Search for


TEXT SIZE

search for



CrossRef (0)
PD-L1 Aptamer-functionalized Liposome Containing SAHA for Anti-lung Cancer Immunotherapy
Biomed Sci Letters 2024;30:37-48
Published online June 30, 2024;  https://doi.org/10.15616/BSL.2024.30.2.37
© 2024 The Korean Society For Biomedical Laboratory Sciences.

Si-Yeon Ryu* , Se-Yun Hong* and Keun-Sik Kim†,**

Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
Correspondence to: Keun-Sik Kim. Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea.
Tel: +82-42-600-8434, Fax: +82-42-600-8408, e-mail: kskim11@konyang.ac.kr

*Graduate student, **Professor.
Received March 11, 2024; Revised May 8, 2024; Accepted June 5, 2024.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
 Abstract
Liposomes are one of the most actively studied and promising drug delivery systems for the treatment of various diseases. In this study, an aptamer-conjugated liposome called "aptamosome" was used, in which an anti-PD-L1 aptamer targeting cancer cells was conjugated to the liposome. These aptamosomes showed remarkable cellular uptake and efficient delivery to Lewis lung carcinoma 2 (LL/2) cancer cells. In addition, suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor (HDACi), was delivered through this aptamer to induce a strong anticancer immunotherapeutic effect. The results of this study showed that when LL/2 cells were treated with SAHA-entrapped aptamosome [SAHA] and liposome [SAHA] and free SAHA, aptamosome [SAHA] improved cell death compared with that of liposomes [SAHA] or free SAHA, and it has demonstrated anticancer efficacy. Moreover, aptamosome [SAHA] induce the secretion of chemokines that promote the migration of activated T cells into tumor tissues. Finally, in vivo experiments showed that aptamosome [SAHA] significantly inhibited the growth rate of LL/2 tumors. Therefore, liposomes combined with an anti-PD-L1 aptamer for efficient SAHA delivery are suggested as an excellent model for drug delivery systems suitable for targeting cancer cells.
Keywords : Aptamer, Drug delivery, HDACi, Liposome, PD-L1, SAHA
INTRODUCTION

Cancer is a major disease that poses challenges for the development of therapeutics. Immunotherapy offers a unique cancer elimination strategy that identifies immune evasion and induces a response (Li et al., 2018). Over the past few decades, cancer immunotherapies have evolved by utilizing T cells, cytokines, vaccines, antibodies, and immune check-point inhibitors (Riley et al., 2019). Blocking programmed cell death protein 1 (PD-1) on T cells enhances immune activation within the tumor microenvironment (TME). Programmed death-ligand 1 (PD-L1), a membrane protein on tumor cells, interacts with PD-1, suppresses T-cell activity, and causes immune evasion (Granier et al., 2017; Chen et al., 2021; Sanmamed and Chen, 2018). Monoclonal antibody-based treatments targeting PD-1 or PD-L1 have proven effective in clinical trials; however, antibody-based immune checkpoint blockade (ICB) does not extend the anticancer response and poses a risk of cancer recurrence (Martins et al., 2019). Additionally, these treatments are difficult to implement and require high costs and time. Thus, combining immune-activating drugs or alternative ligands is crucial to restoring suppressed anticancer responses and addressing issues related to therapeutic production (Kennedy and Salama, 2020; Lv et al., 2022).

Histone deacetylases (HDACs) cause dysregulation and abnormal histone mutations in various diseases, resulting in cancer development and an unfavorable prognosis. Histone deacetylase inhibitors (HDACi) restore cellular acetylation homeostasis and related gene expression and improve tumor immunogenicity by increasing the expression of major histocompatibility complex (MHC) molecules (Conte et al., 2018; West and Johnstone, 2014). Various chemokines, including CXC motif chemokine ligand 10 (CXCL10) secreted by several HDACis, induce antitumor immune responses (Zheng et al., 2016; Terranova-Barberio et al., 2017). However, suberoylanilide hydroxamic acid (SAHA), which was approved by the Food and Drug Administration (FDA) in 2006, is an HDACi that is used as an anticancer agent but has low permeability and bioavailability; therefore, a targeted delivery system is needed to overcome these problems (Mann et al., 2007; Le et al., 2021).

Liposomes, which are used as drug delivery carriers for diverse disease treatments, encapsulate drugs and exhibit amphiphilic characteristics that enable the transport of both hydrophilic and hydrophobic molecules (Large et al., 2021). With excellent biocompatibility, efficient cellular uptake, and the ability to escape endosomes, liposomes have enhanced therapeutic effects in cancer treatment (Akbarzadeh et al., 2013). While clinical trials often use liposomal delivery systems based on enhanced permeation and retention (EPR), enhancing in vivo tumor drug accumulation requires the conjugation of specific ligands such as antibodies, aptamers, or peptides to target cancer cells (Peer et al., 2020; Xu et al., 2013).

