1
|
Takahashi M, Fujishiro J, Nomura S, Harada M, Hinoki A, Arake M, Ozeki E, Hara I, Satoh A, Tainaka T, Uchida HO, Morimoto Y. DDS-type near-infrared light absorber enables deeper lesion treatment in laser photothermal therapy while avoiding damage to surrounding organs. Front Bioeng Biotechnol 2024; 12:1444107. [PMID: 39211012 PMCID: PMC11357940 DOI: 10.3389/fbioe.2024.1444107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The efficacy of drug delivery system (DDS)-type near-infrared (NIR) absorbing agents in enhancing laser photothermal therapy is widely acknowledged. Despite the acknowledged efficacy, the therapeutic advantages of photothermal therapy using DDS-type NIR-absorbing agents over simple photothermal therapy without such agents have not been fully elucidated. This study was designed to investigate two primary objectives: firstly, the ability of DDS-type NIR-absorbing agents to induce cell death at greater depths within tumors, and secondly, their capacity to minimize collateral damage to adjacent healthy organs. To investigate these objectives, we employed a combination of indocyanine green lactosome-a DDS-type NIR-absorbing agent-and a precision-controlled laser hyperthermia system. An orthotopic neuroblastoma tumor model was used to closely simulate clinical conditions. The findings revealed that photothermal therapy using the DDS-type NIR-absorbing agent not only facilitates deeper penetration of cell death within tumors but also significantly mitigates thermal damage to surrounding healthy tissues, when compared to simple phototherapy without the agent. Furthermore, the combined treatment significantly prolonged the survival periods of the animals involved. This study is the first to analyze these therapeutic efficacies using quantitative data from an orthotopic tumor animal model and substantiated the potential of DDS-type NIR-absorbing agents to deepen the therapeutic impact of photothermal therapy while safeguarding vital organs, thereby enhancing overall treatment outcomes.
Collapse
Affiliation(s)
- Masataka Takahashi
- Department of Pediatric Surgery, The University of Tokyo, Tokyo, Japan
- Department of Cell Engineering, National Center for Child Health and Development, Tokyo, Japan
| | - Jun Fujishiro
- Department of Pediatric Surgery, The University of Tokyo, Tokyo, Japan
| | - Shinsuke Nomura
- Department of Surgery, National Defense Medical College, Tokorozawa, Japan
| | - Manabu Harada
- Department of Surgery, National Defense Medical College, Tokorozawa, Japan
| | - Akinari Hinoki
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masashi Arake
- Department of Physiology, National Defense Medical College, Tokorozawa, Japan
| | - Eiichi Ozeki
- Technology Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Isao Hara
- Technology Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Ayano Satoh
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama, Japan
| | - Takahisa Tainaka
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hiro-o Uchida
- Department of Pediatric Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuji Morimoto
- Department of Physiology, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
2
|
Pramanik N, Gupta A, Ghanwatkar Y, Mahato RI. Recent advances in drug delivery and targeting for the treatment of pancreatic cancer. J Control Release 2024; 366:231-260. [PMID: 38171473 PMCID: PMC10922996 DOI: 10.1016/j.jconrel.2023.12.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/24/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Despite significant treatment efforts, pancreatic ductal adenocarcinoma (PDAC), the deadliest solid tumor, is still incurable in the preclinical stages due to multifacet stroma, dense desmoplasia, and immune regression. Additionally, tumor heterogeneity and metabolic changes are linked to low grade clinical translational outcomes, which has prompted the investigation of the mechanisms underlying chemoresistance and the creation of effective treatment approaches by selectively targeting genetic pathways. Since targeting upstream molecules in first-line oncogenic signaling pathways typically has little clinical impact, downstream signaling pathways have instead been targeted in both preclinical and clinical studies. In this review, we discuss how the complexity of various tumor microenvironment (TME) components and the oncogenic signaling pathways that they are connected to actively contribute to the development and spread of PDAC, as well as the ways that recent therapeutic approaches have been targeted to restore it. We also illustrate how many endogenous stimuli-responsive linker-based nanocarriers have recently been developed for the specific targeting of distinct oncogenes and their downstream signaling cascades as well as their ongoing clinical trials. We also discuss the present challenges, prospects, and difficulties in the development of first-line oncogene-targeting medicines for the treatment of pancreatic cancer patients.
Collapse
Affiliation(s)
- Nilkamal Pramanik
- Department of Pharmaceutical Sciences, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aditya Gupta
- Department of Pharmaceutical Sciences, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yashwardhan Ghanwatkar
- Department of Pharmaceutical Sciences, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, the University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
3
|
Bentivoglio V, Nayak P, Varani M, Lauri C, Signore A. Methods for Radiolabeling Nanoparticles (Part 3): Therapeutic Use. Biomolecules 2023; 13:1241. [PMID: 37627307 PMCID: PMC10452659 DOI: 10.3390/biom13081241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Following previously published systematic reviews on the diagnostic use of nanoparticles (NPs), in this manuscript, we report published methods for radiolabeling nanoparticles with therapeutic alpha-emitting, beta-emitting, or Auger's electron-emitting isotopes. After analyzing 234 papers, we found that different methods were used with the same isotope and the same type of nanoparticle. The most common type of nanoparticles used are the PLGA and PAMAM nanoparticles, and the most commonly used therapeutic isotope is 177Lu. Regarding labeling methods, the direct encapsulation of the isotope resulted in the most reliable and reproducible technique. Radiolabeled nanoparticles show promising results in metastatic breast and lung cancer, although this field of research needs more clinical studies, mainly on the comparison of nanoparticles with chemotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, “Sapienza” University of Rome, 00185 Rome, Italy; (V.B.); (P.N.); (M.V.); (C.L.)
