1
|
Kwon MR, Park JS, Ko EJ, Park J, Ju EJ, Shin SH, Son GW, Lee HW, Park YY, Kang MH, Kim YJ, Kim BM, Lee HJ, Kim TW, Kim CJ, Song SY, Park SS, Jeong SY. Ibulocydine Inhibits Migration and Invasion of TNBC Cells via MMP-9 Regulation. Int J Mol Sci 2024; 25:6123. [PMID: 38892310 PMCID: PMC11173234 DOI: 10.3390/ijms25116123] [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: 04/25/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 15-20% of all breast cancer types, indicating a poor survival prognosis with a more aggressive biology of metastasis to the lung and a short response duration to available therapies. Ibulocydine (IB) is a novel (cyclin-dependent kinase) CDK7/9 inhibitor prodrug displaying potent anti-cancer effects against various cancer cell types. We performed in vitro and in vivo experiments to determine whether IB inhibits metastasis and eventually overcomes the poor drug response in TNBC. The result showed that IB inhibited the growth of TNBC cells by inducing caspase-mediated apoptosis and blocking metastasis by reducing MMP-9 expression in vitro. Concurrently, in vivo experiments using the metastasis model showed that IB inhibited metastasis of MDA-MB-231-Luc cells to the lung. Collectively, these results demonstrate that IB inhibited the growth of TNBC cells and blocked metastasis by regulating MMP-9 expression, suggesting a novel therapeutic agent for metastatic TNBC.
Collapse
Affiliation(s)
- Mi-Ri Kwon
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Ji-Soo Park
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Jung Ko
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Jin Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Eun-Jin Ju
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Seol-Hwa Shin
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Ga-Won Son
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Hye-Won Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Yun-Yong Park
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Myoung-Hee Kang
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Yeon-Joo Kim
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Byeong-Moon Kim
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee-Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Tae-Won Kim
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Chong-Jai Kim
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Si-Yeol Song
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Seok-Soon Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Seong-Yun Jeong
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul 05505, Republic of Korea
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| |
Collapse
|
2
|
Digby EM, Tung MT, Kagalwala HN, Ryan LS, Lippert AR, Beharry AA. Dark Dynamic Therapy: Photosensitization without Light Excitation Using Chemiluminescence Resonance Energy Transfer in a Dioxetane-Erythrosin B Conjugate. ACS Chem Biol 2022; 17:1082-1091. [PMID: 35394740 DOI: 10.1021/acschembio.1c00925] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Reactive oxygen species (e.g., singlet oxygen) are the primary cytotoxic agents used in the clinically approved technique photodynamic therapy (PDT). Although singlet oxygen has high potential to effectively kill tumor cells, its production via light excitation of a photosensitizer has been limited by the penetration depth and delivery of light in tissue. To produce singlet oxygen without light excitation, we describe the use of Schaap's chemiluminescent scaffold comprising an adamantylidene-dioxetane motif. Functionalizing this scaffold with a photosensitizer, Erythrosin B, resulted in spontaneous chemiluminescence resonance energy transfer (CRET) leading to the production of singlet oxygen. We show that this compound is cell permeable and that the singlet oxygen produced via CRET is remarkably efficient in killing cancer cells at low micromolar concentrations. Moreover, we demonstrate that protection of the phenol on the chemiluminescent scaffold with a nitroreductase-responsive trigger group allows for cancer-selective dark dynamic cell death. Here, we present the concept of dark dynamic therapy using a small cell-permeable molecule capable of producing the effects of PDT in cells, without light.
Collapse
Affiliation(s)
- Elyse M. Digby
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, Ontario L5L 1C6, Canada
| | - Matthew T. Tung
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, Ontario L5L 1C6, Canada
| | - Husain N. Kagalwala
- Department of Chemistry, Center for Drug Discovery, Design, and Delivery (CD4), and Center for Global Health Impact (CGHI), Southern Methodist University, Dallas, Texas 75205-0314, United States
| | - Lucas S. Ryan
- Department of Chemistry, Center for Drug Discovery, Design, and Delivery (CD4), and Center for Global Health Impact (CGHI), Southern Methodist University, Dallas, Texas 75205-0314, United States
| | - Alexander R. Lippert
- Department of Chemistry, Center for Drug Discovery, Design, and Delivery (CD4), and Center for Global Health Impact (CGHI), Southern Methodist University, Dallas, Texas 75205-0314, United States
| | - Andrew A. Beharry
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, Ontario L5L 1C6, Canada
| |
Collapse
|
3
|
Fuchigami T, Haywood T, Gowrishankar G, Anders D, Namavari M, Wardak M, Gambhir SS. Synthesis and Characterization of 9-(4-[ 18F]Fluoro-3-(hydroxymethyl)butyl)-2-(phenylthio)-6-oxopurine as a Novel PET Agent for Mutant Herpes Simplex Virus Type 1 Thymidine Kinase Reporter Gene Imaging. Mol Imaging Biol 2021; 22:1151-1160. [PMID: 32691392 DOI: 10.1007/s11307-020-01517-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE [18F]FHBG has been used as a positron emission tomography (PET) imaging tracer for the monitoring of herpes simplex virus type 1 thymidine kinase (HSV1-tk), a reporter gene for cell and gene therapy in humans. However, this tracer shows inadequate blood-brain barrier (BBB) penetration and, therefore, would be limited for accurate quantification of reporter gene expression in the brain. Here, we report the synthesis and evaluation of 9-(4-[18F]fluoro-3-(hydroxymethyl)butyl)-2(phenylthio)-6-oxopurine ([18F]FHBT) as a new PET tracer for imaging reporter gene expression of HSV1-tk and its mutant HSV1-sr39tk, with the aim of improved BBB penetration. PROCEDURES [18F]FHBT was prepared by using a tosylate precursor and [18F]KF. The cellular uptake of [18F]FHBT was performed in HSV1-sr39tk-positive (+) or HSV1-sr39tk-negative (-) MDA-MB-231 breast cancer cells. The specificity of [18F]FHBT to assess HSV1-sr39tk expression was evaluated by in vitro blocking studies using 1 mM of ganciclovir (GCV). Penetration of [18F]FHBT and [18F]FHBG across the BBB was assessed by dynamic PET imaging studies in normal mice. RESULTS The tosylate precursor reacted with [18F]KF using Kryptofix2.2.2 followed by deprotection to give [18F]FHBT in 10 % radiochemical yield (decay-corrected). The uptake of [18F]FHBT in HSV1-sr39tk (+) cells was significantly higher than that of HSV1-sr39tk (-) cells. In the presence of GCV (1 mM), the uptake of [18F]FHBT was significantly decreased, indicating that [18F]FHBT serves as a selective substrate of HSV1-sr39TK. PET images and time-activity curves of [18F]FHBT in the brain regions showed similar initial brain uptakes (~ 12.75 min) as [18F]FHBG (P > 0.855). Slower washout of [18F]FHBT was observed at the later time points (17.75 - 57.75 min, P > 0.207). CONCLUSIONS Although [18F]FHBT showed no statistically significant improvement of BBB permeability compared with [18F]FHBG, we have demonstrated that the 2-(phenylthio)-6-oxopurine backbone can serve as a novel scaffold for developing HSV1-tk/HSV1-sr39tk reporter gene imaging agents for additional research in the future.
