51
|
Phan TTV, Bui NQ, Cho SW, Bharathiraja S, Manivasagan P, Moorthy MS, Mondal S, Kim CS, Oh J. Photoacoustic Imaging-Guided Photothermal Therapy with Tumor-Targeting HA-FeOOH@PPy Nanorods. Sci Rep 2018; 8:8809. [PMID: 29891947 PMCID: PMC5995888 DOI: 10.1038/s41598-018-27204-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/24/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer theragnosis agents with both cancer diagnosis and therapy abilities would be the next generation of cancer treatment. Recently, nanomaterials with strong absorption in near-infrared (NIR) region have been explored as promising cancer theragnosis agents for bio-imaging and photothermal therapy (PTT). Herein, we reported the synthesis and application of a novel multifunctional theranostic nanoagent based on hyaluronan (HA)-coated FeOOH@polypyrrole (FeOOH@PPy) nanorods (HA-FeOOH@PPy NRs) for photoacoustic imaging (PAI)-guided PTT. The nanoparticles were intentionally designed with rod-like shape and conjugated with tumor-targeting ligands to enhance the accumulation and achieve the entire tumor distribution of nanoparticles. The prepared HA-FeOOH@PPy NRs showed excellent biocompatible and physiological stabilities in different media. Importantly, HA-FeOOH@PPy NRs exhibited strong NIR absorbance, remarkable photothermal conversion capability, and conversion stability. Furthermore, HA-FeOOH@PPy NRs could act as strong contrast agents to enhance PAI, conducting accurate locating of cancerous tissue, as well as precise guidance for PTT. The in vitro and in vivo photothermal anticancer activity results of the designed nanoparticles evidenced their promising potential in cancer treatment. The tumor-bearing mice completely recovered after 17 days of PTT treatment without obvious side effects. Thus, our work highlights the great potential of using HA-FeOOH@PPy NRs as a theranostic nanoplatform for cancer imaging-guided therapy.
Collapse
Affiliation(s)
- Thi Tuong Vy Phan
- Interdisciplinary Program of Biomedical Mechanical & Electrical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
- Center for Marine-Integrated Biomedical Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Nhat Quang Bui
- Interdisciplinary Program of Biomedical Mechanical & Electrical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Soon-Woo Cho
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Subramaniyan Bharathiraja
- Center for Marine-Integrated Biomedical Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Panchanathan Manivasagan
- Center for Marine-Integrated Biomedical Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Madhappan Santha Moorthy
- Center for Marine-Integrated Biomedical Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Sudip Mondal
- Center for Marine-Integrated Biomedical Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Chang-Seok Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Junghwan Oh
- Interdisciplinary Program of Biomedical Mechanical & Electrical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
- Center for Marine-Integrated Biomedical Technology, Pukyong National University, Busan, 48513, Republic of Korea.
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
52
|
Effect of multiple cyclic RGD peptides on tumor accumulation and intratumoral distribution of IRDye 700DX-conjugated polymers. Sci Rep 2018; 8:8126. [PMID: 29802410 PMCID: PMC5970177 DOI: 10.1038/s41598-018-26593-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/10/2018] [Indexed: 12/20/2022] Open
Abstract
Strategic delivery of IRDye 700DX (photosensitizer) is a key for improving its effect in photodynamic therapy. In this study, we have synthesized IRDye 700DX-conjugated polymers containing multiple cyclic RGD peptides to deliver IRDye 700DX selectively to tumor cells and tumor-associated blood vessels overexpressing αvβ3 integrin. Our polymer has a backbone of hydrophilic poly(ethylene glycol)-poly(L-glutamic acid) block copolymer, and cyclic RGD peptides are conjugated to side chains of the poly(L-glutamic acid) while IRDye 700DX is conjugated to the terminal of poly(ethylene glycol). The polymers exhibited selective accumulation to the target sites in a subcutaneous solid tumor, and the accumulation was augmented with the increased number of cyclic RGD peptides. More importantly, the polymer containing 15 cyclic RGD peptides in one construct revealed preferential accumulation on the tumor-associated blood vessels without compromising penetration to deep portions of the tumor, thereby drastically inhibiting tumor growth upon photoirradiation, while the polymer containing 5 cyclic RGD peptides showed moderate antitumor activity despite efficient accumulation in the tumor with almost homogenous intratumoral distribution. These results suggest that controlling the intratumoral distribution of IRDye 700DX is critical for successful PDT, and our polymer containing multiple cyclic RGD peptides may be a promising carrier for this spatial control.