| |
Collapse
|
4
|
Marshall SK, Saelim B, Taweesap M, Pachana V, Panrak Y, Makchuchit N, Jaroenpakdee P. Anti-EGFR Targeted Multifunctional I-131 Radio-Nanotherapeutic for Treating Osteosarcoma: In Vitro 3D Tumor Spheroid Model. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3517. [PMID: 36234645 PMCID: PMC9565722 DOI: 10.3390/nano12193517] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
The systemic delivery of doxorubicin (DOX) to treat osteosarcoma requires an adequate drug concentration to be effective, but in doing so, it raises the risk of increasing organ off-target toxicity and developing drug resistance. Herein, this study reveals a multiple therapeutic nanocarrier delivery platform that overcomes off-target toxicity by providing good specificity and imparting enhanced tumor penetration in a three-dimensional (3D) human MG-63 spheroid model. By synthesizing PEG-PLGA nanoparticles by the double emulsion method, encapsulating DOX and Na131I in the inner core, and conjugating with an epidermal growth factor receptor (EGFR) antibody, it is intended to specifically target human MG-63 cells. The nanocarrier is biocompatible with blood and has good stability characteristics. Na131I encapsulation efficiency was >96%, and radiochemical purity was >96% over 96 h. A DOX encapsulation efficacy of ~80% was achieved, with a drug loading efficiency of ~3%, and a sustained DOX release over 5 days. The nanocarrier EGFR antibody achieved a ~80-fold greater targeting efficacy to MG-63 cells (EGFR+) than fibroblast cells (EGFR−). The targeted multiple therapeutic DIE-NPs have a higher penetration and uptake of Na131I to the 3D model and a ~3-fold higher cytotoxicity than the DOX monotherapy (D-NPs). The co-administration of DOX and Na131I (DIE-NPs) disrupts DNA repair and generates free radicals resulting in DNA damage, triggering the activation of apoptosis pathways. This leads to inhibition of MG-63 cell proliferation and promotes cell cycle arrest in the G0/G1 phase. Furthermore, the PEGylated anti-EGFR functionalized DIE-NPs were found to be biocompatible with red blood cells and to have no adverse effects. This anti-EGFR targeted multifunctional I-131 radio-nanotherapeutic signifies a customizable specific targeted treatment for osteosarcoma.
Collapse
Affiliation(s)
- Suphalak Khamruang Marshall
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Molecular Imaging and Cyclotron Center, Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Boonyisa Saelim
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Maneerat Taweesap
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Verachai Pachana
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Yada Panrak
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Naritsara Makchuchit
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Passara Jaroenpakdee
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
5
|
Go SI, Ko GH, Lee WS, Lee JH, Jeong SH, Lee YJ, Hong SC, Ha WS. Cyclin D1 Serves as a Poor Prognostic Biomarker in Stage I Gastric Cancer. Curr Issues Mol Biol 2022; 44:1395-1406. [PMID: 35723316 PMCID: PMC8947299 DOI: 10.3390/cimb44030093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/24/2022] Open
Abstract
TNM stage still serves as the best prognostic marker in gastric cancer (GC). The next step is to find prognostic biomarkers that detect subgroups with different prognoses in the same TNM stage. In this study, the expression levels of epidermal growth factor receptor (EGFR) and cyclin D1 were assessed in 96 tissue samples, including non-tumorous tissue, adenoma, and carcinoma. Then, the prognostic impact of EGFR and cyclin D1 was retrospectively investigated in 316 patients who underwent R0 resection for GC. EGFR positivity increased as gastric tissue became malignant, and cyclin D1 positivity was increased in all the tumorous tissues. However, there was no survival difference caused by the EGFR positivity, while the cyclin D1-postive group had worse overall survival (OS) than the cyclin D1-negative group in stage I GC (10-year survival rate (10-YSR): 62.8% vs. 86.5%, p = 0.010). In subgroup analyses for the propensity score-matched (PSM) cohort, there were also significant differences in the OS according to the cyclin D1 positivity in stage I GC but not in stage II and III GC. Upon multivariate analysis, cyclin D1 positivity was an independent prognostic factor in stage I GC. In conclusion, cyclin D1 may be a useful biomarker for predicting prognosis in stage I GC.
Collapse
Affiliation(s)
- Se-Il Go
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Institute of Health Sciences, Gyeongsang National University College of Medicine, Changwon 51472, Korea;
| | - Gyung Hyuck Ko
- Department of Pathology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (G.H.K.); (J.-H.L.)