Collapse
Affiliation(s)
- Takeshi Fuchigami
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, 852-8521, Japan.,Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 318 Campus Drive, Room E150A, Stanford, CA, 94305, USA
| | - Tom Haywood
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 318 Campus Drive, Room E150A, Stanford, CA, 94305, USA
| | - Gayatri Gowrishankar
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 318 Campus Drive, Room E150A, Stanford, CA, 94305, USA
| | - David Anders
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 318 Campus Drive, Room E150A, Stanford, CA, 94305, USA
| | - Mohammad Namavari
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 318 Campus Drive, Room E150A, Stanford, CA, 94305, USA
| | - Mirwais Wardak
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 318 Campus Drive, Room E150A, Stanford, CA, 94305, USA
| | - Sanjiv Sam Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 318 Campus Drive, Room E150A, Stanford, CA, 94305, USA. .,Department of Bioengineering and Materials Science & Engineering, Bio-X Program, Stanford University, 318 Campus Dr., Room E150 Stanford, Stanford, CA, 94305, USA.
| |
Collapse
|
4
|
Kumar SU, Wang H, Telichko AV, Natarajan A, Bettinger T, Cherkaoui S, Massoud TF, Dahl JJ, Paulmurugan R. Ultrasound Triggered Co‐Delivery of Therapeutic MicroRNAs and a Triple Suicide Gene Therapy Vector by Using Biocompatible Polymer Nanoparticles for Improved Cancer Therapy in Mouse Models. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sukumar Uday Kumar
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
| | - Huaijun Wang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
| | - Arsenii V. Telichko
- Canary Center for Cancer Early Detection, Department of Radiology Stanford University Stanford CA 94305 USA
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
| | - Thierry Bettinger
- Novel Agents Department Bracco Suisse SA Route de la Galaise 31 1228 Plan‐les‐Ouates Geneva Switzerland
| | - Samir Cherkaoui
- Novel Agents Department Bracco Suisse SA Route de la Galaise 31 1228 Plan‐les‐Ouates Geneva Switzerland
| | - Tarik F. Massoud
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
| | - Jeremy J. Dahl
- Canary Center for Cancer Early Detection, Department of Radiology Stanford University Stanford CA 94305 USA
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology Stanford University Stanford CA 94305 USA
- Canary Center for Cancer Early Detection, Department of Radiology Stanford University Stanford CA 94305 USA
| |
Collapse
|
5
|
Ruiz de Garibay G, García de Jalón E, Stigen E, Lund KB, Popa M, Davidson B, Safont MM, Rygh CB, Espedal H, Barrett TM, Haug BE, McCormack E. Repurposing 18F-FMISO as a PET tracer for translational imaging of nitroreductase-based gene directed enzyme prodrug therapy. Am J Cancer Res 2021; 11:6044-6057. [PMID: 33897898 PMCID: PMC8058731 DOI: 10.7150/thno.55092] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/25/2021] [Indexed: 12/25/2022] Open
Abstract
Nitroreductases (NTR) are a family of bacterial enzymes used in gene directed enzyme prodrug therapy (GDEPT) that selectively activate prodrugs containing aromatic nitro groups to exert cytotoxic effects following gene transduction in tumours. The clinical development of NTR-based GDEPT has, in part, been hampered by the lack of translational imaging modalities to assess gene transduction and drug cytotoxicity, non-invasively. This study presents translational preclinical PET imaging to validate and report NTR activity using the clinically approved radiotracer, 18F-FMISO, as substrate for the NTR enzyme. Methods: The efficacy with which 18F-FMISO could be used to report NfsB NTR activity in vivo was investigated using the MDA-MB-231 mammary carcinoma xenograft model. For validation, subcutaneous xenografts of cells constitutively expressing NTR were imaged using 18F-FMISO PET/CT and fluorescence imaging with CytoCy5S, a validated fluorescent NTR substrate. Further, examination of the non-invasive functionality of 18F-FMISO PET/CT in reporting NfsB NTR activity in vivo was assessed in metastatic orthotopic NfsB NTR expressing xenografts and metastasis confirmed by bioluminescence imaging. 18F-FMISO biodistribution was acquired ex vivo by an automatic gamma counter measuring radiotracer retention to confirm in vivo results. To assess the functional imaging of NTR-based GDEPT with 18F-FMISO, PET/CT was performed to assess both gene transduction and cytotoxicity effects of prodrug therapy (CB1954) in subcutaneous models. Results:18F-FMISO retention was detected in NTR+ subcutaneous xenografts, displaying significantly higher PET contrast than NTR- xenografts (p < 0.0001). Substantial 18F-FMISO retention was evident in metastases of orthotopic xenografts (p < 0.05). Accordingly, higher 18F-FMISO biodistribution was prevalent ex vivo in NTR+ xenografts. 18F-FMISO NfsB NTR PET/CT imaging proved useful for monitoring in vivo NTR transduction and the cytotoxic effect of prodrug therapy. Conclusions:18F-FMISO NfsB NTR PET/CT imaging offered significant contrast between NTR+ and NTR- tumours and effective resolution of metastatic progression. Furthermore, 18F-FMISO NfsB NTR PET/CT imaging proved efficient in monitoring the two steps of GDEPT, in vivo NfsB NTR transduction and response to CB1954 prodrug therapy. These results support the repurposing of 18F-FMISO as a readily implementable PET imaging probe to be employed as companion diagnostic test for NTR-based GDEPT systems.