Aptamers, which are short single-stranded DNA or RNA oligonucleotides that can bind to target molecules, are promising ligands for targeted drug delivery and can accurately and efficiently deliver drugs through high affinity and specific binding to receptors or antigens on tumor (Liu et al., 2014; Alshaer et al., 2018). The disadvantages of currently used antibody treatments are that they are expensive and may cause inflammatory side effects due to non-specific false positive binding. However, when aptamosomes using aptamers are used for treatment, side effects can be minimized due to the structural stability, low toxicity, and low immunogenicity of the aptamers (Lv et al., 2022; Agnello et al., 2021). The advantage of these aptamer-linked liposomes is that they can target drugs more effectively, increasing the possibility of selective drug delivery to cancer cells (Gao et al., 2021).

In this study, anti-PD-L1 aptamer-conjugated liposomes were used for the targeted delivery of the anticancer agent SAHA to PD-L1-highly expressed cancer cells. First, we formulated a liposome [SAHA] containing SAHA in a neutral liposome containing SAHA based on POPC (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine). Subsequently, we evaluated aptamosome [SAHA] designed for specific cancer cell targeting by combining liposome [SAHA] with an anti-PD-L1 DNA aptamer in comparison with liposome [SAHA] lacking the anti-PD-L1 aptamer. Furthermore, we evaluated the potential of aptamosome [SAHA] to induce an anticancer immune response, including cytotoxicity and secretion of T-cell-activating chemokines, both in vitro and in vivo.

MATERIALS AND METHODS

Materials

DSPE-PEG2000 (1,2-distearoyl-sn-glycero-3-phospho-ethanolamine-N[amino(polyethylene glycol)-2000]), DSPE-PEG2000-Maleimide (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[maleimide(polyethylene-glycol)-2000]), POPC (1-palmitoyl-2-oleoyl-glycero-3-phosphocholine), DOPE (1,2-dioleoyl-sn-glycero-3-phosphatidylethanolamine), Rho-DOPE (1,2-dioleoyl-sn-glycero-3-phosphoenthanol-amine), and cholesterol were purchased from Avanti Polar Lipid, Inc. (Alabaster, AL, USA). Liposome-sized extruders and polycarbonate membranes (100, 200, 400, and 800 nm) were purchased from Whatman (Maidstone, UK). Suberoylanilide hydroxamic acid (SAHA) was purchased from Sigma-Aldrich (St. Louis, MO, USA). Amicon tube (30 k) was purchased from Merck Millipore (Billerica, MA, USA). The anti-PD-L1 DNA aptamer (sequence: 5'-ACGGGCC-ACATCAACTCATTGATAGACAATGCGTCCACTGCC-CGTTTTTTTTTT-3') was synthesized and modified with FAM (6-fluorescein amidite) and thiol at the 5' and 3' ends, respectively (Bioneer, Daejeon, Korea).

Cell culture

LL/2 Lewis lung carcinoma and HepG2 hepatocellular carcinoma cells were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA). The cells were maintained in Dulbeccós Modified Eaglés Medium (DMEM) (Corning, NY, USA) supplemented with 10% heat-inactivated fetal bovine serum (FBS) (Gibco/Invitrogen, Carlsbad, CA, USA), and 1% penicillin/streptomycin (Hyclone, Logan, UT, USA) in an incubator in 5% CO2 at 37℃.

Preparation of liposomes containing SAHA

POPC, cholesterol, PEG-DSPE2000, and DOPE were mixed at a molar ratio of 4:1:0.4:1 in chloroform and methanol (2:1, v/v). The mixtures were evaporated using N2 gas to form a thin lipid film and desiccated using a vacuum pump for a minimum of 1 h to remove the residual organic solvent. The dried lipid film was hydrated in HEPES buffer (20 mM HEPES and 150 mM NaCl, pH 7.5) containing 500 μM SAHA. Then the hydrated solution was incubated for 2 h at 60℃ with stirring to form liposomes containing SAHA. To prepare small unilamellar vesicles, liposomes were sonicated for 3 min in a bath-type sonicator (Branson Inc., Danbury, USA). Finally, the liposome solution was extruded through 800-, 400-, 200-, and 100-nm polycarbonate membranes to adjust the liposome size.

Post-insertion of aptamer-conjugated micelles

To prepare the anti-PD-L1 DNA aptamer for conjugation with micelles, 1 nM anti-PD-L1 aptamers were treated with 10 μL of 100 mM TCEP (tris(2-carboxyethyl) phosphine) for 1 h at 25℃ and then mixed with 3 M NaOAc and 100% cold ethanol. Micelles were prepared using DSPE-PEG2000 /DSPE-PEG2000-maleimide (1:4 molar ratio). Afterward, DNA aptamers were conjugated to the maleimide groups of micelles by incubation for 2 h at 25℃, producing aptamer-conjugated micelles. After the incubation, anti-PD-L1 aptamer-conjugated micelles were mixed with liposomes containing SAHA and incubated for 1 h at 60℃. Finally, the cells were incubated for 1 h at 25℃ to complete the formation of anti-PD-L1 aptamer-conjugated liposomes containing SAHA (aptamosomes [SAHA]).