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
- Correspondence: ; Tel.: +82-55-750-8733
| | - Jeong-Hee Lee
- Department of Pathology, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (G.H.K.); (J.-H.L.)
| | - Sang-Ho Jeong
- Department of Surgery, Gyeongsang National University Changwon Hospital, Institute of Health Sciences, Gyeongsang National University College of Medicine, Changwon 51472, Korea; (S.-H.J.); (Y.-J.L.)
| | - Young-Joon Lee
- Department of Surgery, Gyeongsang National University Changwon Hospital, Institute of Health Sciences, Gyeongsang National University College of Medicine, Changwon 51472, Korea; (S.-H.J.); (Y.-J.L.)
| | - Soon Chan Hong
- Department of Surgery, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (S.C.H.); (W.S.H.)
| | - Woo Song Ha
- Department of Surgery, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea; (S.C.H.); (W.S.H.)
| |
Collapse
|
6
|
Yan Z, Zhang X, Liu Y, Shen Y, Li N, Jia Q, Ji Y, Zhang P, Zhao L, Meng Z. HSA-MnO 2- 131I Combined Imaging and Treatment of Anaplastic Thyroid Carcinoma. Technol Cancer Res Treat 2022; 21:15330338221106557. [PMID: 35702054 PMCID: PMC9208040 DOI: 10.1177/15330338221106557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Purpose Compelling evidence suggests that nanoparticles (NPs) play a crucial role in cancer therapy. NPs templated with human serum albumin (HSA) has good retention in tumors. Manganese dioxide (MnO2) has been used to enhance the effect of radiotherapy. In this study, synthesized NPs using HSA-MnO2 labeled 131I to perform both imaging and therapy for anaplastic thyroid carcinoma (ATC). Method HSA-MnO2 was synthesized via HSA using a simple biomineralization method, and then labeled with Na131I by the chloramine T method. The cytotoxicity and biosafety of HSA-MnO2 were evaluated by the MTT test. The proliferation-inhibiting effect of HSA-MnO2-131I was evaluated in papillary thyroid cancer cell lines (K1, BCPAP, and KTC) and anaplastic thyroid carcinoma cell lines (Cal62, THJ16T, and ARO). For further translational application in medicine, we established a model of transplantable subcutaneously tumors in BALB\c-nu mice to assess the anti-tumor effect of HSA-MnO2-131I. The imaging effects of NPs were evaluated by MRI and SPECT/CT. Results The MTT test proved that the HSA-MnO2 had low toxicity. HSA-MnO2-131I significantly inhibited the proliferation of PTC and ATC cell lines. In addition, the results unveiled that HSA-MnO2-131I exhibited dual-modality MR/SPECT imaging for thyroid cancer visualization. In particular, HSA-MnO2-131I had an enhanced T1 signal in MR. Using SPECT/CT, we observed that HSA-MnO2-131I had good retention in tumor tissue, which was helpful for the diagnosis and treatment of tumor. In vivo assays indicated that the NPs led to a reduction in radioresistance in the tumor hypoxic microenvironment. Conclusion The nanomaterial had a simple synthesis method, good water solubility and biosafety, and good retention in tumor tissue. Hence, it could be used for SPECT/CT and MR dual mode imaging and therapy with radioiodine of tumor cells. The experimental results provided a feasible solution for combining radiotherapy and dual-model imaging by NPs for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Ziyu Yan
- Department of Nuclear Medicine, 117865Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Xuemei Zhang
- Department of Nuclear Medicine, 117865Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Yifan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, 12610Tianjin Medical University, Tianjin, China
| | - Yiming Shen
- Department of Nuclear Medicine, 117865Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Ning Li
- Department of Nuclear Medicine, 117865Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Qiang Jia
- Department of Nuclear Medicine, 117865Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Yanhui Ji
- Department of Nuclear Medicine, 117865Tianjin Medical University General Hospital, Tianjin, P. R. China
| | - Peitao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, 12610Tianjin Medical University, Tianjin, China
| | - Li Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, 12610Tianjin Medical University, Tianjin, China
| | - Zhaowei Meng
- Department of Nuclear Medicine, 117865Tianjin Medical University General Hospital, Tianjin, P. R. China
| |
Collapse
|
7
|
Bayoumi NA, El-Kolaly MT. Utilization of nanotechnology in targeted radionuclide cancer therapy: monotherapy, combined therapy and radiosensitization. RADIOCHIM ACTA 2021. [DOI: 10.1515/ract-2020-0098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
The rapid progress of nanomedicine field has a great influence on the different tumor therapeutic trends. It achieves a potential targeting of the therapeutic agent to the tumor site with neglectable exposure of the normal tissue. In nuclear medicine, nanocarriers have been employed for targeted delivery of therapeutic radioisotopes to the malignant tissues. This systemic radiotherapy is employed to overcome the external radiation therapy drawbacks. This review overviews studies concerned with investigation of different nanoparticles as promising carriers for targeted radiotherapy. It discusses the employment of different nanovehicles for achievement of the synergistic effect of targeted radiotherapy with other tumor therapeutic modalities such as hyperthermia and photodynamic therapy. Radiosensitization utilizing different nanosensitizer loaded nanoparticles has also been discussed briefly as one of the nanomedicine approach in radiotherapy.