Collapse
|
6
|
Pandurangi RS, Tomasetti M, Verapazham ST, Paulmurugan R, Ma C, Rajput S, Anjanappa M, Nakshatri H. A Priori Activation of Apoptosis Pathways of Tumor (AAAPT) technology: Development of targeted apoptosis initiators for cancer treatment. PLoS One 2021; 16:e0225869. [PMID: 33556062 PMCID: PMC7870153 DOI: 10.1371/journal.pone.0225869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/18/2020] [Indexed: 11/24/2022] Open
Abstract
Cancer cells develop tactics to circumvent the interventions by desensitizing themselves to interventions. Amongst many, the principle routes of desensitization include a) activation of survival pathways (e.g. NF-kB, PARP) and b) downregulation of cell death pathways (e.g. CD95/CD95L). As a result, it requires high therapeutic dose to achieve tumor regression which, in turn damages normal cells through the collateral effects. Methods are needed to sensitize the low and non-responsive resistant tumor cells including cancer stem cells (CSCs) in order to evoke a better response from the current treatments. Current treatments including chemotherapy can induce cell death only in bulk cancer cells sparing CSCs and cancer resistant cells (CRCs) which are shown to be responsible for high recurrence of disease and low patient survival. Here, we report several novel tumor targeted sensitizers derived from the natural Vitamin E analogue (AMP-001-003). The drug design is based on a novel concept "A priori activation of apoptosis pathways of tumor technology (AAAPT) which is designed to activate specific cell death pathways and inhibit survival pathways simultaneously and selectively in cancer cells sparing normal cells. Our results indicate that AMP-001-003 sensitize various types of cancer cells including MDA-MB-231 (triple negative breast cancer), PC3 (prostate cancer) and A543 (lung cancer) cells resulting in reducing the IC-50 of doxorubicin in vitro when used as a combination. At higher doses, AMP-001 acts as an anti-tumor agent on its own. The synergy between AMP-001 and doxorubicin could pave a new pathway to use AAAPT leading molecules as neoadjuvant to chemotherapy to achieve better efficacy and reduced off-target toxicity compared to the current treatments.
Collapse
Affiliation(s)
- Raghu S. Pandurangi
- Sci-Engi-Medco Solutions Inc., St Charles, Missouri, United States of America
| | - Marco Tomasetti
- Department of Clinical and Molecular Sciences, Section of Experimental and Occupational Medicine, Polytechnic University of Marche, Ancona, Italy
| | - Sekar T. Verapazham
- Molecular Imaging Program at Stanford (MIPS), Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Canary Center for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Cynthia Ma
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sandeep Rajput
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Manjushree Anjanappa
- Department Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Harikrishna Nakshatri
- Department Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
7
|
Molecular Imaging of Gene Therapy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
8
|
Pan CT, Chang WH, Kumar A, Singh SP, Kaushik AC, Sharma J, Long ZJ, Wen ZH, Mishra SK, Yen CK, Chaudhary RK, Shiue YL. Nanoparticles-mediated Brain Imaging and Disease Prognosis by Conventional as well as Modern Modal Imaging Techniques: a Comparison. Curr Pharm Des 2020; 25:2637-2649. [PMID: 31603057 DOI: 10.2174/1381612825666190709220139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Multimodal imaging plays an important role in the diagnosis of brain disorders. Neurological disorders need to be diagnosed at an early stage for their effective treatment as later, it is very difficult to treat them. If possible, diagnosing at an early stage can be much helpful in curing the disease with less harm to the body. There is a need for advanced and multimodal imaging techniques for the same. This paper provides an overview of conventional as well as modern imaging techniques for brain diseases, specifically for tumor imaging. In this paper, different imaging modalities are discussed for tumor detection in the brain along with their advantages and disadvantages. Conjugation of two and more than two modalities provides more accurate information rather than a single modality. They can monitor and differentiate the cellular processes of normal and diseased condition with more clarity. The advent of molecular imaging, including reporter gene imaging, has opened the door of more advanced noninvasive detection of brain tumors. Due to specific optical properties, semiconducting polymer-based nanoparticles also play a pivotal role in imaging tumors. OBJECTIVE The objective of this paper is to review nanoparticles-mediated brain imaging and disease prognosis by conventional as well as modern modal imaging techniques. CONCLUSION We reviewed in detail various medical imaging techniques. This paper covers recent developments in detail and elaborates a possible research aspect for the readers in the field.