Cell viability assay

The cytotoxic effects of free drugs and formulations were evaluated using the MTT assay. LL/2 cells (1 × 104 cells/ well) were seeded in a 96-well plate and incubated overnight at 37℃. The cells were treated with various concentrations of free SAHA (0.39~50 μM) or different formulations (SAHA, liposome [SAHA], and aptamosome [SAHA]) in 100 μL medium and incubated for 24 and 48 h. After incubation, the cells were washed with DPBS and incubated with a fresh medium containing MTT reagent (5 mg/mL) for 3 h at 37℃. The media were discharged, and the cells were incubated with DMSO to solubilize the purple formazan crystals for 15 min at 37℃. Absorbance was measured at 570 nm using a VersaMax microplate reader (Molecular Devices, Sunnyvale, CA, USA). The cytotoxic effects of formulations: free SAHA, liposome [SAHA], and aptamosome [SAHA] containing the equivalent amount of SAHA (10 μM) were analyzed by the same method described above.

Apoptosis assay

LL/2 cells (4 × 104 cells/well) were seeded on a 24-well plate and grown overnight at 37℃. The cells were treated with SAHA (0, 2, 4, 6, 8, 10, and 12 μM) or various formulations (SAHA, liposome [SAHA], and aptamosome [SAHA]) in 500 μL medium and incubated for 24 and 48 h at 37℃. After incubation, the cells were harvested using trypsinization, and pellets were redispersed in 100 μL of the annexin-binding buffer. The cells were co-stained with a combo of 5 μL of Annexin-V/FITC and 1 μL of PI working solution (Invitrogen, Carlsbad, CA, USA) and incubated for 20 min at room temperature. The stained cells were analyzed using a NovoCyte Flow Cytometer (ACEA Bioscience, San Diego, CA, USA).

Enzyme-linked immunosorbent assay

LL/2 cells (4 × 104 cells/well) were seeded on a 24-well plate and grown overnight at 37℃. The cells were treated with various concentrations of SAHA (0, 1, 3, 10, 25, and 50 μM) or formulations (SAHA, liposome [SAHA], and aptamosome [SAHA]) in 500 μL medium and incubated for 48 h at 37℃. After incubation, cell lysates and supernatants were harvested from each well and assayed to determine the secretion levels of CXCL10 using a Mouse CXCL10 DuoSet ELISA kit (R&D Systems, Minneapolis, MN, USA) following the manufacturer's instructions. Absorbance was measured at 450 nm using a microplate reader.

Cell binding analysis

LL/2 cells and HepG2 cells (8 × 104 cells/well) were cultured on cover glass (Ф12 mm) placed in 24-well plates and grown overnight at 37℃. The cells were treated with various formulations: liposome [SAHA], aptamosome [SAHA] labeled with rhodamine, respectively; containing equivalent lipids (30 μg). After incubation times (10 min, 30 min, 6 h, and 12 h) at 37℃, the cells were washed three times with cold PBS and fixed with 100% chilled methanol. Nuclei were stained with the mounting medium with DAPI (Invitrogen, Carlsbad, CA, USA) overnight at 4℃. The images were captured using a fluorescence microscope. The fluorescence from the rhodamine-conjugated DOPE lipids was monitored using red fluorescence.

Animal model

For all experiments, eight-week-old male C57BL/6N mice were purchased from Koatech (Pyeongtaek, Korea). All the animal experiments were approved by the Institutional Animal Care and Use Committee (IACUC approval no. KU22232). The tumor-bearing models were established by subcutaneous administration of 100 μL of PBS containing LL/2 cells (7 × 105) into the flank of the mice. To establish lung experimental metastasis models, LL/2 cells (4 × 105) in 100 μL PBS were intravenously injected into the mice.

Antitumor efficiency in subcutaneous tumor model mice

The antitumor efficacies of SAHA, liposome [SAHA] and aptamosome [SAHA] were investigated in tumor-bearing mice. When the tumors reached a volume of approximately 100 mm3, the mice were randomly allocated to five groups based on their tumor volumes. All mice were treated by intravenous injection twice a week with different formulations (diluted in 200 μL saline): free SAHA, liposome [SAHA], aptamosome [SAHA] which contain an equivalent amount of SAHA (0.25 mg/kg) in each liposome formulation. Body weight and tumor volume were measured every two days over a period of 28 days. Tumor volume (mm3) = (length × width2)/2.

