ArticlePublisher preview available

Delivery of chemotherapeutic drug targeting folate receptor to oral cancer cells using functionalized carbon nanospheres

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Folate receptor (FR) has long been the subject of active interest as regards its potential to serve as a target for cancer therapy. FR is overexpressed in several cancers, including clinical samples of different stages from Oral Squamous Cell Carcinoma (OSCC) patients. However, no clear correlation or conclusive finding has been obtained so far which might indicate the efficacy of FR as a credible target for the treatment of OSCC. Methods: All cell lines to be used were assessed for FR expression. Subsequently, we developed glucose-derived carbon nanospheres (CSPs) and primed them with a Folate-based cationic lipid FA8 and the chemotherapeutic drug doxorubicin (DOX). CSP-based delivery systems along with the pristine drug Doxorubicin were characterized and treated subsequently to in vitro cultures of OSCC cells and assessed for cancer cell targetability as well as cell death. Subsequently, treatment was administered to immunocompetent C57 mice carrying MOC2-based syngeneic OSCC tumors and assessed for tumor regression and toxicity. Results: Ligand-primed targeted CSPs exhibited commendable drug uptake as well as efficient induction of cell death. Further, receptor-blocking studies revealed FR-mediated uptake, preferentially in cancer cells. Drug once delivered by ligand-primed CSPs was retained longer inside cells than the pristine drug alone, indicating possibilities of better therapeutic outcome. In animal studies, CSP-FA8-DOX (Ligand primed targeted CSP) demonstrated significant regression in tumor size compared to pristine Doxorubicin as well as CSP-DOX (non-targeted CSP) treated animals. Conclusions: FR-mediated system CFD demonstrated targeted drug uptake and apoptotic death selectively in cancer cells. Significant tumor regression was also observed in vivo. Overall, it may be presumed that the Folate receptor is a therapeutic target with substantial potential in OSCC treatment. &#xD.
This content is subject to copyright. Terms and conditions apply.
Biomed. Mater. 18 (2023) 055012 https://doi.org/10.1088/1748-605X/ace8de
Biomedical Materials
RECEIVED
12 March 2023
REVISED
9 July 2023
ACC EPT ED FOR PUB LICATI ON
19 July 2023
PUBLISHED
2 August 2023
PAPER
Delivery of chemotherapeutic drug targeting folate receptor to oral
cancer cells using functionalized carbon nanospheres
Dwaipayan Bhattacharya1, Kalyani Sakhare1, Chhavi Dhiman1, Aasia Ansari2,4, Tapas Kumar Kundu3,
Kumar Pranav Narayan1,and Rajkumar Banerjee2,4,
1Department of Biological Science, Birla Institute of Technology & Science Pilani, Hyderabad Campus, Jawaharnagar, Kapra Mandal,
Hyderabad, Telangana 500078, India
2Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, Telangana 500076, India
3Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centrefor Advanced Scientific Research,
Bangalore 560064, India
4Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
Authors to whom any correspondence should be addressed.
E-mail: pranav@hyderabad.bits-pilani.ac.in and banerjee@iict.res.in
Keywords: carbon nanospheres, folate receptor, oral squamous cell carcinoma, targeted drug retention, apoptosis, EMT,
tumour regression
Supplementary material for this article is available online
Abstract
Folate receptor (FR) (α) has long been the subject of active interest as regards its potential to serve
as a target for cancer therapy. FR has been found to be overexpressed in several cancers, including
clinical samples of different stages from OSCC (oral squamous cell carcinoma) patients. However,
no clear correlation or conclusive finding has been obtained so far which might indicate the
efficacy of FR as a credible target for the treatment of OSCC. All cell lines to be used were assessed
for FR-expression. Subsequently, we developed glucose-derived carbon nanospheres (CSPs) and
primed them with a Folate-based cationic lipid FA8 and the chemotherapeutic drug doxorubicin
(DOX). CSP based delivery systems along with pristine drug DOX were characterized and treated
subsequently to in vitro cultures of OSCC cells and assessed for cancer cell targetability as well as
cell death. Subsequently, treatment was administered to immunocompetent C57 mice carrying
MOC2 based syngeneic OSCC tumours and assessed for tumour regression and toxicity. Ligand
primed targeted CSPs exhibited commendable drug uptake as well as efficient induction of cell
death. Further, receptor blocking studies revealed FR-mediated uptake, preferentially in cancer
cells. Drug once delivered by ligand-primed CSPs was retained longer inside cells than pristine
drug alone, indicating possibilities of better therapeutic outcome. In animal studies,
CSP-FA8-DOX (Ligand primed targeted CSP) demonstrated significant regression in tumour size
compared to pristine DOX as well as CSP-DOX (non-targeted CSP) treated animals. FR-mediated
system CFD demonstrated targeted drug uptake and apoptotic death selectively in cancer cells.