Collapse
Affiliation(s)
- Noha Anwer Bayoumi
- Department of Radiolabeled Compounds , Hot Laboratories Center, Egyptian Atomic Energy Authority , Cairo , Egypt
| | - Mohamed Taha El-Kolaly
- Department of Radiolabeled Compounds , Hot Laboratories Center, Egyptian Atomic Energy Authority , Cairo , Egypt
| |
Collapse
|
8
|
Nanomedicines functionalized with anti-EGFR ligands for active targeting in cancer therapy: Biological strategy, design and quality control. Int J Pharm 2021; 605:120795. [PMID: 34119579 DOI: 10.1016/j.ijpharm.2021.120795] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Recently, active targeting using nanocarriers with biological ligands has emerged as a novel strategy for improving the delivery of therapeutic and/or imaging agents to tumor cells. The presence of active targeting moieties on the surface of nanomedicines has been shown to play an important role in enhancing their accumulation in tumoral cells and tissues versus healthy ones. This property not only helps to increase the therapeutic index but also to minimize possible side effects of the designed nanocarriers. Since the overexpression of epidermal growth factor receptors (EGFR) is a common occurrence linked to the progression of a broad variety of cancers, the potential application of anti-EGFR immunotherapy and EGFR-targeting ligands in active targeting nanomedicines is getting increasing attention. Henceforth, the EGFR-targeted nanomedicines were extensively studied in vitro and in vivo but exhibited both satisfactory and disappointing results, depending on used protocols. This review is designed to give an overview of a variety of EGFR-targeting ligands available for nanomedicines, how to conjugate them onto the surface of nanoparticles, and the main analytical methods to confirm this successful conjugation.
Collapse
|
9
|
Nicolson F, Kircher MF. Theranostics: Agents for Diagnosis and Therapy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
10
|
Liu W, Tang Y, Ma H, Li F, Hu Y, Yang Y, Yang J, Liao J, Liu N. Astatine-211 labelled a small molecule peptide: specific cell killing in vitro and targeted therapy in a nude-mouse model. RADIOCHIM ACTA 2020. [DOI: 10.1515/ract-2020-0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Abstract
Extensive interest in the development of α-emitting radionuclides astatine-211 (211At) stems from the potential superiority for the treatment of smaller tumors, disseminated disease, and metastatic disease. VP2, a small molecule fusion peptide, can specifically bind to the VPAC1 receptor which is over-expressed in malignant epithelial tumors. In our recent study, we performed the preparation of 211At labelled VP2 through a one-step method. In this work, we explored the targeted radionuclide therapy with [211At]At-SPC-VP2 in vitro and in vivo. The cytotoxicity and specific cell killing of [211At]At-SPC-VP2 were evaluated using the CCK-8 assay. Compared with the [211At]NaAt, the VPAC1-targeted radionuclide compound [211At]At-SPC-VP2 showed more effective cytotoxicity in vitro. Targeted radioactive therapy trial was carried out in non-small-cell lung cancer (NSCLC) xenograft mice. For the therapy experiment, 4 groups of mice were injected via the tail vein with 370 kBq, 550 kBq, 740 kBq, 3 × ∼246 kBq of [211At]At-SPC-VP2, of which the second and third injections were given 4 and 8 days after the first injection, respectively. As controls, animals were treated with saline or 550 kBq [211At]NaAt. The body weight and tumor size of mice were monitored before the administration and every 2 days thereafter. Cytotoxic radiation of partial tissue samples such as kidneys, liver and stomach of mice were assessed by immunohistochemical examination. The tumor growth was inhibited and significantly improved survival was achieved in mice treated with [211At]At-SPC-VP2, two-fold prolongation of survival compared with the control group, which received normal saline or 550 kBq [211At]NaAt. No renal or hepatic toxicity was observed in the mice receiving [211At]At-SPC-VP2, but gastric pathological sections showed 211At uptake in stomach resulting in later toxicity, highlighting the importance of further enhancing the stability of labelled compounds.
Collapse
Affiliation(s)
- Weihao Liu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Yu Tang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Huan Ma
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Feize Li
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Yingjiang Hu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Yuanyou Yang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Jijun Yang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Jiali Liao
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University , Chengdu , China
| |
Collapse
|
11
|
Kunjiappan S, Pavadai P, Vellaichamy S, Ram Kumar Pandian S, Ravishankar V, Palanisamy P, Govindaraj S, Srinivasan G, Premanand A, Sankaranarayanan M, Theivendren P. Surface receptor‐mediated targeted drug delivery systems for enhanced cancer treatment: A state‐of‐the‐art review. Drug Dev Res 2020; 82:309-340. [DOI: 10.1002/ddr.21758] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Selvaraj Kunjiappan
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy M.S. Ramaiah University of Applied Sciences Bengaluru Karnataka India
| | - Sivakumar Vellaichamy
- Department of Pharmaceutics Arulmigu Kalasalingam College of Pharmacy Krishnankoil Tamilnadu India
| | | | | | - Ponnusamy Palanisamy
- School of Mechanical Engineering Vellore Institute of Technology Vellore Tamilnadu India
| | - Saravanan Govindaraj
- Department of Pharmaceutical Chemistry MNR College of Pharmacy Sangareddy Telangana India
| | - Gowshiki Srinivasan
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | - Adhvitha Premanand
- Department of Biotechnology Kalasalingam Academy of Research and Education Krishnankoil Tamilnadu India
| | | | - Panneerselvam Theivendren
- Department of Pharmaceutical Chemistry Swamy Vivekananda College of Pharmacy Elayampalayam, Namakkal Tamilnadu India
| |
Collapse
|
12
|
