1
|
Wyszatko K, Janzen N, Silva LR, Kwon L, Komal T, Ventura M, Venugopal C, Singh SK, Valliant JF, Sadeghi S. 89Zr-labeled ImmunoPET targeting the cancer stem cell antigen CD133 using fully-human antibody constructs. EJNMMI Res 2024; 14:29. [PMID: 38498285 PMCID: PMC10948676 DOI: 10.1186/s13550-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/04/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Cancer stem cells play an important role in driving tumor growth and treatment resistance, which makes them a promising therapeutic target to prevent cancer recurrence. Emerging cancer stem cell-targeted therapies would benefit from companion diagnostic imaging probes to aid in patient selection and monitoring response to therapy. To this end, zirconium-89-radiolabeled immunoPET probes that target the cancer stem cell-antigen CD133 were developed using fully human antibody and antibody scFv-Fc scaffolds. RESULTS ImmunoPET probes [89Zr]-DFO-RW03IgG (CA = 0.7 ± 0.1), [89Zr]-DFO-RW03IgG (CA = 3.0 ± 0.3), and [89Zr]-DFO-RW03scFv - Fc (CA = 2.9 ± 0.3) were radiolabeled with zirconium-89 (radiochemical yield 42 ± 5%, 97 ± 2%, 86 ± 12%, respectively) and each was isolated in > 97% radiochemical purity with specific activities of 120 ± 30, 270 ± 90, and 200 ± 60 MBq/mg, respectively. In vitro binding assays showed a low-nanomolar binding affinity of 0.6 to 1.1 nM (95% CI) for DFO-RW03IgG (CA = 0.7 ± 0.1), 0.3 to 1.9 nM (95% CI) for DFO-RW03IgG (CA = 3.0 ± 0.3), and 1.5 to 3.3 nM (95% CI) for DFO-RW03scFv - Fc (C/A = 0.3). Biodistribution studies found that [89Zr]-DFO-RW03scFv - Fc (CA = 2.9 ± 0.3) exhibited the highest tumor uptake (23 ± 4, 21 ± 2, and 23 ± 4%ID/g at 24, 48, and 72 h, respectively) and showed low uptake (< 6%ID/g) in all off-target organs at each timepoint (24, 48, and 72 h). Comparatively, [89Zr]-DFO-RW03IgG (CA = 0.7 ± 0.1) and [89Zr]-DFO-RW03IgG (CA = 3.0 ± 0.3) both reached maximum tumor uptake (16 ± 3%ID/g and 16 ± 2%ID/g, respectively) at 96 h p.i. and showed higher liver uptake (10.2 ± 3%ID/g and 15 ± 3%ID/g, respectively) at that timepoint. Region of interest analysis to assess PET images of mice administered [89Zr]-DFO-RW03scFv - Fc (CA = 2.9 ± 0.3) showed that this probe reached a maximum tumor uptake of 22 ± 1%ID/cc at 96 h, providing a tumor-to-liver ratio that exceeded 1:1 at 48 h p.i. Antibody-antigen mediated tumor uptake was demonstrated through biodistribution and PET imaging studies, where for each probe, co-injection of excess unlabeled RW03IgG resulted in > 60% reduced tumor uptake. CONCLUSIONS Fully human CD133-targeted immunoPET probes [89Zr]-DFO-RW03IgG and [89Zr]-DFO-RW03scFv - Fc accumulate in CD133-expressing tumors to enable their delineation through PET imaging. Having identified [89Zr]-DFO-RW03scFv - Fc (CA = 2.9 ± 0.3) as the most attractive construct for CD133-expressing tumor delineation, the next step is to evaluate this probe using patient-derived tumor models to test its detection limit prior to clinical translation.
Collapse
Affiliation(s)
- Kevin Wyszatko
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Nancy Janzen
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Luis Rafael Silva
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Luke Kwon
- Spatio-Temporal Targeting and Amplification of Radiation Response Innovation Centre (STTARR), University Health Network, Toronto, ON, Canada
| | - Teesha Komal
- Spatio-Temporal Targeting and Amplification of Radiation Response Innovation Centre (STTARR), University Health Network, Toronto, ON, Canada
| | - Manuela Ventura
- Spatio-Temporal Targeting and Amplification of Radiation Response Innovation Centre (STTARR), University Health Network, Toronto, ON, Canada
| | - Chitra Venugopal
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Sheila K Singh
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - John F Valliant
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Saman Sadeghi
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
2
|
Jin C, Luo X, Li X, Zhou R, Zhong Y, Xu Z, Cui C, Xing X, Zhang H, Tian M. Positron emission tomography molecular imaging-based cancer phenotyping. Cancer 2022; 128:2704-2716. [PMID: 35417604 PMCID: PMC9324101 DOI: 10.1002/cncr.34228] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
During the past several decades, numerous studies have provided insights into biological characteristics of cancer cells and identified various hallmarks of cancer acquired in the tumorigenic processes. However, it is still challenging to image these distinctive traits of cancer to facilitate the management of patients in clinical settings. The rapidly evolving field of positron emission tomography (PET) imaging has provided opportunities to investigate cancer's biological characteristics in vivo. This article reviews the current status of PET imaging on characterizing hallmarks of cancer and discusses the future directions of PET imaging strategies facilitating in vivo cancer phenotyping.