Hematoxylin and eosin (H&E) staining

Mice injected with different formulations were euthanized 28 days after inoculation for histological analysis. Tumors were harvested and fixed in a 10% formalin solution. The tissues were dehydrated with 70~100% ethanol and embedded in paraffin. The sections were deparaffinized and stained with hematoxylin and eosin. The stained slides were observed under a light microscope.

Statistical analysis

The experiment results are expressed as the means ± standard deviation (SD). Statistical analyses of in vitro experiments were performed using two-tailed Student's t-tests. Statistical analysis of in vivo experiments was performed using one-way ANOVA followed by GraphPad Prism 9 (GraphPad Software Inc, California, USA). *P<0.05, **P< 0.01 and ***P<0.001 were considered statistically significant.

RESULTS

Anticancer effects of SAHA in mouse lung carcinoma cells LL/2

To evaluate the anticancer effect of SAHA, we analyzed the cytotoxicity and apoptosis of LL/2 cells treated with SAHA. After cells were treated with doubly increasing concentrations of SAHA from 0.39 μM to 50 μM for 24 and 48 h, the cytotoxicity of LL/2 was measured using the MTT assay (Fig. 1A). As a result, cell death gradually increased with increasing concentration of SAHA, and the IC50 value of SAHA was measured to be 10 μM when cultured in LL/2 for 48 hours. Additionally, SAHA-induced apoptosis in LL/2 cells was measured using flow cytometry using Annexin V-FITC/PI staining (Fig. 1B and 1C). These results indicate that SAHA can be used as an anticancer agent by inducing cytotoxicity and apoptosis in lung cancer cells.

Fig. 1. Anticancer efficacy of SAHA against LL/2 mouse lung carcinoma cells. (A) LL/2 cells were treated with SAHA at 2-fold increasing concentrations from 0.3 μM to 50 μM for 24 and 48 h. Cell viability was measured via MTT assay at 570 nm and expressed as % relative to the negative control. (B and C) LL/2 cells were treated with the indicated concentration of SAHA for 24 and 48 h. Apoptosis was analyzed via flow cytometry using Annexin V-FITC/PI staining. The sum of early apoptotic cells and late apoptotic cells indicates total apoptosis. The data are represented as mean ± SD (n = 4).

Targeted cellular binding of anti-PD-L1 aptamer-conjugated aptamosome [SAHA]

Specific binding of the aptamosome [SAHA] to the aptamer was measured by identifying the lipid fluorescence of rhodamine. Liposome [SAHA] and aptamosome [SAHA] with rhodamine-labeled DOPE were treated with LL/2 and HepG2 cells for 10 and 30 min, respectively. After incubation, fluorescence images were captured with a fluorescence microscope (Fig. 2A), and the binding affinity was measured using flow cytometry (Fig. 2B). No noticeable uptake efficiency was observed of aptamosomes [SAHA] in PD-L1-negative control HepG2 cells. However, LL/2 cells expressing PD-L1 showed rhodamine fluorescence signals that were approximately 1.8-fold higher than those in the cells treated with liposomes [SAHA]. These results indicate that the aptamosome [SAHA], through the anti-PD-L1 DNA aptamer, can selectively target cancer cells and effectively deliver SAHA.

Fig. 2. Cell binding affinity of aptamosome[SAHA] by anti-PD-L1 aptamer. (A) LL/2 cells and HepG2 cells were incubated for 10 and 30 min with liposome [SAHA] and aptamosome [SAHA] at 37℃. The intercellular localization of the liposome was visualized through fluorescence microscopy with rhodamine attached to the lipids. The fluorescent images were captured at 40× magnification. DAPI (blue): nuclei, Rhodamine (red): liposomes. (B) In LL/2 cells, the binding affinities of liposome [SAHA] and aptamosome [SAHA] were analyzed using flow cytometry. Red: liposome [SAHA] 10 min, Green: liposome [SAHA] 30 min, Blue: aptamosome [SAHA] 10 min, Brown: aptamosome [SAHA] 30 min.

Anticancer efficacy of the aptamosome [SAHA] in LL/2 cells

The anticancer effect of the aptamosome [SAHA] on the mouse lung cancer cell line LL/2 was examined via the MTT assay (Fig. 3A). The cells were incubated for 48 h after treatment with SAHA, liposome [SAHA], and aptamosome [SAHA] containing an equivalent concentration of SAHA at 10 μM. The cell viability graph shows that cells treated with aptamosomes [SAHA] exhibited higher cytotoxicity than those treated with SAHA or liposomes [SAHA]. Additionally, apoptosis was measured using flow cytometry (Fig. 3B). Apoptosis levels showed the same trend as cytotoxicity levels (Fig. 3C). These results indicate that aptamosomes [SAHA] induce greater cytotoxicity in lung cancer cells than that in liposomes [SAHA] or free SAHA.