Li M, Sagastume EE, Lee D, McAlister D, DeGraffenreid AJ, Olewine KR, Graves S, Copping R, Mirzadeh S, Zimmerman BE, Larsen R, Johnson FL, Schultz MK. 203/212Pb Theranostic Radiopharmaceuticals for Image-guided Radionuclide Therapy for Cancer. Curr Med Chem 2020; 27:7003-7031. [PMID: 32720598 PMCID: PMC10613023 DOI: 10.2174/0929867327999200727190423] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/25/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Receptor-targeted image-guided Radionuclide Therapy (TRT) is increasingly recognized as a promising approach to cancer treatment. In particular, the potential for clinical translation of receptor-targeted alpha-particle therapy is receiving considerable attention as an approach that can improve outcomes for cancer patients. Higher Linear-energy Transfer (LET) of alpha-particles (compared to beta particles) for this purpose results in an increased incidence of double-strand DNA breaks and improved-localized cancer-cell damage. Recent clinical studies provide compelling evidence that alpha-TRT has the potential to deliver a significantly more potent anti-cancer effect compared with beta-TRT. Generator-produced 212Pb (which decays to alpha emitters 212Bi and 212Po) is a particularly promising radionuclide for receptor-targeted alpha-particle therapy. A second attractive feature that distinguishes 212Pb alpha-TRT from other available radionuclides is the possibility to employ elementallymatched isotope 203Pb as an imaging surrogate in place of the therapeutic radionuclide. As direct non-invasive measurement of alpha-particle emissions cannot be conducted using current medical scanner technology, the imaging surrogate allows for a pharmacologically-inactive determination of the pharmacokinetics and biodistribution of TRT candidate ligands in advance of treatment. Thus, elementally-matched 203Pb labeled radiopharmaceuticals can be used to identify patients who may benefit from 212Pb alpha-TRT and apply appropriate dosimetry and treatment planning in advance of the therapy. In this review, we provide a brief history on the use of these isotopes for cancer therapy; describe the decay and chemical characteristics of 203/212Pb for their use in cancer theranostics and methodologies applied for production and purification of these isotopes for radiopharmaceutical production. In addition, a medical physics and dosimetry perspective is provided that highlights the potential of 212Pb for alpha-TRT and the expected safety for 203Pb surrogate imaging. Recent and current preclinical and clinical studies are presented. The sum of the findings herein and observations presented provide evidence that the 203Pb/212Pb theranostic pair has a promising future for use in radiopharmaceutical theranostic therapies for cancer.
Collapse
Affiliation(s)
- Mengshi Li
- Department of Radiology, The University of Iowa, Iowa City, IA USA
- Viewpoint Molecular Targeting, Inc., Coralville, IA USA
| | | | - Dongyoul Lee
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA
| | | | | | | | - Stephen Graves
- Department of Radiology, The University of Iowa, Iowa City, IA USA
| | - Roy Copping
- Oak Ridge National Laboratory, The US Department of Energy, Oak Ridge TN USA
| | - Saed Mirzadeh
- Oak Ridge National Laboratory, The US Department of Energy, Oak Ridge TN USA
| | - Brian E. Zimmerman
- The National Institute of Standards and Technology, Gaithersburg, MD, USA
| | | | - Frances L. Johnson
- Viewpoint Molecular Targeting, Inc., Coralville, IA USA
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa USA
| | - Michael K. Schultz
- Department of Radiology, The University of Iowa, Iowa City, IA USA
- Viewpoint Molecular Targeting, Inc., Coralville, IA USA
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA
- Department of Chemistry, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
13
|
Jeon J. Review of Therapeutic Applications of Radiolabeled Functional Nanomaterials. Int J Mol Sci 2019; 20:E2323. [PMID: 31083402 PMCID: PMC6539387 DOI: 10.3390/ijms20092323] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/10/2023] Open
Abstract
In the last two decades, various nanomaterials have attracted increasing attention in medical science owing to their unique physical and chemical characteristics. Incorporating radionuclides into conventionally used nanomaterials can confer useful additional properties compared to the original material. Therefore, various radionuclides have been used to synthesize functional nanomaterials for biomedical applications. In particular, several α- or β-emitter-labeled organic and inorganic nanoparticles have been extensively investigated for efficient and targeted cancer treatment. This article reviews recent progress in cancer therapy using radiolabeled nanomaterials including inorganic, polymeric, and carbon-based materials and liposomes. We first provide an overview of radiolabeling methods for preparing anticancer agents that have been investigated recently in preclinical studies. Next, we discuss the therapeutic applications and effectiveness of α- or β-emitter-incorporated nanomaterials in animal models and the emerging possibilities of these nanomaterials in cancer therapy.
Collapse
Affiliation(s)
- Jongho Jeon
- Department of Applied Chemistry, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|
14
|
Eslahi N, Shakeri-Zadeh A, Ashtari K, Pirhajati-Mahabadi V, Tohidi Moghadam T, Shabani R, Kamrava K, Madjd Z, Maki C, Asgari HR, Koruji M. In Vitro Cytotoxicity of Folate-Silica-Gold Nanorods on Mouse Acute Lymphoblastic Leukemia and Spermatogonial Cells. CELL JOURNAL 2019; 21:14-26. [PMID: 30507084 PMCID: PMC6275430 DOI: 10.22074/cellj.2019.5691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/27/2018] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The purpose of this study was to evaluate in vitro cytotoxicity of gold nanorods (GNRs) on the viability of spermatogonial cells (SSCs) and mouse acute lymphoblastic leukemia cells (EL4s). MATERIALS AND METHODS In this experimental study, SSCs were isolated from the neonate mice, following enzymatic digestion and differential plating. GNRs were synthesized, then modified by silica and finally conjugated with folic acid to form F-Si-GNRs. Different doses of F-Si-GNRs (25, 50, 75, 100, 125 and 140 μM) were used on SSCs and EL4s. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) proliferation assay was performed to examine the GNRs toxicity. Flow cytometry was used to confirm the identity of the EL4s and SSCs. Also, the identity and functionality of SSCs were determined by the expression of specific spermatogonial genes and transplantation into recipient testes. Apoptosis was determined by flow cytometry using an annexin V/propidium iodide (PI) kit. RESULTS Flow cytometry showed that SSCs and EL4s were positive for Plzf and H-2kb, respectively. The viability percentage of SSCs and EL4s that were treated with 25, 50, 75, 100, 125 and 140 μM of F-Si-GNRs was 65.33 ± 3.51%, 60 ± 3.6%, 51.33 ± 3.51%, 49 ± 3%, 30.66 ± 2.08% and 16.33 ± 2.51% for SSCs and 57.66 ± 0.57%, 54.66 ± 1.5%, 39.66 ± 1.52%, 12.33 ± 2.51%, 10 ± 1% and 5.66 ± 1.15% for EL4s respectively. The results of the MTT assay indicated that 100 μM is the optimal dose to reach the highest and lowest level of cell death in EL4s and in SSCs, respectively. CONCLUSION Cell death increased with increasing concentrations of F-Si-GNRs. Following utilization of F-Si-GNRs, there was a significant difference in the extent of apoptosis between cancer cells and SSCs.