Significant tumour regression was also observed in vivo. Overall, it may be presumed that the FR is
a therapeutic target with substantial potential in OSCC treatment.
1. Introduction
Cancer drug delivery is no longer about just wrap-
ping drugs in new formulations to suit differ-
ent routes of delivery. Surface functionalization for
optimal utilization of advanced nanomaterials is of
paramount interest to today’s researchers and medical
professionals.
Currently, the sixth most common cancer in
the world, head, and neck cancer continues to
take its toll on the lives of millions [1]. When
oral cancer stops responding to radiation and
chemotherapy, targeted therapy may be used to
help control the disease. Targeted drugs often
alter particular aspects of cancer cell growth.
These drugs may be used alone or in combination
© 2023 IOP Publishing Ltd
Preprint
Full-text available
To date, the treatment options for metastatic and recurrent tongue squamous cell carcinoma (TSCC) remain limited due to the lack of effective drug targets and therapeutics. Here we report the identification of ICAM1 as a TSCC target candidate for the development of antibody-drug conjugates (ADC), an emerging class of targeted therapeutics. We first performed an unbiased and quantitative screening of a panel of 69 TSCC cell surface antigens that identified ICAM1 as the most abundant hit. The overexpression level of ICAM1 was validated in 26 TSCC clinical specimens and four cell lines along with genomic information of 153 TSCC patients from the TCGA database. Subsequently, two ICAM1-targeted ADCs were designed and synthesized using two cytotoxic payloads of different mechanisms of action: monomethyl auristatin E (MMAE), a microtubule inhibitor, and deruxtecan (DXd), a type I DNA topoisomerase inhibitor, respectively. The therapeutic efficacy of these ICAM1 ADCs was determined in vitro and in vivo. Both ADCs selectively and potently ablate TSCC tumors in the established SAS cell line and patient-derived xenograft models. Our experimental results strongly support ICAM1 as a promising ADC target candidate for TSCC therapy.
Article
Full-text available
Intraperitoneal (IP) aerosolized anticancer drug delivery was recently introduced in the treatment of patients with peritoneal metastases. However, little is known on the effect of treatment parameters on the spatial distribution of the aerosol droplets in the peritoneal cavity. Here, computational fluid dynamics (CFD) modeling was used in conjunction with experimental validation in order to investigate the effect of droplet size, liquid flow rate and viscosity, and the addition of an electrostatic field on the homogeneity of IP aerosol. We found that spatial distribution is optimal with small droplet sizes (1–5 µm). Using the current clinically used technology (droplet size of 30 µm), the optimal spatial distribution of aerosol is obtained with a liquid flow rate of 0.6 mL/s. Compared to saline, nebulization of higher viscosity liquids results in less homogeneous aerosol distribution. The addition of electrostatic precipitation significantly improves homogeneity of aerosol distribution, but no further improvement is obtained with voltages higher than 6.5 kV. The results of the current study will allow to choose treatment parameters and settings in order to optimize spatial distribution of IP aerosolized drug, with a potential to enhance its anticancer effect.
Article
Full-text available
Aims Acute myeloblastic leukemia (AML) is the most common type of acute leukemia in adults. Despite numerous treatment strategies including chemotherapy and radiotherapy, a large number of patients do not respond to treatment and experience relapse. The main problem of these patients is the development of resistance to anti-cancer drugs. Therefore, any endeavor to reduce drug resistance in these patients is of high priority. In general, several mechanisms such as changes in drug metabolic pathways, drug inactivation, drug target alterations and reduced drug accumulation in the cells contribute to drug resistance of cancer cells. In this context, evidence suggests that exosomes could reduce drug resistance by removing drugs from their parent cells. In the present study, we aimed to investigate the effects of exosome release inhibition on the resistance of U937 cells to PEGylated liposomal doxorubicin (PLD). Main Methods In order to find a suitable ABCG2 (ATP-binding cassette sub-family G member 2) transporter substrate, virtual screening was performed among a list of drugs used in leukemia and PLD was selected. U937 cells were treated with PLD with/without co-treatment with the exosome release inhibitor, GW4869. Released exosomes within different study groups were isolated and characterized to determine the differences between groups. Doxorubicin presence in the isolated exosomes was also measured by high performance liquid chromatography (HPLC) to confirm drug export through the exosomes. Finally, the effect of exosome inhibition on the cytotoxicity of PLD on U937 cells was determined using different cytotoxicity assays including the standard lactate dehydrogenase (LDH) release assay and the flow cytometric analysis of apoptotic and non-apoptotic cell death. Key Findings GW4869 treatment caused a significant decrease in the exosome release of U937 cells compared to the untreated cells, as evidenced by the reduction of the protein content of the isolated exosomes (P<0.05). Co-treatment with GW4869 significantly increased cytotoxic cell death in the groups treated with 0.5 and 1 µM PLD, compared to the same groups without GW4869 co-treatment (P<0.05). Interestingly, co-treatment with GW4896 and 0.5 µM PLD was enough to induce the same cytotoxic effect as that of the sole 1 µM PLD group. Significance Our findings showed that U937 cells increase their resistance against the cytotoxic effects of PLD through the exosome-mediated expelling of the drug. Inhibition of exosome release could prevent PLD efflux and consequently increase the vulnerability of the U937 cells to the cytotoxic effects of PLD. Our results along with prior studies indicate that the integration of exosome release inhibitors into the common PLD-containing chemotherapy regimens could significantly lower the required concentrations of the drug and consequently reduce its associated side effects. Further studies are warranted to identify clinically safe inhibitors and investigate their clinical efficacy.