Collapse
Affiliation(s)
- Cheng-Tang Pan
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan.,Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
| | - Wei-Hsi Chang
- Department of Emergency Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Ajay Kumar
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
| | - Satya P Singh
- School of EEE, Nanyang Technological University, Nanyang Ave, Singapore
| | - Aman Chandra Kaushik
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, ShanghaiJia Tong University, Shanghai 200240, China
| | - Jyotsna Sharma
- Amity School of Applied Sciences, Amity University Haryana, Gurugram-122413, Manesai, Panchgaon, Haryana, India
| | - Zheng-Jing Long
- Department of Emergency Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Sunil Kumar Mishra
- Patronage Institute of Management Studies, Greater Noida, Uttar Pradesh, India
| | - Chung-Kun Yen
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
| | - Ravi Kumar Chaudhary
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pardesh, India, India
| | - Yow-Ling Shiue
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
| |
Collapse
|
9
|
Sukumar UK, Rajendran JCB, Gambhir SS, Massoud TF, Paulmurugan R. SP94-Targeted Triblock Copolymer Nanoparticle Delivers Thymidine Kinase-p53-Nitroreductase Triple Therapeutic Gene and Restores Anticancer Function against Hepatocellular Carcinoma in Vivo. ACS APPLIED MATERIALS & INTERFACES 2020; 12:11307-11319. [PMID: 32048820 PMCID: PMC7997290 DOI: 10.1021/acsami.9b20071] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Gene-directed enzyme-prodrug therapy (GDEPT) is a promising approach for cancer therapy, but it suffers from poor targeted delivery in vivo. Polyethylenimine (PEI) is a cationic polymer efficient in delivering negatively charged nucleic acids across cell membranes; however, it is highly toxic in vivo. Hence, we efficiently reduced PEI toxicity without compromising its transfection efficiency by conjugating it with poly(d,l-lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) as triblock copolymers through a multistep synthetic process. The synthesized nanoparticles showed efficient delivery of loaded nucleic acids to tumor cells in vitro and in vivo in mice. We used this nanoparticle to deliver a rationally engineered thymidine kinase (TK)-p53-nitroreductase (NTR) triple therapeutic gene against hepatocellular carcinoma (HCC), where p53 tumor suppressor gene is mutated in more than 85% of cancers. TK-p53-NTR triple gene therapy restores p53 function and potentiates cancer cell response to delivered prodrugs (ganciclovir (GCV) and CB1954). We used SP94 peptide-functionalized PLGA-PEG-PEI nanoparticles for the optimal delivery of TK-p53-NTR therapeutic gene in vivo. The nanoparticles prepared from the conjugated polymer showed high loading efficiency for the DNA and markedly enhanced TK-NTR-mediated gene therapy upon the simultaneous coexpression of p53 by the concurrent rescue of the endogenous apoptotic pathway in HCC cells of both p53-mutant and wild-type phenotypes in vitro. In vivo delivery of TK-p53-NTR genes by SP94-targeted PLGA-PEG-PEI NP in mice resulted in a strong expression of suicide genes selectively in tumors, and subsequent administration of GCV and CB1954 led to a decline in tumor growth, and established a superior therapeutic outcome against HCC. We demonstrate a highly efficient approach that exogenously supplements p53 to enable synergy with the outcome of TK-NTR suicide gene therapy against HCC.
Collapse
Affiliation(s)
- Uday K Sukumar
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, California 94305, United States
| | - Jagadesh Chandra Bose Rajendran
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, California 94305, United States
| | - Sanjiv S Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, California 94305, United States
| | - Tarik F Massoud
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, California 94305, United States
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, California 94305, United States
| |
Collapse
|
10
|
Kanada M, Kim BD, Hardy JW, Ronald JA, Bachmann MH, Bernard MP, Perez GI, Zarea AA, Ge TJ, Withrow A, Ibrahim SA, Toomajian V, Gambhir SS, Paulmurugan R, Contag CH. Microvesicle-Mediated Delivery of Minicircle DNA Results in Effective Gene-Directed Enzyme Prodrug Cancer Therapy. Mol Cancer Ther 2019; 18:2331-2342. [PMID: 31451563 DOI: 10.1158/1535-7163.mct-19-0299] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/13/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022]
Abstract
An emerging approach for cancer treatment employs the use of extracellular vesicles, specifically exosomes and microvesicles, as delivery vehicles. We previously demonstrated that microvesicles can functionally deliver plasmid DNA to cells and showed that plasmid size and sequence, in part, determine the delivery efficiency. In this study, delivery vehicles comprised of microvesicles loaded with engineered minicircle (MC) DNA that encodes prodrug converting enzymes developed as a cancer therapy in mammary carcinoma models. We demonstrated that MCs can be loaded into shed microvesicles with greater efficiency than their parental plasmid counterparts and that microvesicle-mediated MC delivery led to significantly higher and more prolonged transgene expression in recipient cells than microvesicles loaded with the parental plasmid. Microvesicles loaded with MCs encoding a thymidine kinase (TK)/nitroreductase (NTR) fusion protein produced prolonged TK-NTR expression in mammary carcinoma cells. In vivo delivery of TK-NTR and administration of prodrugs led to the effective killing of both targeted cells and surrounding tumor cells via TK-NTR-mediated conversion of codelivered prodrugs into active cytotoxic agents. In vivo evaluation of the bystander effect in mouse models demonstrated that for effective therapy, at least 1% of tumor cells need to be delivered with TK-NTR-encoding MCs. These results suggest that MC delivery via microvesicles can mediate gene transfer to an extent that enables effective prodrug conversion and tumor cell death such that it comprises a promising approach to cancer therapy.
Collapse
Affiliation(s)
- Masamitsu Kanada
- Department of Pediatrics, Stanford University, Stanford, California. .,Department of Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan.,Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan
| | - Bryan D Kim
- Deptartment of Chemistry, University of California, Santa Cruz, California
| | - Jonathan W Hardy
- Department of Pediatrics, Stanford University, Stanford, California.,Department of Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan.,Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, Michigan
| | - John A Ronald
- Department of Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Department of Radiology, Stanford University, Stanford, California.,Robarts Research Institute, Western University, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - Michael H Bachmann
- Department of Pediatrics, Stanford University, Stanford, California.,Department of Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan.,Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, Michigan
| | - Matthew P Bernard
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan.,Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan
| | - Gloria I Perez
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan
| | - Ahmed A Zarea
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan
| | - T Jessie Ge
- Department of Radiology, Stanford University, Stanford, California
| | - Alicia Withrow
- Center for Advanced Microscopy, Michigan State University, East Lansing, Michigan
| | - Sherif A Ibrahim
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan.,Deptartment of Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Victoria Toomajian
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan.,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan
| | - Sanjiv S Gambhir
- Department of Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Department of Radiology, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California.,Department of Materials Science, Stanford University, Stanford, California
| | - Ramasamy Paulmurugan
- Department of Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California. .,Department of Radiology, Stanford University, Stanford, California
| | - Christopher H Contag
- Department of Pediatrics, Stanford University, Stanford, California. .,Department of Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan.,Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, Michigan.,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan
| |
Collapse
|
11
|
Devulapally R, Lee T, Barghava-Shah A, Sekar TV, Foygel K, Bachawal SV, Willmann JK, Paulmurugan R. Ultrasound-guided delivery of thymidine kinase-nitroreductase dual therapeutic genes by PEGylated-PLGA/PIE nanoparticles for enhanced triple negative breast cancer therapy. Nanomedicine (Lond) 2018; 13:1051-1066. [PMID: 29790803 PMCID: PMC6219432 DOI: 10.2217/nnm-2017-0328] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 03/06/2018] [Indexed: 11/21/2022] Open
Abstract
AIM Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype. Since no targeted therapy is available, gene-directed enzyme prodrug therapy (GDEPT) could be an attractive strategy for treating TNBC. MATERIALS & METHODS Polyethylene glycol (PEG)ylated-poly(lactic-co-glycolic acid)/polyethyleneimine nanoparticles (PLGA/PEI NPs) were synthesized and complexed with TK-NTR fusion gene. Ultrasound (US) and microbubble (MB) mediated sonoporation was used for efficient delivery of the TK-NTR-DNA-NP complex to TNBC tumor in vivo for cancer therapy. Therapeutic effect was evaluated by treating TNBC cells in vitro and tumor xenograft in vivo by using prodrugs ganciclovir (GCV) and CB1954. RESULTS TNBC cells treated with GCV/CB1954 prodrugs after transfection of TK-NTR-DNA by PEGylated-PLGA/PEI NP resulted in high apoptotic-index. US-MB image-guided delivery of TK-NTR-DNA-NP complex displayed significant expression level of TK-NTR protein and showed tumor reduction when treated with GCV/CB1954 prodrugs in TNBC xenograft in vivo. CONCLUSION US-MB image-guided delivery of TK-NTR gene by PEGylated-PLGA/PEI NPs could be a potential prodrug therapy for TNBC in the clinic.