Collapse
|
53
|
Fujimoto S, Muguruma N, Okamoto K, Kurihara T, Sato Y, Miyamoto Y, Kitamura S, Miyamoto H, Taguchi T, Tsuneyama K, Takayama T. A Novel Theranostic Combination of Near-infrared Fluorescence Imaging and Laser Irradiation Targeting c-KIT for Gastrointestinal Stromal Tumors. Theranostics 2018; 8:2313-2328. [PMID: 29721082 PMCID: PMC5928892 DOI: 10.7150/thno.22027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 02/27/2018] [Indexed: 01/13/2023] Open
Abstract
It is difficult to distinguish gastrointestinal stromal tumors (GISTs) from other types of submucosal tumors under conventional gastrointestinal endoscopy. We aimed to detect GISTs by molecular fluorescence imaging using a near-infrared (NIR) photosensitizer (IR700)-conjugated anti-c-KIT antibody and to treat GISTs by photoimmunotherapy with NIR irradiation as a non-invasive theranostic procedure. We also investigated the therapeutic mechanisms. Methods: Human GIST cell lines GIST-T1 and GIST-882M were incubated with IR700-conjugated anti-c-KIT antibody, IR700-12A8, and observed by confocal laser microscopy. Mice with GIST-T1 xenografts or rats with orthotopic xenografts were injected with IR700-12A8 or AF488-conjugated antibody, and observed under IVIS or autofluorescence imaging (AFI) endoscopy. GIST cells were treated with IR700-12A8 and NIR light in vitro and vivo, and cell viability, histology and apoptosis were evaluated. Results: Strong red fluorescence of IR700-12A8 was observed on the cell membrane of GIST cells and was gradually internalized into the cytoplasm. Tumor-specific accumulation of IR700-12A8 was observed in GIST-T1 xenografts in mice. Under AFI endoscopy, a strong fluorescence signal was observed in orthotopic GIST xenografts in rats through the normal mucosa covering the tumor. The percentage of dead cells significantly increased in a light-dose-dependent manner and both acute necrotic and late apoptotic cell death was observed with annexin/PI staining. Cleaved PARP expression was significantly increased after IR700-12A8-mediated NIR irradiation, which was almost completely reversed by NaN3. All xenograft tumors (7/7) immediately regressed and 4/7 tumors completely disappeared after IR700-12A8-mediated NIR irradiation. Histologic assessment and TUNEL staining revealed apoptosis in the tumors. Conclusion: NIR fluorescence imaging using IR700-12A8 and subsequent NIR irradiation could be a very effective theranostic technology for GIST, the underlying mechanism of which appears to involve acute necrosis and supposedly late apoptosis induced by singlet oxygen.