Collapse
Affiliation(s)
- Neda Eslahi
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Shakeri-Zadeh
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Khadijeh Ashtari
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Tahereh Tohidi Moghadam
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ronak Shabani
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Kamran Kamrava
- Clinical Nanomedicine Laboratory, ENT-Head and Neck Research Center, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center and Dep Pathology, Faculty of Medicine Iran University of Medical Sciences, Tehran, Iran
| | - Chad Maki
- VetCell Therapeutics, Daimler St, Santa Ana CA, USA
| | - Hamid Reza Asgari
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran. Electronic Address:
| |
Collapse
|
15
|
Molecular Imaging with 68Ga Radio-Nanomaterials: Shedding Light on Nanoparticles. APPLIED SCIENCES-BASEL 2018. [DOI: 10.3390/app8071098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
16
|
Ji A, Zhang Y, Lv G, Lin J, Qi N, Ji F, Du M. 131
I radiolabeled immune albumin nanospheres loaded with doxorubicin for in vivo combinatorial therapy. J Labelled Comp Radiopharm 2018; 61:362-369. [PMID: 29247459 DOI: 10.1002/jlcr.3593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Anping Ji
- Nuclear Medicine Department; the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine; Nanjing China
| | - Yuanchao Zhang
- Nuclear Medicine Department; the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine; Nanjing China
| | - Gaochao Lv
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine; Jiangsu Institute of Nuclear Medicine; Wuxi China
| | - Jianguo Lin
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine; Jiangsu Institute of Nuclear Medicine; Wuxi China
| | - Ning Qi
- College of Clinical Medicine; Southeast University; Nanjing China
| | - Faquan Ji
- Nuclear Medicine Department; the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine; Nanjing China
| | - Minghua Du
- Nuclear Medicine Department; the Affiliated Hospital of Nanjing University of Traditional Chinese Medicine; Nanjing China
| |
Collapse
|
17
|
Li M, Zhang X, Quinn TP, Lee D, Liu D, Kunkel F, Zimmerman BE, McAlister D, Olewein K, Menda Y, Mirzadeh S, Copping R, Johnson FL, Schultz MK. Automated cassette-based production of high specific activity [ 203/212Pb]peptide-based theranostic radiopharmaceuticals for image-guided radionuclide therapy for cancer. Appl Radiat Isot 2017; 127:52-60. [PMID: 28521118 PMCID: PMC6295910 DOI: 10.1016/j.apradiso.2017.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/29/2017] [Accepted: 05/06/2017] [Indexed: 01/28/2023]
Abstract
A method for preparation of Pb-212 and Pb-203 labeled chelator-modified peptide-based radiopharmaceuticals for cancer imaging and radionuclide therapy has been developed and adapted for automated clinical production. Pre-concentration and isolation of radioactive Pb2+ from interfering metals in dilute hydrochloric acid was optimized using a commercially-available Pb-specific chromatography resin packed in disposable plastic columns. The pre-concentrated radioactive Pb2+ is eluted in NaOAc buffer directly to the reaction vessel containing chelator-modified peptides. Radiolabeling was found to proceed efficiently at 85°C (45min; pH 5.5). The specific activity of radiolabeled conjugates was optimized by separation of radiolabeled conjugates from unlabeled peptide via HPLC. Preservation of bioactivity was confirmed by in vivo biodistribution of Pb-203 and Pb-212 labeled peptides in melanoma-tumor-bearing mice. The approach has been found to be robustly adaptable to automation and a cassette-based fluid-handling system (Modular Lab Pharm Tracer) has been customized for clinical radiopharmaceutical production. Our findings demonstrate that the Pb-203/Pb-212 combination is a promising elementally-matched radionuclide pair for image-guided radionuclide therapy for melanoma, neuroendocrine tumors, and potentially other cancers.