Collapse
Affiliation(s)
| | - Taehwa Lee
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | | | - Thillai V Sekar
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Kira Foygel
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | | | | | | |
Collapse
|
12
|
Regulated Mesenchymal Stem Cells Mediated Colon Cancer Therapy Assessed by Reporter Gene Based Optical Imaging. Int J Mol Sci 2018; 19:ijms19041002. [PMID: 29584688 PMCID: PMC5979455 DOI: 10.3390/ijms19041002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/24/2018] [Accepted: 03/25/2018] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer is the most common cancer in both men and women and the second most common cause of cancer-related deaths. Suicide gene-based therapy with suicide gene-transduced mesenchymal stem cells (MSCs) is a promising therapeutic strategy. A tetracycline-controlled Tet-On inducible system used to regulate gene expression may be a useful tool for gene-based therapies. The aim of this study was to develop therapeutic MSCs with a suicide gene that is induced by an artificial stimulus, to validate therapeutic gene expression, and to monitor the MSC therapy for colon cancer using optical molecular imaging. For our study, we designed the Tet-On system using a retroviral vector and developed a response plasmid RetroX-TRE (tetracycline response element) expressing a mutant form of herpes simplex virus thymidine kinase (HSV1-sr39TK) with dual reporters (eGFP-Fluc2). Bone marrow-derived MSCs were transduced using a RetroX-Tet3G (Clontech, CA, USA) regulatory plasmid and RetroX-TRE-HSV1-sr39TK-eGFP-IRES-Fluc2, for a system with a Tet-On (MSC-Tet-TK/Fluc2 or MSC-Tet-TK) or without a Tet-On (MSC-TK/Fluc2 or MSC-TK) function. Suicide gene engineered MSCs were co-cultured with colon cancer cells (CT26/Rluc) in the presence of the prodrug ganciclovir (GCV) after stimulation with or without doxycycline (DOX). Treatment efficiency was monitored by assessing Rluc (CT26/Rluc) and Fluc (MSC-Tet-TK and MSC-TK) activity using optical imaging. The bystander effect of therapeutic MSCs was confirmed in CT26/Rluc cells after GCV treatment. Rluc activity in CT26/Rluc cells decreased significantly with GCV treatment of DOX(+) cells (p < 0.05 and 0.01) whereas no significant changes were observed in DOX(-) cells. In addition, Fluc activity in also decreased significantly with DOX(+) MSC-Tet-TK cells, but no signal was observed in DOX(-) cells. In addition, an MSC-TK bystander effect was also confirmed. We assessed therapy with this system in a colon cancer xenograft model (CT26/Rluc). We successfully transduced cells and developed a Tet-On system with the suicide gene HSV1-sr39TK. Our results confirmed the therapeutic efficiency of a suicide gene with the Tet-On system for colon cancer. In addition, our results provide an innovative therapeutic approach using the Tet-On system to eradicate tumors by administration of MSC-Tet-TK cells with DOX and GCV.
Collapse
|
13
|
Drug Discovery by Molecular Imaging and Monitoring Therapy Response in Lymphoma. Int J Mol Sci 2017; 18:ijms18081639. [PMID: 28749424 PMCID: PMC5578029 DOI: 10.3390/ijms18081639] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/23/2017] [Accepted: 07/23/2017] [Indexed: 12/12/2022] Open
Abstract
Molecular imaging allows a noninvasive assessment of biochemical and biological processes in living subjects. Treatment strategies for malignant lymphoma depend on histology and tumor stage. For the last two decades, molecular imaging has been the mainstay diagnostic test for the staging of malignant lymphoma and the assessment of response to treatment. This technology enhances our understanding of disease and drug activity during preclinical and clinical drug development. Here, we review molecular imaging applications in drug development, with an emphasis on oncology. Monitoring and assessing the efficacy of anti-cancer therapies in preclinical or clinical models are essential and the multimodal molecular imaging approach may represent a new stage for pharmacologic development in cancer. Monitoring the progress of lymphoma therapy with imaging modalities will help patients. Identifying and addressing key challenges is essential for successful integration of molecular imaging into the drug development process. In this review, we highlight the general usefulness of molecular imaging in drug development and radionuclide-based reporter genes. Further, we discuss the different molecular imaging modalities for lymphoma therapy and their preclinical and clinical applications.