Collapse
Affiliation(s)
- Chentao Jin
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoyun Luo
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoyi Li
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Rui Zhou
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Yan Zhong
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Zhoujiao Xu
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Chunyi Cui
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoqing Xing
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Hong Zhang
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
- College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
- Key Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouChina
| | - Mei Tian
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
3
|
Shigdar S, Schrand B, Giangrande PH, de Franciscis V. Aptamers: Cutting edge of cancer therapies. Mol Ther 2021; 29:2396-2411. [PMID: 34146729 PMCID: PMC8353241 DOI: 10.1016/j.ymthe.2021.06.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
The development of an aptamer-based therapeutic has rapidly progressed following the first two reports in the 1990s, underscoring the advantages of aptamer drugs associated with their unique binding properties. In 2004, the US Food and Drug Administration (FDA) approved the first therapeutic aptamer for the treatment of neovascular age-related macular degeneration, Macugen developed by NeXstar. Since then, eleven aptamers have successfully entered clinical trials for various therapeutic indications. Despite some of the pre-clinical and clinical successes of aptamers as therapeutics, no aptamer has been approved by the FDA for the treatment of cancer. This review highlights the most recent and cutting-edge approaches in the development of aptamers for the treatment of cancer types most refractory to conventional therapies. Herein, we will review (1) the development of aptamers to enhance anti-cancer immunity and as delivery tools for inducing the expression of immunogenic neoantigens; (2) the development of the most promising therapeutic aptamers designed to target the hard-to-treat cancers such as brain tumors; and (3) the development of "carrier" aptamers able to target and penetrate tumors and metastasis, delivering RNA therapeutics to the cytosol and nucleus.
Collapse
Affiliation(s)
- Sarah Shigdar
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, 75 Pigdons Road, Waurn Ponds, VIC 3216, Australia
| | - Brett Schrand
- TCR(2) Therapeutics, Inc., 100 Binney Street, Cambridge, MA 02142, USA
| | - Paloma H Giangrande
- Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; VP Platform Discovery Sciences, Biology, Wave Life Sciences, Cambridge, MA 02138, USA
| | - Vittorio de Franciscis
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan, Italy; Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
4
|
Schmidtova S, Dorssers LCJ, Kalavska K, Gillis AJM, Oosterhuis JW, Stoop H, Miklikova S, Kozovska Z, Burikova M, Gercakova K, Durinikova E, Chovanec M, Mego M, Kucerova L, Looijenga LHJ. Napabucasin overcomes cisplatin resistance in ovarian germ cell tumor-derived cell line by inhibiting cancer stemness. Cancer Cell Int 2020; 20:364. [PMID: 32774158 PMCID: PMC7397611 DOI: 10.1186/s12935-020-01458-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/25/2020] [Indexed: 02/08/2023] Open
Abstract
Background Cisplatin resistance of ovarian yolk sac tumors (oYST) is a clinical challenge due to dismal patient prognosis, even though the disease is extremely rare. We investigated potential association between cisplatin resistance and cancer stem cell (CSC) markers in chemoresistant oYST cells and targeting strategies to overcome resistance in oYST. Methods Chemoresistant cells were derived from chemosensitive human oYST cells by cultivation in cisplatin in vitro. Derivative cells were characterized by chemoresistance, functional assays, flow cytometry, gene expression and protein arrays focused on CSC markers. RNAseq, methylation and microRNA profiling were performed. Quail chorioallantoic membranes (CAM) with implanted oYST cells were used to analyze the micro-tumor extent and interconnection with the CAM. Tumorigenicity in vivo was determined on immunodeficient mouse model. Chemoresistant cells were treated by inhibitors intefering with the CSC properties to examine the chemosensitization to cisplatin. Results Long-term cisplatin exposure resulted in seven-fold higher IC50 value in resistant cells, cross-resistance to oxaliplatin and carboplatin, and increased migratory capacity, invasiveness and tumorigenicity, associated with hypomethylation of differentially methylated genes/promotors. Resistant cells exhibited increased expression of prominin-1 (CD133), ATP binding cassette subfamily G member 2 (ABCG2), aldehyde dehydrogenase 3 isoform A1 (ALDH3A1), correlating with reduced gene and promoter methylation, as well as increased expression of ALDH1A3 and higher overall ALDH enzymatic activity, rendering them cross-resistant to DEAB, disulfiram and napabucasin. Salinomycin and tunicamycin were significantly more toxic to resistant cells. Pretreatment with napabucasin resensitized the cells to cisplatin and reduced their tumorigenicity in vivo. Conclusions The novel chemoresistant cells represent unique model of refractory oYST. CSC markers are associated with cisplatin resistance being possible targets in chemorefractory oYST.
Collapse
Affiliation(s)
- Silvia Schmidtova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.,Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
| | - Lambert C J Dorssers
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Katarina Kalavska
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.,Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Ad J M Gillis
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - J Wolter Oosterhuis
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Hans Stoop
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Svetlana Miklikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Zuzana Kozovska
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Monika Burikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Katarina Gercakova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Erika Durinikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Michal Chovanec
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Michal Mego
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Lucia Kucerova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Leendert H J Looijenga
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
5
|
Pastò A, Consonni FM, Sica A. Influence of Innate Immunity on Cancer Cell Stemness. Int J Mol Sci 2020; 21:ijms21093352. [PMID: 32397392 PMCID: PMC7247585 DOI: 10.3390/ijms21093352] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/02/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Even if cancer stem cells (CSCs) represent only a small proportion of the tumor mass, they significantly account for tumor maintenance, resistance to therapies, relapse and metastatic spread, due to their increased capacity of self-renewal, multipotency, tumorigenicity and quiescence. Emerging evidence suggests that the immune contexture within the tumor microenvironment (TME) determines both the response to therapy and the clinical outcome. In this context, CSCs acquire immune evasion skills by editing immune cell functions and sculpting the immunosuppressive landscape of TME. Reciprocally, infiltrating immune cells influence CSCs self-renewal, tumorigenicity and metastasis. In this review, we summarize the immunomodulatory properties of CSCs, as well as the impact of innate immune cells on cancer cells stemness in the different phases of cancer immunoediting process and neoplastic progression.