Fig. 3. Anticancer efficacy by aptamosome [SAHA] in LL/2 cells. (A) LL/2 cells were treated with different formulations for 48 h. The cell viability is evaluated via MTT assay and measured at 570 nm. The cell viability is indicated as % compared to an untreated control group. (B and C) LL/2 cells were treated with different formulations for 48 h. Apoptosis assays were performed with the annexin-V-FITC/PI flow cytometry analysis to measure total apoptosis levels. Data are represented as the mean ± SD for triplicate experiments. A; ***P<0.001 compared with liposome [SAHA] and aptamosome [SAHA]. C; **P<0.01 compared with liposome [SAHA] and aptamosome [SAHA]. A and C; *P<0.05 compared with SAHA and aptamosome [SAHA].

Secretion of chemokine CXCL10 by SAHA and aptamosome [SAHA] in LL/2 cells

To determine whether T cell-derived chemokine CXCL10 expression was induced by SAHA, it was measured using ELISA. LL/2 cells were treated with 1, 5, 10, 25, and 50 μM SAHA for 48 hours, and CXCL10 expression was confirmed to be increased in a dose-dependent manner in SAHA cells (Fig. 4A). Next, to confirm whether aptamosome [SAHA] induces immune activity, cells were treated with SAHA, liposome [SAHA], and aptamosome [SAHA] for 48 hours, respectively, and then the secretion of CXCL10 was examined. As a result, CXCL10 levels were higher in cells treated with aptamosomes [SAHA] than in cells treated with other agents (Fig. 4B). These results suggest that aptamosomes [SAHA] have a high potential to recruit cytotoxic T cells and induce immune activity.

Fig. 4. Secretion of chemokine CXCL10 by SAHA and aptamosome [SAHA] in LL/2 cells. (A) The Secretion level of CXCL10 was confirmed using the ELISA method in LL/2 cells treated with SAHA (0~50 μM) for 48 h. (B) LL/2 cells were treated with SAHA, liposome [SAHA], and aptamosome [SAHA] for 48 h, respectively, and the CXCL10 secretion level was evaluated using ELISA. The data are represented as the mean ± SD (n = 3). A; **P<0.01 compared with the control group. B; *P<0.05 compared with SAHA and aptamosome [SAHA], **P<0.01 compared with liposome [SAHA] and aptamosome [SAHA].

Tumor growth inhibition by aptamosome [SAHA] in LL/2 tumor-bearing mouse models

Based on these in vitro results, aptamosome [SAHA] was evaluated in a lung cancer xenograft model to demonstrate that aptamosome [SAHA] treatment induces immune responses and enhances anticancer activity. As shown in Fig. 5A, the lung cancer retention model was created by subcutaneously injecting LL/2 lung cancer cells into C57BL/6N mice. When the tumor volume reached an average of 60~ 80 mm3, various agents were administered intravenously twice a week. Each preparation contained the same amount of SAHA (0.25 mg/kg). To demonstrate the toxicity and tumor treatment effects of the different formulations, mouse body weight (Fig. 5B) and tumor volume (Fig. 5C) were measured for 28 days. As a result, Fig. 5B confirmed that the mouse body weight was maintained appropriately and that all preparations were non-toxic. Changes in tumor volume showed relative differences, with free SAHA (0.25 mg/kg) treatment not reducing tumor volume compared to the untreated control group. Liposome [SAHA] treatment demonstrated that liposomes could passively target tumors through the EPR effect and inhibit tumor growth better than free SAHA, but no significant reduction in tumor volume was observed. Aptamosome [SAHA] treatment suppressed tumor size compared to liposome [SAHA] treatment at day 28 due to active targeting by PD-L1 aptamer. This significant tumor growth inhibition trend indicates that SAHA can be delivered systemically to tumors in vivo through targeting effects using aptamers.

Fig. 5. In vivo tumor growth inhibition by aptamosome [SAHA] in LL/2 tumor-bearing models. (A) Experimental timeline of a designed antitumor study using LL/2 tumor-bearing mice. During the experimental period of 28 d, (B) the body weights and (C) the tumor volume was measured (n = 4). C; *P<0.05 compared with administration of liposome [SAHA] and aptamosome [SAHA].

Histopathological variation of tumors by aptamosome [SAHA] in LL/2 tumor-bearing mouse models

Apoptotic cells within a tumor can be identified by distinct features such as cell shrinkage, condensed nuclei, and hypereosinophilic cytoplasm. Accordingly, we inoculated the LL/2 tumor-bearing mouse model with various formulations for 28 days, and isolated tumor tissues were stained with H&E reagent for histological analysis (Fig. 6). In tumor tissues treated with various agents, progressive changes were observed that induce apoptosis in tumor cells. Tumors treated with free SAHA appeared similar to those treated with untreated controls. However, changes were observed in liposomes [SAHA] and aptamosomes [SAHA]. PD-L1 aptamer-binding aptamosomes [SAHA] showed a significant reduction in tumor cell numbers. These results indicate that aptamosome [SAHA] has antitumor efficacy in vivo.

