalization of anti-HER2 synthetic affibody monomer 111In-DOTA-ZHER2:342-pep2: Implications for development of labeled tracers. Cancer Biother Radiopharm. 23:435–442. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Cornford P, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, Fanti S, Fossati N, Gandaglia G, Gillessen S, et al: EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer. Part II-2020 update: Treatment of relapsing and metastatic prostate cancer. Eur Urol. 79:263–282. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Drago JZ, Modi S and Chandarlapaty S: Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat Rev Clin Oncol. 18:327–344. 2021. View Article : Google Scholar : PubMed/NCBI

42 

Altai M, Liu H, Orlova A, Tolmachev V and Gräslund T: Influence of molecular design on biodistribution and targeting properties of an Affibody-fused HER2-recognising anticancer toxin. Int J Oncol. 49:1185–1194. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Ding H, Altai M, Yin W, Lindbo S, Liu H, Garousi J, Xu T, Orlova A, Tolmachev V, Hober S and Gräslund T: HER2-specific Pseudomonas exotoxin A PE25 based fusions: Influence of targeting domain on target binding, toxicity, and in vivo biodistribution. Pharmaceutics. 12:3912020. View Article : Google Scholar : PubMed/NCBI

44 

Sokolova E, Proshkina G, Kutova O, Shilova O, Ryabova A, Schulga A, Stremovskiy O, Zdobnova T, Balalaeva I and Deyev S: Recombinant targeted toxin based on HER2-specific DARPin possesses a strong selective cytotoxic effect in vitro and a potent antitumor activity in vivo. J Control Release. 233:48–56. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Martin-Killias P, Stefan N, Rothschild S, Plückthun A and Zangemeister-Wittke U: A novel fusion toxin derived from an EpCAM-specific designed ankyrin repeat protein has potent antitumor activity. Clin Cancer Res. 17:100–110. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Simon M, Stefan N, Borsig L, Plückthun A and Zangemeister-Wittke U: Increasing the antitumor effect of an EpCAM-targeting fusion toxin by facile click PEGylation. Mol Cancer Ther. 13:375–385. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Proshkina GM, Kiseleva DV, Shilova O, Ryabova AV, Shramova EI, Stremovskiy OA and Deyev SM: Bifunctional toxin DARP-LoPE based on the HER2-specific innovative module of a non-immunoglobulin scaffold as a promising agent for theranostics. Mol Biol. 51:865–873. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Shramova E, Proshkina G, Shipunova V, Ryabova A, Kamyshinsky R, Konevega A, Schulga A, Konovalova E, Telegin G and Deyev S: Dual targeting of cancer cells with DARPin-based toxins for overcoming tumor escape. Cancers (Basel). 12:30142020. View Article : Google Scholar : PubMed/NCBI

49 

Altai M, Liu H, Ding H, Mitran B, Edqvist PH, Tolmachev V, Orlova A and Gräslund T: Affibody-derived drug conjugates: Potent cytotoxic molecules for treatment of HER2 over-expressing tumors. J Control Release. 288:84–95. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Liu H, Seijsing J, Frejd FY, Tolmachev V and Gräslund T: Target-specific cytotoxic effects on HER2-expressing cells by the tripartite fusion toxin ZHER2:2891-ABD-PE38X8, including a targeting affibody molecule and a half-life extension domain. Int J Oncol. 47:601–609. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Garousi J, Ding H, von Witting E, Xu T, Vorobyeva A, Oroujeni M, Orlova A, Hober S, Gräslund T and Tolmachev V: Targeting HER2 expressing tumors with a potent drug conjugate based on an albumin binding domain-derived affinity protein. Pharmaceutics. 13:18472021. View Article : Google Scholar : PubMed/NCBI

52 

Schmidt M, Rüttinger D, Sebastian M, Hanusch CA, Marschner N, Baeuerle PA, Wolf A, Göppel G, Oruzio D, Schlimok G, et al: Phase IB study of the EpCAM antibody adecatumumab combined with docetaxel in patients with EpCAM-positive relapsed or refractory advanced-stage breast cancer. Ann Oncol. 23:2306–2313. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2022
Volume 47 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu T, Liu Y, Schulga A, Konovalova E, Deyev SM, Tolmachev V and Vorobyeva A: Epithelial cell adhesion molecule‑targeting designed ankyrin repeat protein‑toxin fusion Ec1‑LoPE exhibits potent cytotoxic action in prostate cancer cells. Oncol Rep 47: 94, 2022
APA
Xu, T., Liu, Y., Schulga, A., Konovalova, E., Deyev, S.M., Tolmachev, V., & Vorobyeva, A. (2022). Epithelial cell adhesion molecule‑targeting designed ankyrin repeat protein‑toxin fusion Ec1‑LoPE exhibits potent cytotoxic action in prostate cancer cells. Oncology Reports, 47, 94. https://doi.org/10.3892/or.2022.8305
MLA
Xu, T., Liu, Y., Schulga, A., Konovalova, E., Deyev, S. M., Tolmachev, V., Vorobyeva, A."Epithelial cell adhesion molecule‑targeting designed ankyrin repeat protein‑toxin fusion Ec1‑LoPE exhibits potent cytotoxic action in prostate cancer cells". Oncology Reports 47.5 (2022): 94.
Chicago
Xu, T., Liu, Y., Schulga, A., Konovalova, E., Deyev, S. M., Tolmachev, V., Vorobyeva, A."Epithelial cell adhesion molecule‑targeting designed ankyrin repeat protein‑toxin fusion Ec1‑LoPE exhibits potent cytotoxic action in prostate cancer cells". Oncology Reports 47, no. 5 (2022): 94. https://doi.org/10.3892/or.2022.8305