Collapse
Affiliation(s)
- Anna Pastò
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center–IRCCS–, via Manzoni 56, 20089 Rozzano (MI), Italy;
| | - Francesca Maria Consonni
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, via Bovio 6, 28100 Novara, Italy;
| | - Antonio Sica
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center–IRCCS–, via Manzoni 56, 20089 Rozzano (MI), Italy;
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, via Bovio 6, 28100 Novara, Italy;
- Correspondence: ; Tel.: +39-0321-375-881; Fax: +39-0321-375-621
| |
Collapse
|
6
|
Arnold CR, Mangesius J, Skvortsova II, Ganswindt U. The Role of Cancer Stem Cells in Radiation Resistance. Front Oncol 2020; 10:164. [PMID: 32154167 PMCID: PMC7044409 DOI: 10.3389/fonc.2020.00164] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSC) are a distinct subpopulation within a tumor. They are able to self-renew and differentiate and possess a high capability to repair DNA damage, exhibit low levels of reactive oxygen species (ROS), and proliferate slowly. These features render CSC resistant to various therapies, including radiation therapy (RT). Eradication of all CSC is a requirement for an effective antineoplastic treatment and is therefore of utmost importance for the patient. This makes CSC the prime targets for any therapeutic approach. Albeit clinical data is still scarce, experimental data and first clinical trials give hope that CSC-targeted treatment has the potential to improve antineoplastic therapies, especially for tumors that are known to be treatment resistant, such as glioblastoma. In this review, we will discuss CSC in the context of RT, describe known mechanisms of resistance, examine the possibilities of CSC as biomarkers, and discuss possible new treatment approaches.
Collapse
Affiliation(s)
- Christoph Reinhold Arnold
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Julian Mangesius
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ira-Ida Skvortsova
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria.,EXTRO-Lab, Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Ute Ganswindt
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
7
|
Kim DM, Kim M, Park HB, Kim KS, Kim DE. Anti-MUC1/CD44 Dual-Aptamer-Conjugated Liposomes for Cotargeting Breast Cancer Cells and Cancer Stem Cells. ACS APPLIED BIO MATERIALS 2019; 2:4622-4633. [DOI: 10.1021/acsabm.9b00705] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Dong-Min Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Minhee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Hee-Bin Park
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea
| | - Keun-Sik Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
8
|
Wong ALA, Bellot GL, Hirpara JL, Pervaiz S. Understanding the cancer stem cell phenotype: A step forward in the therapeutic management of cancer. Biochem Pharmacol 2019; 162:79-88. [PMID: 30689981 DOI: 10.1016/j.bcp.2019.01.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/24/2019] [Indexed: 12/15/2022]
Abstract
The experimental validation of the existence of cancer stem cells (CSC) has had a significant impact on our understanding of the cellular mechanisms and signaling networks involved in the process of carcinogenesis and its progression. These findings provide insights into the critical role that tumor microenvironment and metabolism play in the acquisition of the drug resistance phenotype as well as provide potential targets for therapeutic exploitation. Here we briefly review the literature on the involvement of key signaling pathways such as Wnt/β-catenin, Notch, Hedgehog and STAT3 in the appearance of cancer cells with stem cells-like characteristics. In addition, we also highlight some of the recent therapeutic strategies used to target these pathways as well as approaches aiming to specifically target CSCs through their distinctive metabolic features.
Collapse
Affiliation(s)
- Andrea Li Ann Wong
- Cancer Science Institute, National University of Singapore, Singapore; Department of Hematology-Oncology, National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gregory Lucien Bellot
- Department of Hand & Reconstructive Microsurgery, University Orthopedic, Hand & Reconstructive Microsurgery Cluster, National University Health System, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jayshree L Hirpara
- Cancer Science Institute, National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.
| |
Collapse
|
9
|
Abstract
Cancer is one of the most serious diseases all over the world, and the cancer stem cell (CSC) model accounts for tumor initiation, metastasis, drug resistance, and relapse. The CSCs within tumor bulk have the capacity to self-renew, differentiate, and give rise to a new tumor. The self-renewal of CSCs is precisely regulated by various modulators, including Wnt/β-catenin signaling, Notch signaling, Hedgehog signaling, transcription factors, chromatin remodeling complexes, and non-coding RNAs. CSCs reside in their niches that are also involved in the self-renewal maintenance of CSCs and protection of CSCs from chemotherapy, radiotherapy, and even endogenous damages. Moreover, CSCs can also remodel their niches to initiate tumorigenesis. The mutual interactions between CSCs and their niches play a critical role in the regulation of CSC self-renewal and tumorigenesis as well. Many surface markers of CSCs have been identified, and these markers become first choices for CSC targeting. Due to heterogeneity and plasticity, targeting CSCs is still a big challenge for tumor elimination. In this review, we summarize recent progresses on the biological features of CSCs and targeting strategies against CSCs.