Fig. 6. Histopathological variation of tumor tissues in LL/2 tumor-bearing models treated by aptamosome [SAHA]. Mouse tumor tissues treated with different formulations were stained with hematoxylin and eosin (H&E) staining and observed under a light microscope. The untreated group represented LL/2 tumor-bearing mice that were not treated with any drugs. The regions in the red boxes indicate images at 40× magnification. (Scale bar, 20X: 200 μm, 40X: 100 μm).
DISCUSSION

In recent years, the blockade of immune checkpoints has led to advances in cancer immunity therapeutics. Among ICB therapies, PD-1/PD-L1 blockade has been widely used for cancer treatment. Numerous methods, such as anti-PD-1 or anti-PD-L1 drugs and monoclonal antibodies, have been proposed to block PD-1/PD-L1 (Melosky et al., 2019; Li et al., 2021). Although these methods are less toxic than chemotherapy, negative effects related to immune activation such as diarrhea, fatigue, and rashes have been reported (Abdel-Rahman et al., 2016). Accordingly, developing more effective therapeutic approaches to avoid negative immune-related effects and increase the immune response in solid cancers is a challenge for cancer immunotherapy (Baxi et al., 2018). Therefore, ICB therapies may enhance anticancer effects when combined with other immune-activating agents.

In this study, SAHA, an HDAC inhibitor used as an anticancer drug, was selected and its immune activation ability was analyzed. First, to determine whether SAHA has anticancer and apoptotic effects in LL/2 cells, we performed cytotoxicity and apoptosis assays and confirmed that SAHA is effective (Fig. 1). Next, the immune activity of SAHA was demonstrated through the secretion of CXCL10, a chemokine that recruits T cells (Fig. 4A).

Accordingly, the main purpose of this study was to utilize the liposome delivery system to increase the efficacy of SAHA drug delivery. Liposome delivery systems have been considered an ideal delivery vehicle for clinical applications due to their excellent biocompatibility and loading capacity for hydrophobic and hydrophilic drugs. Liposomes can encapsulate drugs in lipid bilayers, and the EPR effect of liposomes gives the encapsulated drugs a long circulation time (Peer et al., 2020). Based on these advantages, it was expected that using liposomes to deliver SAHA would improve the drug delivery effect. Liposomes have excellent delivery capabilities, but the need for combination with antibodies, aptamers, etc. has been raised to complement affinity and specificity (Bagalkot et al., 2006). The primary objective of this study was to establish an approach for cancer immunotherapy by conjugating aptamers to liposomes and delivering SAHA. In this study, we designed POPC-based liposomes to encapsulate SAHA. Subsequently, PD-L1 aptamers were conjugated with liposomal carriers to specifically target cancer cells (aptamosomes). The binding affinity of the aptamosomes was evaluated in LL/2 cells in vitro and compared with that of liposomes lacking aptamers (Fig. 2).

In Fig. 3, the cancer cell inhibition effects of the SAHA-treated group and the aptamosome-treated group were similar, but aptamosomes with targeting ability had a higher cancer cell inhibition effect than non-targeting liposomes. In vitro results showed that SAHA, a free drug, was effective in suppressing cancer cells at the IC50 concentration, and Aptamosome, a targeting liposome, was also shown to have an effect in suppressing cancer cells similar to free SAHA especially. The fact that the early apoptosis after drug treatment is significantly higher in Aptamosome than in SAHA and Liposome means that the drug delivery effect is more specific. This can be compared with the results of animal experiments showing that non-targeting liposomes and SAHA drugs lack the ability to target cancer cells and thus lack the cancer cell inhibition effect (Fig. 5). These findings highlight the important role of PD-L1 aptamers in enhancing tumor targeting ability.

Anticancer immune responses to aptamosomes were evaluated in vitro and in vivo. These results confirmed the cytotoxic effect and induction of CXCL10, a chemokine that recruits T cells, through aptamosome treatment of LL/2 cells (Figs. 3 and 4B). Aptamosomes (SAHA) increased toxic chemokine levels, and these results show that aptamosomes [SAHA] deliver SAHA to the effector group through an aptamer bound to liposomes and at the same time target cancer cells by the PD-L1 aptamer. Evidence that blocking PD-L1 on the surface to some extent activates immune responses and inhibits cell growth is also explained through research results from other research institutes (Kim et al., 2022). Next, in vivo inhibition of tumor growth was demonstrated in LL/2 tumor-xenograft mice treated with aptamosome therapeutics. The tumor volume of mice treated with free SAHA or liposomes [SAHA] was significantly increased, whereas the tumor volume of mice treated with aptamosomes [SAHA] was not significantly increased (Fig. 5).