Collapse
|
14
|
Kalimuthu S, Oh JM, Gangadaran P, Zhu L, Lee HW, Jeon YH, Jeong SY, Lee SW, Lee J, Ahn BC. Genetically engineered suicide gene in mesenchymal stem cells using a Tet-On system for anaplastic thyroid cancer. PLoS One 2017; 12:e0181318. [PMID: 28727740 PMCID: PMC5519161 DOI: 10.1371/journal.pone.0181318] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 06/29/2017] [Indexed: 12/21/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is the most aggressive malignancy of the thyroid, during which undifferentiated tumors arise from the thyroid follicular epithelium. ATC has a very poor prognosis due to its aggressive behavior and poor response to conventional therapies. Gene-directed enzyme/prodrug therapy using genetically engineered mesenchymal stromal cells (MSC) is a promising therapeutic strategy. The doxycycline (DOX)-controlled Tet inducible system is the most widely utilized regulatory system and could be a useful tool for therapeutic gene-based therapies. For example, use a synthetic "tetracycline-on" switch system to control the expression of the therapeutic gene thymidine kinase, which converts prodrugs to active drugs. The aim of this study was to develop therapeutic MSCs, harboring an inducible suicide gene, and to validate therapeutic gene expression using optical molecular imaging of ATC. We designed the Tet-On system using a retroviral vector expressing herpes simplex virus thymidine kinase (HSV1-sr39TK) with dual reporters (eGFP-Fluc2). Mouse bone marrow-derived mesenchymal stromal cells (BM-MSC) were transduced using this system with (MSC-Tet-TK/Fluc2) or without (MSC-TK/Fluc) the Tet-On system. Transduced cells were screened and characterized. Engineered MSCs were co-cultured with ATC (CAL62/Rluc) cells in the presence of the prodrug ganciclovir (GCV) and stimulated with DOX. The efficiency of cell killing monitored by assessing Rluc (CAL62/Rluc) and Fluc (MSC-Tet-TK/Fluc and MSC-TK/Fluc) activities using IVIS imaging. Fluc activity increased in MSC-Tet-TK/Fluc cells in a dose dependent manner following DOX treatment (R2 = 0.95), whereas no signal was observed in untreated cells. eGFP could also be visualized after induction with DOX, and the HSV1-TK protein could be detected by western blotting. In MSC-TK/Fluc cells, the Fluc activity increased with increasing cell number (R2 = 0.98), and eGFP could be visualized by fluorescence microscopy. The Fluc activity and cell viability of MSC-Tet-TK/Fluc and MSC-TK/Fluc cells decreased significantly following GCV treatment. A bystander effect of the therapeutic cells confirmed in co-cultures of CAL62 cells, an anaplastic thyroid cancer cell line, with either MSC-Tet-TK/Fluc cells or MSC-TK/Fluc cells. The Rluc activity in MSC-Tet-TK/Fluc co-cultures, derived from the CAL62/Rluc cells, decreased significantly with GCV treatment of DOX treated cultures, whereas no significant changes were observed in untreated cultures. In addition, the Fluc activity of MSC-Tet-TK/Fluc cells also decreased significantly with DOX treatment whereas no signal was present in untreated cultures. A bystander effect also be demonstrated in co-cultures with MSC-TK/Fluc cells and CAL62/Rluc; both the Rluc activity and the Fluc activity were significantly decreased following GCV treatment. We have successfully developed a Tet-On system of gene-directed enzyme/prodrug delivery using MSCs. We confirmed the therapeutic bystander effect in CAL62/Rluc cells with respect to MSC-Tet-TK/Fluc and MSC-TK/Fluc cells after GCV treatment with and without DOX. Our results confirm the therapeutic efficiency of a suicide gene, with or without the Tet-On system, for ATC therapy. In addition, our findings provide an innovative therapeutic approach for using the Tet-On system to eradicate tumors by simple, repeated administration of MSC-Tet-TK/Fluc cells with DOX and GCV.
Collapse
Affiliation(s)
- Senthilkumar Kalimuthu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Ji Min Oh
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Liya Zhu
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Ho Won Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Yong Hyun Jeon
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine/Hospital, Daegu, Republic of Korea
| |
Collapse
|
15
|
Heterologous Overexpression and Biochemical Characterization of a Nitroreductase from Gluconobacter oxydans 621H. Mol Biotechnol 2017; 58:428-40. [PMID: 27138989 DOI: 10.1007/s12033-016-9942-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A NADPH-dependent and FMN-containing nitroreductase (Gox0834) from Gluconobacter oxydans was cloned and heterogeneously expressed in Escherichia coli. The purified enzyme existed as a dimer with an apparent molecular mass of about 31.4 kDa. The enzyme displayed broad substrate specificity and reduced a variety of mononitrated, polynitrated, and polycyclic nitroaromatic compounds to the corresponding amino products. The highest activity was observed for the reduction of CB1954 (5-(1-aziridinyl)-2,4-dinitrobenzamide). The enzyme kinetics analysis showed that Gox0834 had relatively low K m (54 ± 11 μM) but high k cat/K m value (0.020 s(-1)/μM) for CB1954 when compared with known nitroreductases. Nitrobenzene and 2,4,6-trinitrotoluene (TNT) were preferred substrates for this enzyme with specific activity of 11.0 and 8.9 μmol/min/mg, respectively. Gox0834 exhibited a broad temperature optimum of 40-60 °C for the reduction of CB1954 with a pH optimum between 7.5 and 8.5. The purified enzyme was very stable below 37 °C over a broad pH range of 6.0-10.0. These characteristics suggest that the nitroreductase Gox0834 may be a possible candidate for catalyzing prodrug activation, bioremediation, or biocatalytic processes.
Collapse
|
16
|
An electrochemical biosensor for double-stranded Wnt7B gene detection based on enzymatic isothermal amplification. Biosens Bioelectron 2016; 86:75-82. [DOI: 10.1016/j.bios.2016.06.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/10/2016] [Accepted: 06/11/2016] [Indexed: 11/23/2022]
|
17
|
Teng G, Ju Y, Yang Y, Hua H, Chi J, Mu X. Combined antitumor activity of the nitroreductase/CB1954 suicide gene system and γ-rays in HeLa cells in vitro. Mol Med Rep 2016; 14:5164-5170. [PMID: 27840931 PMCID: PMC5355654 DOI: 10.3892/mmr.2016.5917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 10/11/2016] [Indexed: 11/16/2022] Open
Abstract
Escherichia coli nitroreductase (NTR) may convert the prodrug CB1954 (5-(aziridin-1-yl)-2,4-dinitrobenzamide) into a bifunctional alkylating agent, which may lead to DNA crosslinks and the apoptosis of cancer cells. NTR/CB1954 has been demonstrated to be an effective gene therapy in cancer cells. The present study examined whether the NTR/CB1954 suicide gene system had cytotoxic effects on HeLa cells and may improve the radiosensitivity of HeLa cells to γ-rays. It was observed that the NTR/CB1954 suicide gene system exerted marked cytotoxic effects on HeLa cells. The combined therapeutic effects of NTR/CB1954 and γ-rays on HeLa cells demonstrated a synergistic effect. CB1954 at concentrations of 12.5 and 25 µmol/l increased the sensitization enhancement ratio of HeLa cells to 1.54 and 1.66, respectively. Therefore, when compared with monotherapy, the combined therapy of NTR/CB1954 and γ-rays may increase the apoptotic rate and enhance the radiosensitivity of HeLa cells. The combined therapy of γ-ray radiation and the NTR/CB1954 suicide gene system may be a novel and potent therapeutic method for the treatment of cervical carcinoma.