Collapse
Affiliation(s)
- Pingping Zhu
- 1CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Zusen Fan
- 1CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China.,2University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
10
|
Chen F, Zeng Y, Qi X, Chen Y, Ge Z, Jiang Z, Zhang X, Dong Y, Chen H, Yu Z. Targeted salinomycin delivery with EGFR and CD133 aptamers based dual-ligand lipid-polymer nanoparticles to both osteosarcoma cells and cancer stem cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2115-2127. [PMID: 29898423 DOI: 10.1016/j.nano.2018.05.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/17/2018] [Accepted: 05/29/2018] [Indexed: 12/24/2022]
Abstract
We previously developed salinomycin (sali)-entrapped nanoparticles labeled with CD133 aptamers which could efficiently eliminate CD133+ osteosarcoma cancer stem cells (CSCs). However, sufficient evidences suggest that the simultaneous targeting both CSCs and cancer cells is pivotal in achieving preferable cancer therapeutic efficacy, due to the spontaneous conversion between cancer cells and CSCs. We hereby constructed sali-entrapped lipid-polymer nanoparticles labeled with CD133 and EGFR aptamers (CESP) to target both osteosarcoma cells and CSCs. The cytotoxicity of CESP in osteosarcoma cells and CSCs was superior to that of single targeting or nontargeted sali-loaded nanoparticles. Administration of CESP in vivo showed the best efficacy in inhibiting tumor growth than other controls in osteosarcoma-bearing mice. Thus, CESP was demonstrated to be capable of efficiently targeting both osteosarcoma CSCs and cancer cells, and it represents an effective potential approach to treat osteosarcoma.
Collapse
Affiliation(s)
- Fangyi Chen
- Department of Orthopedics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yibin Zeng
- Department of Dermatology, Central Hospital of Minhang District, Shanghai, China
| | - Xiaoxia Qi
- The Wound Care Center, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yanchao Chen
- Department of Orthopedics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Zhe Ge
- Department of Orthopedics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Zengxin Jiang
- Department of Orthopedics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Xinchao Zhang
- Department of Orthopedics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Yinmei Dong
- Center of Clinical and Translational Medicine, Shanghai Changhai Hospital, Shanghai, China
| | - Huaiwen Chen
- Sunlipo Biotech Research Center for Nanomedicine, Shanghai, China; Center of Clinical and Translational Medicine, Shanghai Changhai Hospital, Shanghai, China.
| | - Zuochong Yu
- Department of Orthopedics, Jinshan Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Acheampong DO, Adokoh CK, Asante DB, Asiamah EA, Barnie PA, Bonsu DOM, Kyei F. Immunotherapy for acute myeloid leukemia (AML): a potent alternative therapy. Biomed Pharmacother 2017; 97:225-232. [PMID: 29091870 DOI: 10.1016/j.biopha.2017.10.100] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/11/2017] [Accepted: 10/21/2017] [Indexed: 12/20/2022] Open
Abstract
The standard therapy of AML for many years has been chemotherapy with or without stem transplantation. However, there has not been any tangible improvement in this treatment beyond induction through chemotherapy and consolidation with allogeneic stem cell transplantation or chemotherapy. Residual AML cells which later cause relapse mostly persist even after rigorous standard therapy. It is imperative therefore to find an alternative therapy that can take care of the residual AML cells. With a better understanding of how the immune system works to destroy tumor cells and inhibit their growth, another therapeutic option immunotherapy has emerged to address the difficulties associated with the standard therapy. Identification of leukemia-associated antigens (LAA) and the fact that T and NK cells can be activated to exert cytotoxicity on AML cells have further introduced diverse immunotherapeutic development strategies. This review discusses the merits of current immunotherapeutic strategies such as the use of antibodies, adoptive T cells and alloreactive NK cell, and vaccination as against the standard therapy of AML.
Collapse
Affiliation(s)
| | - Christian K Adokoh
- Department of Forensic Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Du-Bois Asante
- Department of Forensic Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Ernest A Asiamah
- Department of Biomedical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Prince A Barnie
- Department of Forensic Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Dan O M Bonsu
- Department of Forensic Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Foster Kyei
- Department of Molecular Biology and Biotechnology, University of Cape Coast, Ghana
| |
Collapse
|
12
|
Finley J. Elimination of cancer stem cells and reactivation of latent HIV-1 via AMPK activation: Common mechanism of action linking inhibition of tumorigenesis and the potential eradication of HIV-1. Med Hypotheses 2017; 104:133-146. [PMID: 28673572 DOI: 10.1016/j.mehy.2017.05.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 02/28/2017] [Accepted: 05/26/2017] [Indexed: 12/25/2022]
Abstract
Although promising treatments are currently in development to slow disease progression and increase patient survival, cancer remains the second leading cause of death in the United States. Cancer treatment modalities commonly include chemoradiation and therapies that target components of aberrantly activated signaling pathways. However, treatment resistance is a common occurrence and recent evidence indicates that the existence of cancer stem cells (CSCs) may underlie the limited efficacy and inability of current treatments to effectuate a cure. CSCs, which are largely resistant to chemoradiation therapy, are a subpopulation of cancer cells that exhibit characteristics similar to embryonic stem cells (ESCs), including self-renewal, multi-lineage differentiation, and the ability to initiate tumorigenesis. Interestingly, intracellular mechanisms that sustain quiescence and promote self-renewal in adult stem cells (ASCs) and CSCs likely also function to maintain latency of HIV-1 in CD4+ memory T cells. Although antiretroviral therapy is highly effective in controlling HIV-1 replication, the persistence of latent but replication-competent proviruses necessitates the development of compounds that are capable of selectively reactivating the latent virus, a method known as the "shock and kill" approach. Homeostatic proliferation in central CD4+ memory T (TCM) cells, a memory T cell subset that exhibits limited self-renewal and differentiation and is a primary reservoir for latent HIV-1, has been shown to reinforce and stabilize the latent reservoir in the absence of T cell activation and differentiation. HIV-1 has also been found to establish durable and long-lasting latency in a recently discovered subset of CD4+ T cells known as T memory stem (TSCM) cells. TSCM cells, compared to TCM cells, exhibit stem cell properties that more closely match those of ESCs and ASCs, including self-renewal and differentiation into all memory T cell subsets. It is our hypothesis that activation of AMPK, a master regulator of cellular metabolism that plays a critical role in T cell activation and differentiation of ESCs and ASCs, will lead to both T cell activation-induced latent HIV-1 reactivation, facilitating virus destruction, as well as "activation", differentiation, and/or apoptosis of CSCs, thus inhibiting tumorigenesis. We also propose the novel observation that compounds that have been shown to both facilitate latent HIV-1 reactivation and promote CSC differentiation/apoptosis (e.g. bryostatin-1, JQ1, metformin, butyrate, etc.) likely do so through a common mechanism of AMPK activation.