Finally, tumor shrinkage was confirmed via histopathological analysis. H&E images of tumor tissues treated with aptamosome [SAHA] showed an increase in eosinophilic cytoplasm and more cell shrinkage than the other treatment groups (Fig. 6). These results suggest that there was a significant occurrence of tumor apoptosis, as indicated by the observed cell shrinkage and hypereosinophilic cytoplasm in apoptotic tumor cells (Nayak et al., 2016; Jain et al., 2009).

In summary, we have developed aptamer-conjugated liposomes for drug delivery to induce anticancer immune responses. Liposomes containing SAHA were conjugated to PD-L1 aptamers to target the surface of PD-L1-expressing cancer cells (aptamosome [SAHA]). These aptamer-linked liposomes were delivered to LL/2 cells in vitro and demonstrated anticancer and chemokine-inducing effects. Treatment of LL/2 experimental xenograft mice with aptamosome [SAHA] suppressed tumor growth and lung metastasis, demonstrating its therapeutic functions in vivo. Moreover, the engineered aptamosome [SAHA] induced T cells and increased chemokine secretion.

However, there are research reports that aptamers can cause immune plaques and rarely cause allergic reactions when introduced into in vivo, and there are literature reports that some aptamers can cause cytotoxicity when used in high concentrations, so an appropriate dosage is required when using them (Ganson et al., 2016; Gilboa et al., 2015). We believe that this shortcoming can be overcome if aptamer-based therapy is adjusted, and we think that a lot of additional immune-linked research should be conducted in the future. In conclusion, this aptamosome is a promising delivery system that may lead to anticancer immune responses due to its aggressive targeting ability and synergistic tumor-suppressive effects.

ACKNOWLEDGEMENT

Schematic Fig. 5A created with BioRender.com (2024).

CONFLICT OF INTEREST

The authors have no conflict of interest to declare.