Collapse
Affiliation(s)
- Mengshi Li
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA.
| | - Xiuli Zhang
- Department of Biochemistry, University of Missouri, Columbia, MO USA
| | - Thomas P Quinn
- Department of Biochemistry, University of Missouri, Columbia, MO USA
| | - Dongyoul Lee
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA
| | - Dijie Liu
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Falk Kunkel
- Eckert & Ziegler Radiopharma GmbH, Berlin, Germany
| | - Brian E Zimmerman
- National Institute of Standards and Technology, Gaithersburg, MD, USA
| | | | | | - Yusuf Menda
- Department of Radiology, The University of Iowa, Iowa City, IA, USA
| | - Saed Mirzadeh
- Oak Ridge National Laboratory, The US Department of Energy, Oak Ridge, TN, USA
| | - Roy Copping
- Oak Ridge National Laboratory, The US Department of Energy, Oak Ridge, TN, USA
| | - Frances L Johnson
- Viewpoint Molecular Targeting, LLC, Coralville, IA, USA; Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Michael K Schultz
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA, USA; Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA; Department of Radiology, The University of Iowa, Iowa City, IA, USA; Viewpoint Molecular Targeting, LLC, Coralville, IA, USA; Department of Radiation Oncology (Free Radical and Radiation Biology Program), Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Chemistry, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
18
|
Antitumor Effect of Nanoparticle 131I-Labeled Arginine-Glycine-Aspartate–Bovine Serum Albumin–Polycaprolactone in Lung Cancer. AJR Am J Roentgenol 2017; 208:1116-1126. [PMID: 28301223 DOI: 10.2214/ajr.16.16947] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
19
|
Sedlacek O, Kucka J, Monnery BD, Slouf M, Vetrik M, Hoogenboom R, Hruby M. The effect of ionizing radiation on biocompatible polymers: From sterilization to radiolysis and hydrogel formation. Polym Degrad Stab 2017. [DOI: 10.1016/j.polymdegradstab.2017.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
20
|
Samadian H, Hosseini-Nami S, Kamrava SK, Ghaznavi H, Shakeri-Zadeh A. Folate-conjugated gold nanoparticle as a new nanoplatform for targeted cancer therapy. J Cancer Res Clin Oncol 2016; 142:2217-29. [PMID: 27209529 DOI: 10.1007/s00432-016-2179-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/09/2016] [Indexed: 01/17/2023]
Abstract
Conventional cancer treatment methods suffer from many limitations such as non-specificity and low efficacy in discrimination between healthy and cancer cells. Recent developments in nanotechnology have introduced novel and smart therapeutic nanomaterials that basically take advantage of various targeting approaches. Targeted nanomaterials selectively bind to the cancer cells and affect them with minor effects on healthy cells. Folic acid (folate) is an essential molecule in DNA synthesis pathway which is highly needed for cancer cell duplication. Some certain cancer cells overexpress folate receptors higher than normal cells, and this fact is the basis of folate targeting strategy. There are many publications reporting various folate conjugated nanomaterials among which folate-conjugated gold nanoparticles hold great promises in targeted cancer therapy. Gold nanoparticles have been identified as promising candidates for new cancer therapy modalities because of biocompatibility, easy synthesis and functionalization, chemo-physical stability, and optical tunable characteristics. In the last decade, there has been a significant explosion in gold nanoparticles research, with a rapid increase in publications related to the area of biomedicine. Although there are many reports published on "gold nanoparticles" and "folate targeting," there are a few reports on "folate-conjugated gold nanoparticles" in biomedical literature. This paper intends to review and illustrate the recent advances in biomedicine which have been designed on the basis of folate-conjugated gold nanoparticles.
Collapse
Affiliation(s)
- Hadi Samadian
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Samira Hosseini-Nami
- Clinical Nanomedicine Laboratory, ENT and Head and Neck Surgery Research Center, Rasoul Akram Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Seyed Kamran Kamrava
- Clinical Nanomedicine Laboratory, ENT and Head and Neck Surgery Research Center, Rasoul Akram Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Habib Ghaznavi
- Zahedan University of Medical Sciences (ZaUMS), Zahedan, Iran.
| | - Ali Shakeri-Zadeh
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Diagnostic Nanoparticles Research Core, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
21
|
Liu X, Song M, Gao Z, Cai X, Dixon W, Chen X, Cao Y, Xiao H. Stereoisomers of Astaxanthin Inhibit Human Colon Cancer Cell Growth by Inducing G2/M Cell Cycle Arrest and Apoptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:7750-7759. [PMID: 27726394 DOI: 10.1021/acs.jafc.6b03636] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Astaxanthin (AST) is a xanthophyll carotenoid with potential protective effects against carcinogenesis. Different stereoisomers of AST (ASTs) exist in a variety of food sources. Due to limited information on the bioactivities of ASTs, the present study investigated the inhibitory effects of ASTs on HCT116 and HT29 human colon cancer cells. ASTs investigated herein included 3S,3'S (S) from Haematococcus pluvialis, 3R,3'R (R) from Phaffia rhodozyma, and a statistical mixture (S: meso: R = 1:2:1) (M) from synthetic AST. Cell viability assay showed that ASTs all inhibited colon cancer cell growth in a time-dependent (24-72 h) and dose-dependent (4-16 μM) manner, and there was no significant difference among the IC50 values of ASTs (p > 0.05). Flow cytometry analysis indicated that ASTs induced G2/M cell cycle arrest and cellular apoptosis in cancer cells. The cell cycle arrest caused by ASTs was associated with increases in the expression levels of p21Cip1/Waf1, p27, and p53, as well as decreases in the levels of CDK4 and CDK6. Meanwhile, the apoptosis induced by ASTs was confirmed by activation of caspase-3 and PARP in the cancer cells. The results indicated that hydroxyl (OH) at C3 and C3' of terminal ring structure might not be the major factor that affects the anticancer activity of AST. This study revealed important information on the inhibitory effects of ASTs on human colon cancer cells, which provided a basis for using ASTs as chemopreventive agents for colon cancer.