Collapse
Affiliation(s)
- Jahahreeh Finley
- Finley BioSciences, 9900 Richmond Avenue, #823, Houston, TX 77042-4539, United States.
| |
Collapse
|
13
|
Gao J, Li W, Guo Y, Feng SS. Nanomedicine strategies for sustained, controlled and targeted treatment of cancer stem cells. Nanomedicine (Lond) 2016; 11:3261-3282. [PMID: 27854161 DOI: 10.2217/nnm-2016-0261] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cancer stem cells (CSCs) are original cancer cells that are of characteristics associated with normal stem cells. CSCs are toughest against various treatments and thus responsible for cancer metastasis and recurrence. Therefore, development of specific and effective treatment of CSCs plays a key role in improving survival and life quality of cancer patients, especially those in the metastatic stage. Nanomedicine strategies, which include prodrugs, micelles, liposomes and nanoparticles of biodegradable polymers, could substantially improve the therapeutic index of conventional therapeutics due to its manner of sustained, controlled and targeted delivery of high transportation efficiency across the cell membrane and low elimination by intracellular autophagy, and thus provide a practical solution to solve the problem encountered in CSCs treatment. This review gives briefly the latest information to summarize the concept, strategies, mechanisms and current status as well as future promises of nanomedicine strategies for treatment of CSCs.
Collapse
Affiliation(s)
- Jie Gao
- Department of Pharmaceutical Sciences, School of Pharmacy, the Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wei Li
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| | - Yajun Guo
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| | - Si-Shen Feng
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China.,Department of Chemical & Biomolecular Engineering, National University of Singapore, Block E5, 02-11, 4 Engineering Drive 4, Singapore 117576, Singapore.,Suzhou NanoStar Biopharm Inc. Ltd, BioBay, Bld B2, Unit 604, 218 Xing-Hu Street, Suzhou Industrial Park, Suzhou 215123, China
| |
Collapse
|
14
|
Ruan Z, Liu J, Kuang Y. Isolation and characterization of side population cells from the human ovarian cancer cell line SK-OV-3. Exp Ther Med 2015; 10:2071-2078. [PMID: 26668597 PMCID: PMC4665172 DOI: 10.3892/etm.2015.2836] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 09/29/2015] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OC) is the most malignant type of gynecological tumor due to its high recurrence rate following initial treatment. Previous studies have indicated that cancer stem cells (CSCs) may be a potential cause underlying the high proportion of recurrence. Side population (SP) cells isolated from cancer cell lines have been shown to exhibit characteristics associated with CSCs, but studies on SP cells in human ovarian SK-OV-3 cell line are limited. In the present study, the SP cell fraction (4.83% of the total cell population) was isolated using flow cytometry, and analyzed by immunocytochemical analysis and reverse transcription-quantitative polymerase chain reaction. The results showed that SP cells exhibited a high mean fluorescence intensity for CD44, a CSC marker, in addition to elevated expression of the CSCs-associated genes, ATP-binding cassette sub-family G member 2 and Nestin. These findings indicated the stem cell-like features of the SP cells. Furthermore, a colony formation test showed that the isolated SP cells possessed a marked capacity for self-regeneration and proliferation. In addition, a cell cycle assay involving cisplatin indicated that the SP cells were strongly resistant to chemotherapy. In conclusion, the present results suggested that SP cells isolated from the SK-OV-3 cell line exhibited properties typically associated with CSCs. Therefore, the isolated SP cells may be used to provide novel insight into potential therapies against OC.
Collapse
Affiliation(s)
- Zhengyi Ruan
- Department of Gynecology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Jianhua Liu
- Department of Gynecology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
15
|
Curcumin suppresses stem-like traits of lung cancer cells via inhibiting the JAK2/STAT3 signaling pathway. Oncol Rep 2015; 34:3311-7. [DOI: 10.3892/or.2015.4279] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/10/2015] [Indexed: 11/05/2022] Open
|
16
|
Cancer stem cells and tumor-associated macrophages: a roadmap for multitargeting strategies. Oncogene 2015; 35:671-82. [PMID: 25961921 DOI: 10.1038/onc.2015.132] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/16/2015] [Accepted: 03/20/2015] [Indexed: 12/12/2022]
Abstract
The idea that tumor initiation and progression are driven by a subset of cells endowed with stem-like properties was first described by Rudolf Virchow in 1855. 'Cancer stem cells', as they were termed more than a century later, represent a subset of tumor cells that are able to generate all tumorigenic and nontumorigenic cell types within the malignancy. Although their existence was hypothesized >150 years ago, it was only recently that stem-like cells started to be isolated from different neoplastic malignancies. Interestingly, Virchow, in suggesting a correlation between cancer and the inflammatory microenvironment, also paved the way for the 'Seed and Soil' theory proposed by Paget a few years later. Despite the time that has passed since these two important concepts were suggested, the relationships between Virchow's 'stem-like cells' and Paget's 'soil' are far from being fully understood. One emerging topic is the importance of a stem-like niche in modulating the biological properties of stem-like cancer cells and thus in affecting the response of the tumor to drugs. This review aims to summarize the recent molecular data concerning the multilayered relationship between cancer stem cells and tumor-associated macrophages that form a key component of the tumor microenvironment. We also discuss the therapeutic implications of targeting this synergistic interplay.