References
  1. Abdel-Rahman O, Helbling D, Schmidt JSchmidt J et al. Treatment-associated fatigue in cancer patients treated with immune checkpoint inhibitors; a systematic review and meta-analysis. Clin Oncol (R. Coll. Radiol). 2016. 28: 127.
    Pubmed CrossRef
  2. Agnello L, Camorani S, Fedele M, Cerchia L. Aptamers and antibodies: Rivals or allies in cancer targeted therapy? Explor Target Antitumor Ther. 2021. 2: 107-121.
    Pubmed KoreaMed CrossRef
  3. Akbarzadeh A, Rezaei-Sadabady R, Davaran SDavaran S et al. Liposome: Classification, preparation, and applications. Nanoscale Res Lett. 2013. 8: 102-102.
    Pubmed KoreaMed CrossRef
  4. Alshaer W, Hillaireau H, Vergnaud JVergnaud J et al. Aptamer-guided siRNA-loaded nanomedicines for systemic gene silencing in CD-44 expressing murine triple-negative breast cancer model. J Control Release. 2018. 271: 98-106.
    Pubmed CrossRef
  5. Bagalkot V, Farokhzad OC, Langer R, Jon S. An aptamer-doxorubicin physical conjugate as a novel targeted drug-delivery platform. Angewandte Chemie International Edition. 2006. 45: 8149-8152.
    Pubmed CrossRef
  6. Baxi S, Yang A, Gennarelli RLGennarelli RL et al. Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: Systematic review and meta-analysis. BMJ. 2018. 360: k793.
    Pubmed KoreaMed CrossRef
  7. Chen S, Dominik PK, Stanfield JStanfield J et al. Dual checkpoint blockade of CD47 and PD-L1 using an affinity-tuned bispecific antibody maximizes antitumor immunity. J Immunother Cancer. 2021. 9: e003464. doi: 10.1136/jitc-003464.
    Pubmed KoreaMed CrossRef
  8. Conte M, De Palma R, Altucci L. HDAC inhibitors as epigenetic regulators for cancer immunotherapy. Int J Biochem Cell Biol. 2018. 98: 65-74.
    Pubmed CrossRef
  9. Ganson NJ, Povsic TJ, Sullenger BASullenger BA et al. Pre-existing anti-polyethylene glycol antibody linked to first-exposure allergic reactions to pegnivacogin, a PEGylated RNA aptamer. J Allergy Clin Immunol. 2016. 137: 1610-1613.e7.
    Pubmed KoreaMed CrossRef
  10. Gao T, Mao Z, Li W, Pei R. Anti-PD-L1 DNA aptamer antagonizes the interaction of PD-1/PD-L1 with antitumor effect. J Mater Chem B. 2021. 9: 746-756.
    Pubmed CrossRef
  11. Gilboa E, Berezhnoy A, Schrand B. Reducing toxicity of immune therapy using aptamer-targeted drug delivery. Cancer Immunol Res. 2015. 3: 1195-1200.
    Pubmed CrossRef
  12. Granier C, De Guillebon E, Blanc CBlanc C et al. Mechanisms of action and rationale for the use of checkpoint inhibitors in cancer. ESMO Open. 2017. 2: e000213-000213. eCollection 2017.
    Pubmed KoreaMed CrossRef
  13. Jain A, Maheshwari V, Alam K, Mehdi G, Sharma SC. Apoptosis in premalignant and malignant squamous cell lesions of the oral cavity: A light microscopic study. Indian J Pathol Microbiol. 2009. 52: 164-166.
    Pubmed CrossRef
  14. Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin. 2020. 70: 86-104.
    Pubmed CrossRef
  15. Kim M, Lee JS, Kim WKim W et al. Aptamer-conjugated nano-liposome for immunogenic chemotherapy with reversal of immunosuppression. J Control Release. 2022. 348: 893-910.
    Pubmed CrossRef
  16. Large DE, Abdelmessih RG, Fink EA, Auguste DT. Liposome composition in drug delivery design, synthesis, characterization, and clinical application. Adv Drug Deliv Rev. 2021. 176: 113851.
    Pubmed CrossRef
  17. Le VKH, Pham TPD, Truong DH. Delivery systems for vorinostat in cancer treatment: An updated review. Journal of Drug Delivery Science and Technology. 2021. 61: 102334.
    CrossRef
  18. Li T, Yao F, An Y, Li X, Duan J, Yang X. Novel complex of PD-L1 aptamer and holliday junction enhances antitumor efficacy in vivo. Molecules. 2021. 26: 1067. doi: 10.3390/molecules 26041067.
    Pubmed KoreaMed CrossRef
  19. Li Z, Song W, Rubinstein M, Liu D. Recent updates in cancer immunotherapy: A comprehensive review and perspective of the 2018 china cancer immunotherapy workshop in beijing. J. Hematol Oncol. 2018. 11: 142-143.
    Pubmed KoreaMed CrossRef
  20. Liu B, Zhang J, Liao JLiao J et al. Aptamer-functionalized nanoparticles for drug delivery. J Biomed Nanotechnol. 2014. 10: 3189-3203.
    Pubmed CrossRef
  21. Lv H, Wang T, Ma FMa F et al. Aptamer-functionalized targeted siRNA delivery system for tumor immunotherapy. Biomed Mater. 2022. 17. 10.1088/1748-605X/ac5415.
    CrossRef
  22. Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 2007. 12: 1247-1252.
    Pubmed CrossRef
  23. Martins F, Sofiya L, Sykiotis GPSykiotis GP et al. Adverse effects of immune-checkpoint inhibitors: Epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019. 16: 563-580.
    Pubmed CrossRef
  24. Melosky B, Juergens R, McLeod DMcLeod D et al. Immune checkpoint-inhibitors and chemoradiation in stage III unresectable non-small cell lung cancer. Lung Cancer. 2019. 134: 259-267.
    Pubmed CrossRef
  25. Nayak A, Raikar A, Kotrashetti V, Nayak R, Shree S, Kambali S. Histochemical detection and comparison of apoptotic cells in the gingival epithelium using hematoxylin and eosin and methyl green-pyronin: A pilot study. J Indian Soc Periodontol. 2016. 20: 294-298.
    Pubmed KoreaMed CrossRef
  26. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nano-Enabled Medical Applications. 2020. 61-91.
    CrossRef
  27. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019. 18: 175-196.
    Pubmed KoreaMed CrossRef
  28. Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: From enhancement to normalization. Cell. 2018. 175: 313-326.
    Pubmed KoreaMed CrossRef
  29. Terranova-Barberio M, Thomas S, Ali NAli N et al. HDAC inhibition potentiates immunotherapy in triple negative breast cancer. Oncotarget. 2017. 8: 114156-114172.
    Pubmed KoreaMed CrossRef
  30. West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Invest. 2014. 124: 30-39.
    Pubmed KoreaMed CrossRef
  31. Xu W, Siddiqui IA, Nihal MNihal M et al. Aptamer-conjugated and doxorubicin-loaded unimolecular micelles for targeted therapy of prostate cancer. Biomaterials. 2013. 34: 5244-5253.
    Pubmed KoreaMed CrossRef
  32. Zheng H, Zhao W, Yan CYan C et al. HDAC inhibitors enhance T-cell chemokine expression and augment response to PD-1 immunotherapy in lung adenocarcinoma. Clin Cancer Res. 2016. 22: 4119-4132.
    Pubmed KoreaMed CrossRef