Article
Full-text available
Cancer remains one of the highest leading causes of morbidity and mortality worldwide. Anthracyclines, specifically Doxorubicin (DOX), have been used for the past three decades as a treatment against a number of cancers. However, its use has been limited due to its severe side effects and toxicity arising during or after treatment. Ample research has already taken place and is still being undertaken in order to understand the mode of action of anthracyclines, including DOX. However, despite the work carried out; the mechanisms proposed remain controversial. Other research has also taken place to get a better understanding of the cell death and growth arrest pathways triggered by DOX. Even though DOX remains one of the most effective chemotherapeutic drugs, resistance development in cancer cells remains a major barrier to effective treatment when using this drug. Apart from the already known mechanisms of DOX chemoresistance, research has shown that post-translational modifications on certain proteins can also contribute to DOX chemoresistance. However, the mechanisms by which DOX resistance arises remain poorly defined. This review tackles some of the currently understood and proposed models for the mode of action of DOX, including the cell death mechanisms triggered by DOX and the DOX resistance mechanisms arising during treatment. By further understanding how DOX functions, its influence on cell biological events and the mechanisms contributing to DOX resistance; it can further help in improving the efficiency and efficacy of the drug, together with decreasing its toxicity.
Article
Full-text available
Background The receptor-interacting protein kinase 3 (RIP3/RIPK3) was recently found to be a critical regulator of programmed necrosis/necroptosis. However, the biological role and clinical significance of RIP3 in prostate cancer remain obscure.Methods Western blotting and QRT-PCR were performed to detect the level of RIP3 in prostate cancer cells. Fixed cancer tissue and normal tissue specimens were subjected to immunohistochemical analysis of RIP3. Cell migration and invasion abilities were evaluated by transwell assays. In vitro proliferative ability was examed by MTS. And in vivo nude mice model were used to evaluate the effect of RIP3 ectopic expression on proliferative capability. Cell cycle of prostate cancer cells were analyzed by flow cytometry. Changes in some related proteins caused by RIP3 overexpression were explored using Western blotting.ResultsRIP3 was significantly down-regulated in prostate cancer cell lines and clinical prostate tumor samples. And over-expressing RIP3 suppressed the migration and invasion of prostate cancer cells. Two important matrix metalloproteinases MMP2, MMP9 which enables the destruction of the histological barrier of tumor cell invasion and three mesenchymal markers Vimentin, fibronectin, and N-cadherin were under-expressed due to the overexpression of RIP3, but the E-cadherin level which is the epithelial marker was increased. Furthermore, our results also showed that RIP3 can inhibit the proliferation and tumorigenicity of prostate cancer cells both in vitro and in vivo by phosphorylating MLKL, which were reversed by MLKL inhibitor treatment, indicating that necroptosis was involved in cell death.Conclusion Taken together, these findings indicated that RIP3 is responsible for the progression of prostate cancer, suggesting that RIP3 might have the potential to be a prognostic marker or a therapeutic target against prostate cancer.
Article
Full-text available
Recent advances in the development of new methods of cancer immunotherapy require the production of complex cancer animal models that reliably reflect the complexity of the tumor and its microenvironment. Mice are good animals to create tumor models because they are low cost, have a short reproductive cycle, exhibit high tumor growth rates, and can be easily genetically modified. However, the obvious problem of these models is the high failure rate observed in human clinical trials after promising results obtained in mouse models. In order to increase the reliability of the results obtained in mice, the tumor model should reflect the heterogeneity of the tumor, contain components of the tumor microenvironment, in particular immune cells, to which the action of immunotherapeutic drugs are directed. This review discusses the current immunocompetent and immunocompromised mouse models of human tumors that are used to evaluate the effectiveness of immunotherapeutic agents, in particular chimeric antigen receptor (CAR) T-cells and immune checkpoint inhibitors.