Collapse
|
17
|
Cojoc M, Mäbert K, Muders MH, Dubrovska A. A role for cancer stem cells in therapy resistance: Cellular and molecular mechanisms. Semin Cancer Biol 2015; 31:16-27. [DOI: 10.1016/j.semcancer.2014.06.004] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/04/2014] [Accepted: 06/11/2014] [Indexed: 12/11/2022]
|
18
|
Cancer stem cells: a systems biology view of their role in prognosis and therapy. Anticancer Drugs 2014; 25:353-67. [PMID: 24418909 DOI: 10.1097/cad.0000000000000075] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Evidence has accumulated that characterizes highly tumorigenic cancer cells residing in heterogeneous populations. The accepted term for such a subpopulation is cancer stem cells (CSCs). While many questions still remain about their precise role in the origin, progression, and drug resistance of tumors, it is clear they exist. In this review, a current understanding of the nature of CSC, their potential usefulness in prognosis, and the need to target them will be discussed. In particular, separate studies now suggest that the CSC is plastic in its phenotype, toggling between tumorigenic and nontumorigenic states depending on both intrinsic and extrinsic conditions. Because of this, a static view of gene and protein levels defined by correlations may not be sufficient to either predict disease progression or aid in the discovery and development of drugs to molecular targets leading to cures. Quantitative dynamic modeling, a bottom up systems biology approach whereby signal transduction pathways are described by differential equations, may offer a novel means to overcome the challenges of oncology today. In conclusion, the complexity of CSCs can be captured in mathematical models that may be useful for selecting molecular targets, defining drug action, and predicting sensitivity or resistance pathways for improved patient outcomes.
Collapse
|
19
|
Peitzsch C, Perrin R, Hill RP, Dubrovska A, Kurth I. Hypoxia as a biomarker for radioresistant cancer stem cells. Int J Radiat Biol 2014; 90:636-52. [DOI: 10.3109/09553002.2014.916841] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
Mäbert K, Cojoc M, Peitzsch C, Kurth I, Souchelnytskyi S, Dubrovska A. Cancer biomarker discovery: current status and future perspectives. Int J Radiat Biol 2014; 90:659-77. [PMID: 24524284 DOI: 10.3109/09553002.2014.892229] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Cancer is a multigene disease which arises as a result of mutational and epigenetic changes coupled with activation of complex signaling networks. The use of biomarkers for early cancer detection, staging and individualization of therapy might improve patient care. A few fundamental issues such as tumor heterogeneity, a highly dynamic nature of the intrinsic and extrinsic determinants of radio- and chemoresistance, along with the plasticity and diversity of cancer stem cells (CSC) make biomarker development a challenging task. In this review we outline the preclinical strategies of cancer biomarker discovery including genomic, proteomic, metabolomic and microRNomic profiling, comparative genome hybridization (CGH), single nucleotide polymorphism (SNP) analysis, high throughput screening (HTS) and next generation sequencing (NGS). Other promising approaches such as assessment of circulating tumor cells (CTC), analysis of CSC-specific markers and cell-free circulating tumor DNA (ctDNA) are also discussed. CONCLUSIONS The emergence of powerful proteomic and genomic technologies in conjunction with advanced bioinformatic tools allows the simultaneous analysis of thousands of biological molecules. These techniques yield the discovery of new tumor signatures, which are sensitive and specific enough for early cancer detection, for monitoring disease progression and for proper treatment selection, paving the way to individualized cancer treatment.
Collapse
Affiliation(s)
- Katrin Mäbert
- OncoRay-National Center for Radiation Research in Oncology, Medical Faculty Dresden Carl Gustav Carus , TU Dresden , Germany
| | | | | | | | | | | |
Collapse
|
21
|
Li L, Xiang D, Shigdar S, Yang W, Li Q, Lin J, Liu K, Duan W. Epithelial cell adhesion molecule aptamer functionalized PLGA-lecithin-curcumin-PEG nanoparticles for targeted drug delivery to human colorectal adenocarcinoma cells. Int J Nanomedicine 2014; 9:1083-96. [PMID: 24591829 PMCID: PMC3937189 DOI: 10.2147/ijn.s59779] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
To improve the efficacy of drug delivery, active targeted nanotechnology-based drug delivery systems are gaining considerable attention as they have the potential to reduce side effects, minimize toxicity, and improve efficacy of anticancer treatment. In this work CUR-NPs (curcumin-loaded lipid-polymer-lecithin hybrid nanoparticles) were synthesized and functionalized with ribonucleic acid (RNA) Aptamers (Apts) against epithelial cell adhesion molecule (EpCAM) for targeted delivery to colorectal adenocarcinoma cells. These CUR-encapsulated bioconjugates (Apt-CUR-NPs) were characterized for particle size, zeta potential, drug encapsulation, stability, and release. The in vitro specific cell binding, cellular uptake, and cytotoxicity of Apt-CUR-NPs were also studied. The Apt-CUR-NP bioconjugates exhibited increased binding to HT29 colon cancer cells and enhancement in cellular uptake when compared to CUR-NPs functionalized with a control Apt (P<0.01). Furthermore, a substantial improvement in cytotoxicity was achieved toward HT29 cells with Apt-CUR-NP bioconjugates. The encapsulation of CUR in Apt-CUR-NPs resulted in the increased bioavailability of delivered CUR over a period of 24 hours compared to that of free CUR in vivo. These results show that the EpCAM Apt-functionalized CUR-NPs enhance the targeting and drug delivery of CUR to colorectal cancer cells. Further development of CUR-encapsulated, nanosized carriers will lead to improved targeted delivery of novel chemotherapeutic agents to colorectal cancer cells.