Collapse
Affiliation(s)
- Geling Teng
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yuanrong Ju
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yepeng Yang
- Department of Radiation Medicine, Peking University Health Science Centre, Beijing 100191, P.R. China
| | - Hu Hua
- Department of Respiratory Medicine, Chest Hospital, Jinan, Shandong 250013, P.R. China
| | - Jingyu Chi
- Department of Respiratory Medicine, Chest Hospital, Jinan, Shandong 250013, P.R. China
| | - Xiuan Mu
- Foreign Language Department, Shandong Medical College, Jinan, Shandong 250002, P.R. China
| |
Collapse
|
18
|
Progress of Multimodal Molecular Imaging Technology in Diagnosis of Tumor. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2016. [DOI: 10.1016/s1872-2040(16)60966-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
19
|
Kim J, Won Y, Goh SH, Choi Y. A redox-responsive theranostic agent for target-specific fluorescence imaging and photodynamic therapy of EGFR-overexpressing triple-negative breast cancers. J Mater Chem B 2016; 4:6787-6790. [DOI: 10.1039/c6tb01519k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A redox-responsive specific theranostic agent for target-cell-specific activatable fluorescence imaging and photodynamic therapy of triple-negative breast cancers.
Collapse
Affiliation(s)
- Jisu Kim
- Molecular Imaging & Therapy Branch
- National Cancer Center
- Goyang-si
- Republic of Korea
| | - Youngjae Won
- Medical Device Development Center
- Osong Medical Innovation Foundation
- Republic of Korea
| | - Sung-Ho Goh
- Precision Medicine Branch
- National Cancer Center
- Goyang-si
- Republic of Korea
| | - Yongdoo Choi
- Molecular Imaging & Therapy Branch
- National Cancer Center
- Goyang-si
- Republic of Korea
- Precision Medicine Branch
| |
Collapse
|
20
|
Nitroreductase gene-directed enzyme prodrug therapy: insights and advances toward clinical utility. Biochem J 2015; 471:131-53. [PMID: 26431849 DOI: 10.1042/bj20150650] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review examines the vast catalytic and therapeutic potential offered by type I (i.e. oxygen-insensitive) nitroreductase enzymes in partnership with nitroaromatic prodrugs, with particular focus on gene-directed enzyme prodrug therapy (GDEPT; a form of cancer gene therapy). Important first indications of this potential were demonstrated over 20 years ago, for the enzyme-prodrug pairing of Escherichia coli NfsB and CB1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide]. However, it has become apparent that both the enzyme and the prodrug in this prototypical pairing have limitations that have impeded their clinical progression. Recently, substantial advances have been made in the biodiscovery and engineering of superior nitroreductase variants, in particular development of elegant high-throughput screening capabilities to enable optimization of desirable activities via directed evolution. These advances in enzymology have been paralleled by advances in medicinal chemistry, leading to the development of second- and third-generation nitroaromatic prodrugs that offer substantial advantages over CB1954 for nitroreductase GDEPT, including greater dose-potency and enhanced ability of the activated metabolite(s) to exhibit a local bystander effect. In addition to forging substantial progress towards future clinical trials, this research is supporting other fields, most notably the development and improvement of targeted cellular ablation capabilities in small animal models, such as zebrafish, to enable cell-specific physiology or regeneration studies.
Collapse
|
21
|
Vorobyeva AG, Stanton M, Godinat A, Lund KB, Karateev GG, Francis KP, Allen E, Gelovani JG, McCormack E, Tangney M, Dubikovskaya EA. Development of a Bioluminescent Nitroreductase Probe for Preclinical Imaging. PLoS One 2015; 10:e0131037. [PMID: 26110789 PMCID: PMC4482324 DOI: 10.1371/journal.pone.0131037] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 05/28/2015] [Indexed: 12/14/2022] Open
Abstract
Bacterial nitroreductases (NTRs) have been widely utilized in the development of novel antibiotics, degradation of pollutants, and gene-directed enzyme prodrug therapy (GDEPT) of cancer that reached clinical trials. In case of GDEPT, since NTR is not naturally present in mammalian cells, the prodrug is activated selectively in NTR-transformed cancer cells, allowing high efficiency treatment of tumors. Currently, no bioluminescent probes exist for sensitive, non-invasive imaging of NTR expression. We therefore developed a "NTR caged luciferin" (NCL) probe that is selectively reduced by NTR, producing light proportional to the NTR activity. Here we report successful application of this probe for imaging of NTR in vitro, in bacteria and cancer cells, as well as in vivo in mouse models of bacterial infection and NTR-expressing tumor xenografts. This novel tool should significantly accelerate the development of cancer therapy approaches based on GDEPT and other fields where NTR expression is important.