Article
Full-text available
Cancer is the second leading cause of death in the US. Current major treatments for cancer management include surgery, cytotoxic chemotherapy, targeted therapy, radiation therapy, endocrine therapy and immunotherapy. Despite the endeavors and achievements made in treating cancers during the past decades, resistance to classical chemotherapeutic agents and/or novel targeted drugs continues to be a major problem in cancer therapies. Drug resistance, either existing before treatment (intrinsic) or generated after therapy (acquired), is responsible for most relapses of cancer, one of the major causes of death of the disease. Heterogeneity among patients and tumors, and the versatility of cancer to circumvent therapies make drug resistance more challenging to deal with. Better understanding the mechanisms of drug resistance is required to provide guidance to future cancer treatment and achieve better outcomes. In this review, intrinsic and acquired resistance will be discussed. In addition, new discoveries in mechanisms of drug resistance will be reviewed. Particularly, we will highlight roles of ATP in drug resistance by discussing recent findings of exceptionally high levels of intratumoral extracellular ATP as well as intracellular ATP internalized from extracellular environment. The complexity of drug resistance development suggests that combinational and personalized therapies, which should take ATP into consideration, might provide better strategies and improved efficacy for fighting drug resistance in cancer.
Article
There is an increasing interest in intraperitoneal delivery of chemotherapy as an aerosol in patients with peritoneal metastasis. The currently used technology is hampered by inhomogenous drug delivery throughout the peritoneal cavity because of gravity, drag, and inertial impaction. Addition of an electrical force to aerosol particles, exerted by an electrostatic field, can improve spatial aerosol homogeneity and enhance tissue penetration. A computational fluid dynamics model shows that electrostatic precipitation (EP) results in a significantly improved aerosol distribution. Fluorescent nanoparticles (NPs) remain stable after nebulization in vitro, while EP significantly improves spatial homogeneity of NP distribution. Next, pressurized intraperitoneal chemotherapy with and without EP using NP albumin bound paclitaxel (Nab‐PTX) in a novel rat model is examined. EP does not worsen the effects of CO2 insufflation and intraperitoneal Nab‐PTX on mesothelial structural integrity or the severity of peritoneal inflammation. Importantly, EP significantly enhances tissue penetration of Nab‐PTX in the anatomical regions not facing the nozzle of the nebulizer. Also, the addition of EP leads to more homogenous peritoneal tissue concentrations of Nab‐PTX, in parallel with higher plasma concentrations. In conclusion, EP enhances spatial homogeneity and tissue uptake after intraperitoneal nebulization of anticancer NPs.
Article
Folate receptor α (FRα) came into focus as an anticancer target many decades after the successful development of drugs targeting intracellular folate metabolism, such as methotrexate and pemetrexed. Binding to FRα is one of several methods by which folate is taken up by cells; however, this receptor is an attractive anticancer drug target owing to the overexpression of FRα in a range of solid tumours, including ovarian, lung and breast cancers. Furthermore, using FRα to better localize effective anticancer therapies to their target tumours using platforms such as antibody–drug conjugates, small-molecule drug conjugates, radioimmunoconjugates and, more recently, chimeric antigen receptor T cells could further improve the outcomes of patients with FRα-overexpressing cancers. FRα can also be harnessed for predictive biomarker research. Moreover, imaging FRα radiologically or in real time during surgery can lead to improved functional imaging and surgical outcomes, respectively. In this Review, we describe the current status of research into FRα in cancer, including data from several late-phase clinical trials involving FRα-targeted therapies, and the use of new technologies to develop FRα-targeted agents with improved therapeutic indices.
Article
In this study, a drug delivery system called glutathione (GSH)-sensitive and folate acid(FA)-targeted nanoparticles loaded paclitaxel (FA-PEG-S-S-PCL@PTX, FA-NPs) was developed. First, we proved that FA receptor was significantly expressed in 95 oral squamous cancer cell (OSCC) specimens (57.9%). It provided the feasibility for FA targeted nanoparticles be released in tumour site for patients with OSCC. Next, the FA-NPs was synthesized and characterized. In vitro, we found enhancement of FA-mediated endocytosis in HSC3 cells with FA overexpression. Therefore, Paclitaxel (PTX) from the FA-NPs can be precisely released due to disulfide bonds cleaved by redox reaction. In vivo, the FA-NPs could be accumulated in mice bearing HSC3 cells, where it exhibited effectively antitumor effects, compared to treatment with free PTX, PEG-S-S-PCL@PTX. In summary, this novel drug system has an opportunity to improve OSCC treatment.