Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization

Metal nanoparticles are effective radiosensitizers that locally enhance radiation doses in targeted cancer cells. Compared with other metal nanoparticles, gold nanoparticles (GNPs) exhibit high biocompatibility, low toxicity, and they increase secondary electron scatter. Herein, we investigated the...

Full description

Bibliographic Details
Main Authors: Daiki Hara, Wensi Tao, Ryder M. Schmidt, Yu-Ping Yang, Sylvia Daunert, Nesrin Dogan, John Chetley Ford, Alan Pollack, Junwei Shi
Format: Article
Language:English
Published: MDPI AG 2022-12-01
Series:Nanomaterials
Subjects:
Online Access:https://www.mdpi.com/2079-4991/12/24/4440
_version_ 1797455969299464192
author Daiki Hara
Wensi Tao
Ryder M. Schmidt
Yu-Ping Yang
Sylvia Daunert
Nesrin Dogan
John Chetley Ford
Alan Pollack
Junwei Shi
author_facet Daiki Hara
Wensi Tao
Ryder M. Schmidt
Yu-Ping Yang
Sylvia Daunert
Nesrin Dogan
John Chetley Ford
Alan Pollack
Junwei Shi
author_sort Daiki Hara
collection DOAJ
description Metal nanoparticles are effective radiosensitizers that locally enhance radiation doses in targeted cancer cells. Compared with other metal nanoparticles, gold nanoparticles (GNPs) exhibit high biocompatibility, low toxicity, and they increase secondary electron scatter. Herein, we investigated the effects of active-targeting GNPs on the radiation-induced bystander effect (RIBE) in prostate cancer cells. The impact of GNPs on the RIBE presents implications for secondary cancers or spatially fractionated radiotherapy treatments. Anti-prostate-specific membrane antigen (PSMA) antibodies were conjugated with PEGylated GNPs through EDC–NHS chemistry. The media transfer technique was performed to induce the RIBE on the non-irradiated bystander cells. This study focused on the LNCaP cell line, because it can model a wide range of stages relating to prostate cancer progression, including the transition from androgen dependence to castration resistance and bone metastasis. First, LNCaP cells were pretreated with phosphate buffered saline (PBS) or PSMA-targeted GNPs (PGNPs) for 24 h and irradiated with 160 kVp X-rays (0–8 Gy). Following that, the collected culture media were filtered (sterile 0.45 µm polyethersulfone) in order to acquire PBS- and PGNP- conditioned media (CM). Then, PBS- and PGNP-CM were transferred to the bystander cells that were loaded with/without PGNPs. MTT, γ-H2AX, clonogenic assays and reactive oxygen species assessments were performed to compare RIBE responses under different treatments. Compared with 2 Gy-PBS-CM, 8 Gy-PBS-CM demonstrated a much higher RIBE response, thus validating the dose dependence of RIBE in LNCaP cells. Compared with PBS-CM, PGNP-CM exhibited lower cell viability, higher DNA damage, and a smaller survival fraction. In the presence of PBS-CM, bystander cells loaded with PGNPs showed increased cell death compared with cells that did not have PGNPs. These results demonstrate the PGNP-boosted expression and sensitivity of RIBE in prostate cancer cells.
first_indexed 2024-03-09T16:01:44Z
format Article
id doaj.art-dbbd367be14b41738e00c4f715684e17
institution Directory Open Access Journal
issn 2079-4991
language English
last_indexed 2024-03-09T16:01:44Z
publishDate 2022-12-01
publisher MDPI AG
record_format Article
series Nanomaterials
spelling doaj.art-dbbd367be14b41738e00c4f715684e172023-11-24T17:04:24ZengMDPI AGNanomaterials2079-49912022-12-011224444010.3390/nano12244440Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer RadiosensitizationDaiki Hara0Wensi Tao1Ryder M. Schmidt2Yu-Ping Yang3Sylvia Daunert4Nesrin Dogan5John Chetley Ford6Alan Pollack7Junwei Shi8Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USADepartment of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USADepartment of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USASylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USASylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USADepartment of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USADepartment of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USADepartment of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USADepartment of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USAMetal nanoparticles are effective radiosensitizers that locally enhance radiation doses in targeted cancer cells. Compared with other metal nanoparticles, gold nanoparticles (GNPs) exhibit high biocompatibility, low toxicity, and they increase secondary electron scatter. Herein, we investigated the effects of active-targeting GNPs on the radiation-induced bystander effect (RIBE) in prostate cancer cells. The impact of GNPs on the RIBE presents implications for secondary cancers or spatially fractionated radiotherapy treatments. Anti-prostate-specific membrane antigen (PSMA) antibodies were conjugated with PEGylated GNPs through EDC–NHS chemistry. The media transfer technique was performed to induce the RIBE on the non-irradiated bystander cells. This study focused on the LNCaP cell line, because it can model a wide range of stages relating to prostate cancer progression, including the transition from androgen dependence to castration resistance and bone metastasis. First, LNCaP cells were pretreated with phosphate buffered saline (PBS) or PSMA-targeted GNPs (PGNPs) for 24 h and irradiated with 160 kVp X-rays (0–8 Gy). Following that, the collected culture media were filtered (sterile 0.45 µm polyethersulfone) in order to acquire PBS- and PGNP- conditioned media (CM). Then, PBS- and PGNP-CM were transferred to the bystander cells that were loaded with/without PGNPs. MTT, γ-H2AX, clonogenic assays and reactive oxygen species assessments were performed to compare RIBE responses under different treatments. Compared with 2 Gy-PBS-CM, 8 Gy-PBS-CM demonstrated a much higher RIBE response, thus validating the dose dependence of RIBE in LNCaP cells. Compared with PBS-CM, PGNP-CM exhibited lower cell viability, higher DNA damage, and a smaller survival fraction. In the presence of PBS-CM, bystander cells loaded with PGNPs showed increased cell death compared with cells that did not have PGNPs. These results demonstrate the PGNP-boosted expression and sensitivity of RIBE in prostate cancer cells.https://www.mdpi.com/2079-4991/12/24/4440gold nanoparticleradiosensitizationradiation-induced bystander effectprostate LNCaP cancer cellsprostate-specific membrane antigenactive targeting
spellingShingle Daiki Hara
Wensi Tao
Ryder M. Schmidt
Yu-Ping Yang
Sylvia Daunert
Nesrin Dogan
John Chetley Ford
Alan Pollack
Junwei Shi
Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization
Nanomaterials
gold nanoparticle
radiosensitization
radiation-induced bystander effect
prostate LNCaP cancer cells
prostate-specific membrane antigen
active targeting
title Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization
title_full Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization
title_fullStr Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization
title_full_unstemmed Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization
title_short Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization
title_sort boosted radiation bystander effect of psma targeted gold nanoparticles in prostate cancer radiosensitization
topic gold nanoparticle
radiosensitization
radiation-induced bystander effect
prostate LNCaP cancer cells
prostate-specific membrane antigen
active targeting
url https://www.mdpi.com/2079-4991/12/24/4440
work_keys_str_mv AT daikihara boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization
AT wensitao boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization
AT rydermschmidt boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization
AT yupingyang boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization
AT sylviadaunert boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization
AT nesrindogan boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization
AT johnchetleyford boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization
AT alanpollack boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization
AT junweishi boostedradiationbystandereffectofpsmatargetedgoldnanoparticlesinprostatecancerradiosensitization