Collapse
Affiliation(s)
- Lei Li
- College of Pharmacy, Dalian Medical University, Dalian, People's Republic of China
| | - Dongxi Xiang
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, Australia
| | - Sarah Shigdar
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, Australia
| | - Wenrong Yang
- School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, Waurn Ponds, VIC, Australia
| | - Qiong Li
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, Australia
| | - Jia Lin
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian, People's Republic of China
| | - Wei Duan
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, Australia
| |
Collapse
|
22
|
Gao J, Feng SS, Guo Y. Nanomedicine for treatment of cancer stem cells. Nanomedicine (Lond) 2014; 9:181-4. [DOI: 10.2217/nnm.13.195] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Jie Gao
- International Joint Cancer Institute, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| | - Si-Shen Feng
- Department of Chemical & Biomolecular Engineering, National University of Singapore, Block E5, 02-11, Engineering Drive 4, 117576, Singapore
| | - Yajun Guo
- International Joint Cancer Institute, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China
| |
Collapse
|
23
|
Noninvasive positron emission tomography and fluorescence imaging of CD133+ tumor stem cells. Proc Natl Acad Sci U S A 2014; 111:E692-701. [PMID: 24469819 DOI: 10.1073/pnas.1314189111] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A technology that visualizes tumor stem cells with clinically relevant tracers could have a broad impact on cancer diagnosis and treatment. The AC133 epitope of CD133 currently is one of the best-characterized tumor stem cell markers for many intra- and extracranial tumor entities. Here we demonstrate the successful noninvasive detection of AC133(+) tumor stem cells by PET and near-infrared fluorescence molecular tomography in subcutaneous and orthotopic glioma xenografts using antibody-based tracers. Particularly, microPET with (64)Cu-NOTA-AC133 mAb yielded high-quality images with outstanding tumor-to-background contrast, clearly delineating subcutaneous tumor stem cell-derived xenografts from surrounding tissues. Intracerebral tumors as small as 2-3 mm also were clearly discernible, and the microPET images reflected the invasive growth pattern of orthotopic cancer stem cell-derived tumors with low density of AC133(+) cells. These data provide a basis for further preclinical and clinical use of the developed tracers for high-sensitivity and high-resolution monitoring of AC133(+) tumor stem cells.
Collapse
|
24
|
O'Connor ML, Xiang D, Shigdar S, Macdonald J, Li Y, Wang T, Pu C, Wang Z, Qiao L, Duan W. Cancer stem cells: A contentious hypothesis now moving forward. Cancer Lett 2013; 344:180-7. [PMID: 24333726 DOI: 10.1016/j.canlet.2013.11.012] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/11/2013] [Accepted: 11/19/2013] [Indexed: 02/07/2023]
Abstract
Cancer stem cells are a progressive concept to account for the cell biological nature of cancer. Despite the controversies regarding the cancer stem cell model, it has the potential to provide a foundation for new innovative treatment targeting the roots of cancer. The last two years have witnessed exceptional progress in cancer stem cell research, in particular on solid tumours, which holds promise for improved treatment outcomes. Here, we review recent advances in cancer stem cell research, discuss challenges in the field and explore future strategies and opportunities in cancer stem cell studies to overcome resistance to chemotherapy.
Collapse
Affiliation(s)
- Michael L O'Connor
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Dongxi Xiang
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Sarah Shigdar
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Joanna Macdonald
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Yong Li
- Cancer Care Centre, St. George Hospital, and St. George Clinical School, Faculty of Medicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Tao Wang
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Chunwen Pu
- Dalian Sixth People's Hospital, Dalian 116033, China
| | - Zhidong Wang
- Dalian Sixth People's Hospital, Dalian 116033, China
| | - Liang Qiao
- Storr Liver Unit, Westmead Millennium Institute, The University of Sydney at the Westmead Hospital, Westmead, NSW 2145, Australia.
| | - Wei Duan
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia.
| |
Collapse
|
25
|
Basuki JS, Duong HTT, Macmillan A, Erlich RB, Esser L, Akerfeldt MC, Whan RM, Kavallaris M, Boyer C, Davis TP. Using fluorescence lifetime imaging microscopy to monitor theranostic nanoparticle uptake and intracellular doxorubicin release. ACS NANO 2013; 7:10175-10189. [PMID: 24131276 DOI: 10.1021/nn404407g] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
We describe the synthesis of iron oxide nanoparticles (IONPs) with excellent colloidal stability in both water and serum, imparted by carefully designed grafted polymer shells. The polymer shells were built with attached aldehyde functionality to enable the reversible attachment of doxorubicin (DOX) via imine bonds, providing a controlled release mechanism for DOX in acidic environments. The IONPs were shown to be readily taken up by cell lines (MCF-7 breast cancer cells and H1299 lung cancer cells), and intracellular release of DOX was proven using in vitro fluorescence lifetime imaging microscopy (FLIM) measurements. Using the fluorescence lifetime difference exhibited by native DOX (~1 ns) compared to conjugated DOX (~4.6 ns), the intracellular release of conjugated DOX was in situ monitored in H1299 and was estimated using phasor plot representation, showing a clear increase of native DOX with time. The results obtained from FLIM were corroborated using confocal microscopy, clearly showing DOX accumulation in the nuclei. The IONPs were also assessed as MRI negative contrast agents. We observed a significant change in the transverse relaxivity properties of the IONPs, going from 220 to 390 mM(-1) s(-1), in the presence or absence of conjugated DOX. This dependence of MRI signal on IONP-DOX/water interactions may be exploited in future theranostic applications. The in vitro studies were then extended to monitor cell uptake of the DOX loaded IONPs (IONP@P(HBA)-b-P(OEGA) + DOX) into two 3D multicellular tumor spheroids (MCS) grown from two independent cell lines (MCF-7 and H1299) using multiphoton excitation microscopy.