Collapse
Affiliation(s)
- Anzhelika G. Vorobyeva
- School of Basic Sciences, Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne, Lausanne, Switzerland
| | - Michael Stanton
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | - Aurélien Godinat
- School of Basic Sciences, Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne, Lausanne, Switzerland
| | - Kjetil B. Lund
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Grigory G. Karateev
- School of Basic Sciences, Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne, Lausanne, Switzerland
| | | | - Elizabeth Allen
- School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology of Lausanne, Lausanne, Switzerland
| | - Juri G. Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Emmet McCormack
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Mark Tangney
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | - Elena A. Dubikovskaya
- School of Basic Sciences, Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
22
|
Zhang TY, Huang B, Wu HB, Wu JH, Li LM, Li YX, Hu YL, Han M, Shen YQ, Tabata Y, Gao JQ. Synergistic effects of co-administration of suicide gene expressing mesenchymal stem cells and prodrug-encapsulated liposome on aggressive lung melanoma metastases in mice. J Control Release 2015; 209:260-71. [PMID: 25966361 DOI: 10.1016/j.jconrel.2015.05.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 04/17/2015] [Accepted: 05/08/2015] [Indexed: 11/30/2022]
Abstract
The success of conventional suicide gene therapy for cancer treatment is still limited because of lack of efficient delivery methods, as well as poor penetration into tumor tissues. Mesenchymal stem cells (MSCs) have recently emerged as potential vehicles in improving delivery issues. However, these stem cells are usually genetically modified using viral gene vectors for suicide gene overexpression to induce sufficient therapeutic efficacy. This approach may result in safety risks for clinical translation. Therefore, we designed a novel strategy that uses non-viral gene vector in modifying MSCs with suicide genes to reduce risks. In addition, these cells were co-administrated with prodrug-encapsulated liposomes for synergistic anti-tumor effects. Results demonstrate that this strategy is effective for gene and prodrug delivery, which co-target tumor tissues, to achieve a significant decrease in tumor colonization and a subsequent increase in survival in a murine melanoma lung metastasis model. Moreover, for the first time, we demonstrated the permeability of MSCs within tumor nests by using an in vitro 3D tumor spheroid model. Thus, the present study provides a new strategy to improve the delivery problem in conventional suicide gene therapy and enhance the therapeutic efficacy. Furthermore, this study also presents new findings to improve our understanding of MSCs in tumor-targeted gene delivery.
Collapse
Affiliation(s)
- Tian-Yuan Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Bing Huang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Hai-Bin Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Jia-He Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Li-Ming Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Yan-Xin Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Yu-Lan Hu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Min Han
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - You-Qing Shen
- Center for Bionanoengineering and State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou, PR China
| | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Jian-Qing Gao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China.
| |
Collapse
|
23
|
Zhu J, Huang H, Dong S, Ge L, Zhang Y. Progress in aptamer-mediated drug delivery vehicles for cancer targeting and its implications in addressing chemotherapeutic challenges. Theranostics 2014; 4:931-44. [PMID: 25057317 PMCID: PMC4107293 DOI: 10.7150/thno.9663] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/23/2014] [Indexed: 12/28/2022] Open
Abstract
Aptamers are novel oligonucleotides with flexible three-dimensional configurations that recognize and bind to their cognate targets, including tumor surface receptors, in a high-affinity and highly specific manner. Because of their unique intrinsic properties, a variety of aptamer-mediated nanovehicles have been developed to directionally transport anti-cancer drugs to tumor sites to minimize systemic cytotoxicity and to enhance permeation by these tumoricidal agents. Despite advances in the selection and synthesis of aptamers and in the conjugation and self-assembly of nanotechnologies, current chemotherapy and drug delivery systems face great challenges. These challenges are due to the limitations of aptamers and vehicles and because of complicated tumor mechanisms, including heterogeneity, anti-cancer drug resistance, and hypoxia-induced aberrances. In this review, we will summarize current approaches utilizing tumor surface hallmarks and aptamers and their roles and mechanisms in therapeutic nanovehicles targeting tumors. Delivery forms include nanoparticles, nanotubes, nanogels, aptamer-drug conjugates, and novel molecular trains. Moreover, the obstacles posed by the aforementioned issues will be highlighted, and possible solutions will be acknowledged. Furthermore, future perspectives will be presented, including cutting-edge integration with RNA interference nanotechnology and personalized chemotherapy, which will facilitate innovative approaches to aptamer-based therapeutics.
Collapse
|
24
|
Zhan Y, Yu B, Wang Z, Zhang Y, Zhang HH, Wu H, Feng X, Geng RS, Kong W, Yu XH. A fiber-modified adenovirus co-expressing HSV-TK and Coli.NTR enhances antitumor activities in breast cancer cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:2850-2860. [PMID: 25031704 PMCID: PMC4097267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 05/26/2014] [Indexed: 06/03/2023]
Abstract
Breast cancers especially in late and metastatic stages remain refractory to treatment despite advances in surgical techniques and chemotherapy. Suicide gene therapy based on adenoviral technology will be promising strategies for such advanced diseases. We previously showed that co-expression of herpes simplex virus thymidine kinase (HSV-TK) and Escherichia coli nitroreductase (Coli.NTR) by an hTERT-driven adenovirus vector resulted in additive anti-tumor effects in breast cancer cells in vitro and in vivo. As many tumor tissue and cancer cells express low level of coxsackie-adenovirus receptor (CAR), which is the functional receptor for the fiber protein of human adenovirus serotype 5 (Ad5), novel Ad5 vectors containing genetically modifi ed fiber are attractive vehicles for achieving targeted gene transfer and improving suicide gene expression in these cancer cells. In the present study, we first built a simplified Ad5 vector platform for fiber modification and quick detection for gene transfer. Then a fiber-modified adenovirus vector containing an RGD motif in the HI loop of the fiber knob was constructed. After recombined with HSV-TK and Coli.NTR gene, this fiber-modified Ad5 vector (Ad-RGD-hT-TK/NTR) was compared with that of our previously constructed Ad5 vector (Ad-hT-TK/NTR) for its therapeutic effects in human breast cancer cell lines. The anti-tumor activity of Ad-RGD-hT-TK/NTR was significantly enhanced compared with Ad-hT-TK/NTR both in vitro and in vivo. This new vector platform provided a robust and simplified approach for capsid modification, and the fiber-modified Ad5 with double suicide genes under the control of hTERT promoter would be a useful gene therapy strategy for breast cancer.
Collapse
Affiliation(s)
- Yang Zhan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Zhen Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Yu Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Hai-Hong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Hao Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Xiao Feng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Ran-Shen Geng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| | - Xiang-Hui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University2699 Qianjin Street, Changchun 130012, China
| |
Collapse
|