Collapse
Affiliation(s)
- Xiaojuan Liu
- Department of Food Science, College of Food Science, South China Agricultural University , Guangzhou 510642, China
- Department of Food Science, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Mingyue Song
- Department of Food Science, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Zili Gao
- Department of Food Science, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Xiaokun Cai
- Department of Food Science, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - William Dixon
- Department of Food Science, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Xiaofeng Chen
- Department of Food Science, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Yong Cao
- Department of Food Science, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| |
Collapse
|
22
|
Lin M, Huang J, Jiang X, Zhang J, Yu H, Ye J, Zhang D. A combination hepatoma-targeted therapy based on nanotechnology: pHRE-Egr1-HSV-TK/(131)I-antiAFPMcAb-GCV/MFH. Sci Rep 2016; 6:33524. [PMID: 27642033 PMCID: PMC5027595 DOI: 10.1038/srep33524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/26/2016] [Indexed: 12/13/2022] Open
Abstract
Combination targeted therapy is a promising cancer therapeutic strategy. Here, using PEI-Mn0.5Zn0.5Fe2O4 nanoparticles (PEI-MZF-NPs) as magnetic media for MFH (magnetic fluid hyperthermia) and gene transfer vector for gene-therapy, a combined therapy, pHRE-Egr1-HSV-TK/(131)I-antiAFPMcAb-GCV/MFH, for hepatoma is developed. AntiAFPMcAb (Monoclonal antibody AFP) is exploited for targeting. The plasmids pHRE-Egr1-HSV-TK are achieved by incorporation of pEgr1-HSV-TK and pHRE-Egr1-EGFP. Restriction enzyme digestion and PCR confirm the recombinant plasmids pHRE-Egr1-HSV-TK are successfully constructed. After exposure to the magnetic field, PEI-MZF-NPs/pHRE-Egr1-EGFP fluid is warmed rapidly and then the temperature is maintained at 43 °C or so, which is quite appropriate for cancer treatment. The gene expression reaches the peak when treated with 200 μCi (131)I for 24 hours, indicating that the dose of 200 μCi might be the optimal dose for irradiation and 24 h irradiation later is the best time to initiate MFH. The in vitro and in vivo experiments demonstrate that pHRE-Egr1-HSV-TK/(131)I-antiAFPMcAb-GCV/MFH can greatly suppress hepatic tumor cell proliferation and induce cell apoptosis and necrosis and effectively inhibit the tumor growth, much better than any monotherapy does alone. Furthermore, the combination therapy has few or no adverse effects. It might be applicable as a strategy to treat hepatic cancer.
Collapse
Affiliation(s)
- Mei Lin
- Taizhou People’s Hospital Affiliated to Nantong University, Taizhou, 225300, China
- Medical School of Southeast University, Nanjing, 210009, China
| | - Junxing Huang
- Taizhou People’s Hospital Affiliated to Nantong University, Taizhou, 225300, China
| | - Xingmao Jiang
- Key Laboratory of Advanced Catalytic Material and Technology, Changzhou University, Changzhou, 213000, China
| | - Jia Zhang
- Medical School of Southeast University, Nanjing, 210009, China
| | - Hong Yu
- Taizhou People’s Hospital Affiliated to Nantong University, Taizhou, 225300, China
| | - Jun Ye
- Taizhou People’s Hospital Affiliated to Nantong University, Taizhou, 225300, China
| | - Dongsheng Zhang
- Medical School of Southeast University, Nanjing, 210009, China
- Southeast University, Jiangsu Key Laboratory For Biomaterials and Devices, Nanjing, 210009, China
| |
Collapse
|
23
|
Feiner RC, Müller KM. Recent progress in protein-protein interaction study for EGFR-targeted therapeutics. Expert Rev Proteomics 2016; 13:817-32. [PMID: 27424502 DOI: 10.1080/14789450.2016.1212665] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Epidermal growth factor receptor (EGFR) expression is upregulated in many tumors and its aberrant signaling drives progression of many cancer types. Consequently, EGFR has become a clinically validated target as extracellular tumor marker for antibodies as well as for tyrosine kinase inhibitors. Within the last years, new mechanistic insights were uncovered and, based on clinical experience as well as progress in protein engineering, novel bio-therapeutic approaches were developed and tested. AREAS COVERED The potential therapeutic targeting arsenal in the fight against cancer now encompasses bispecific or biparatopic antibodies, DARPins, Adnectins, Affibodies, peptides and combinations of these binding molecules with viral- and nano-particles. We review past and recent binding proteins from the literature and include a brief description of the various targeting approaches. Special attention is given to the binding modes with the EGFR. Expert commentary: Clinical data from the three approved anti EGFR antibodies indicate that there is room for improved therapeutic efficacy. Having choices in size, affinity, avidity and the mode of EGFR binding as well as the possibility to combine various effector functions opens the possibility to rationally design more effective therapeutics.
Collapse
Affiliation(s)
- Rebecca Christine Feiner
- a Cellular and Molecular Biotechnology group, Faculty of Technology , Bielefeld University , Bielefeld , Germany
| | - Kristian Mark Müller
- a Cellular and Molecular Biotechnology group, Faculty of Technology , Bielefeld University , Bielefeld , Germany
| |
Collapse
|