Collapse
Affiliation(s)
- Johan S Basuki
- Australian Centre for Nanomedicine, University of New South Wales , Sydney, New South Wales 2052, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
DCA increases the antitumor effects of capecitabine in a mouse B16 melanoma allograft and a human non-small cell lung cancer A549 xenograft. Cancer Chemother Pharmacol 2013; 72:1031-41. [PMID: 24043136 DOI: 10.1007/s00280-013-2281-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/27/2013] [Indexed: 12/26/2022]
Abstract
PURPOSE Capecitabine is one of the few chemotherapy drugs with high oral availability. Recently, sodium dichloroacetate (DCA) has shown great potential as an anticancer agent. In the present study, we assessed the anticancer effect of DCA in combination with capecitabine for cancers that modestly expressed TP. METHODS A mouse B16 melanoma allograft and a human non-small cell lung cancer A549 xenograft were used to assess the effect of DCA and capecitabine combined treatment. Histology and immunohistochemistry were used to detect the apoptosis and proliferation of cancer cells. Real-time PCR and Western blot were carried out to detect the expression of TP and caspases, respectively. RESULTS For the first time, we report that DCA increased the antitumor effects of capecitabine in a mouse B16 allograft and a human A549 xenograft by promoting apoptosis of tumor cells. DCA has little effect on the expression of TP. CONCLUSIONS Our finding suggests that DCA in combination with capecitabine might be potential as a new therapeutic regimen against some cancers.
Collapse
|
27
|
Shigdar S, Li Y, Bhattacharya S, O'Connor M, Pu C, Lin J, Wang T, Xiang D, Kong L, Wei MQ, Zhu Y, Zhou S, Duan W. Inflammation and cancer stem cells. Cancer Lett 2013; 345:271-8. [PMID: 23941828 DOI: 10.1016/j.canlet.2013.07.031] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 07/19/2013] [Accepted: 07/28/2013] [Indexed: 12/18/2022]
Abstract
Cancer stem cells are becoming recognised as being responsible for metastasis and treatment resistance. The complex cellular and molecular network that regulates cancer stem cells and the role that inflammation plays in cancer progression are slowly being elucidated. Cytokines, secreted by tumour associated immune cells, activate the necessary pathways required by cancer stem cells to facilitate cancer stem cells progressing through the epithelial-mesenchymal transition and migrating to distant sites. Once in situ, these cancer stem cells can secrete their own attractants, thus providing an environment whereby these cells can continue to propagate the tumour in a secondary niche.
Collapse
Affiliation(s)
- Sarah Shigdar
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia.
| | - Yong Li
- Cancer Care Centre, St. George Hospital, and St. George Clinical School, Faculty of Medicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Santanu Bhattacharya
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560 012, India
| | - Michael O'Connor
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Chunwen Pu
- Dalian Sixth People's Hospital, Dalian 116033, China
| | - Jia Lin
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Tao Wang
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Dongxi Xiang
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Lingxue Kong
- School of Medical Science and Griffith Health Institute, Griffith University, Gold Coast Campus, Southport, Australia
| | - Ming Q Wei
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria, Australia
| | - Yimin Zhu
- Suzhou Key Laboratory of Nanobiomedicine, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Shufeng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Wei Duan
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia.
| |
Collapse
|
28
|
Noninvasive identification of viable cell populations in docetaxel-treated breast tumors using ferritin-based magnetic resonance imaging. PLoS One 2013; 8:e52931. [PMID: 23301003 PMCID: PMC3534651 DOI: 10.1371/journal.pone.0052931] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 11/22/2012] [Indexed: 12/01/2022] Open
Abstract
Background Cancer stem cells (CSCs) are highly tumorigenic and are responsible for tumor progression and chemoresistance. Noninvasive imaging methods for the visualization of CSC populations within tumors in vivo will have a considerable impact on the development of new CSC-targeting therapeutics. Methodology/Principal Findings In this study, human breast cancer stem cells (BCSCs) transduced with dual reporter genes (human ferritin heavy chain [FTH] and enhanced green fluorescence protein [EGFP]) were transplanted into NOD/SCID mice to allow noninvasive tracking of BCSC-derived populations. No changes in the properties of the BCSCs were observed due to ferritin overexpression. Magnetic resonance imaging (MRI) revealed significantly different signal intensities (R2* values) between BCSCs and FTH-BCSCs in vitro and in vivo. In addition, distinct populations of pixels with high R2* values were detected in docetaxel-treated FTH-BCSC tumors compared with control tumors, even before the tumor sizes changed. Histological analysis revealed that areas showing high R2* values in docetaxel-treated FTH-BCSC tumors by MRI contained EGFP+/FTH+ viable cell populations with high percentages of CD44+/CD24− cells. Conclusions/Significance These findings suggest that ferritin-based MRI, which provides high spatial resolution and tissue contrast, can be used as a reliable method to identify viable cell populations derived from BCSCs after chemotherapy and may serve as a new tool to monitor the efficacy of CSC-targeting therapies in vivo.
Collapse
|