Collapse
Affiliation(s)
- Shota Fujimoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naoki Muguruma
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koichi Okamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Kurihara
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yasushi Sato
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoshihiko Miyamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shinji Kitamura
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hiroshi Miyamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takahiro Taguchi
- Division of Human Health & Medical Science, Graduate School of Kuroshio Science, Kochi University, Nankoku, Kochi, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
54
|
Zarrintaj P, Ahmadi Z, Hosseinnezhad M, Saeb MR, Laheurte P, Mozafari M. Photosensitizers in medicine: Does nanotechnology make a difference? ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.matpr.2018.05.082] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
55
|
Zhang C, Gao H, Li C, Tu J, Chen Z, Su W, Geng X, Chen X, Wang J, Pan W. TGFβ1 Promotes Breast Cancer Local Invasion and Liver Metastasis by Increasing the CD44 high/CD24 - Subpopulation. Technol Cancer Res Treat 2018; 17:1533033818764497. [PMID: 29658391 PMCID: PMC5909851 DOI: 10.1177/1533033818764497] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 01/10/2018] [Accepted: 02/16/2018] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Previous studies have shown that the transforming growth factor β1 pathway plays an important role in breast cancer metastasis to the liver. However, the mechanism of this metastasis has not been fully clarified. Cancer stem cells are essential for the initiation and propagation of tumor metastasis. The objective of our current study was to define the role of cancer stem cells in transforming growth factor β1-mediated breast cancer hepatic metastases. METHOD Hematoxylin and eosin staining was used to assess the formation of breast cancer liver metastases and local invasion. Cancer stem cells surface markers (CD44, CD24, and Epithelial cell adhesion molecule [EpCAM]), luminal/mesenchymal markers (keratin8 and alpha smooth muscle actin), and proliferation markers (Ki-67 and cyclinD1) were detected by immunohistochemistry assays. Flow cytometry was used to evaluate the effect of transforming growth factor β1 on the CD44+/CD24- cancer stem cell population. Quantitative real-time polymerase chain reaction was employed to assess the gene expression of the stem cell self-renewal markers nanog, pou5f1 (coding for Oct4), and sox2. RESULTS Transforming growth factor β1 increased the formation of liver metastases by the MDA-MB231 (MDA) breast cancer cell line but did not affect the liver metastasis of CD44+/CD24+ noncancer stem cells. Transforming growth factor β1 treatment did not significantly affect tumor proliferation in vitro or in vivo. Transforming growth factor β1 promoted mammary tumor local invasion. Furthermore, the CD44high/CD24- cancer stem cell population was also significantly increased by transforming growth factor β1 treatment. Besides, the gene expression of the stem cell self-renewal markers (nanog, pou5f1, and sox2) and another stem cell surface marker (EpCAM) was increased by transforming growth factor β1 treatment. Finally, clusters of CD44-positive breast cancer cells were observed in the livers of mice from the control and transforming growth factor β1 pretreatment groups. CONCLUSION Our results indicate that transforming growth factor β1 increases the local invasive capacity and liver metastasis of breast cancer cells by inducing the CD44high/CD24- cancer stem cell population.
Collapse
Affiliation(s)
- Chenjing Zhang
- Department of Gastroenterology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huiqin Gao
- Department of Gastroenterology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- Bengbu Medical College, Bengbu, Anhui, China
| | - Chao Li
- Department of Gastroenterology, Nanjing First Hospital, Nanjing, Jiangsu, China
| | - Jiangfeng Tu
- Department of Gastroenterology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhihao Chen
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weiwei Su
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoge Geng
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaojun Chen
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingya Wang
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wensheng Pan
- Department of Gastroenterology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
56
|
Yu X, Gao D, Gao L, Lai J, Zhang C, Zhao Y, Zhong L, Jia B, Wang F, Chen X, Liu Z. Inhibiting Metastasis and Preventing Tumor Relapse by Triggering Host Immunity with Tumor-Targeted Photodynamic Therapy Using Photosensitizer-Loaded Functional Nanographenes. ACS NANO 2017; 11:10147-10158. [PMID: 28901740 DOI: 10.1021/acsnano.7b04736] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Effective cancer therapy depends not only on destroying the primary tumor but also on conditioning the host immune system to recognize and eliminate residual tumor cells and prevent metastasis. In this study, a tumor integrin αvβ6-targeting peptide (the HK peptide)-functionalized graphene oxide (GO) was coated with a photosensitizer (HPPH). The resulting GO conjugate, GO(HPPH)-PEG-HK, was investigated whether it could destroy primary tumors and boost host antitumor immunity. We found that GO(HPPH)-PEG-HK exhibited significantly higher tumor uptake than GO(HPPH)-PEG and HPPH. Photodynamic therapy (PDT) using GO(HPPH)-PEG suppressed tumor growth in both subcutaneous and lung metastatic mouse models. Necrotic tumor cells caused by GO(HPPH)-PEG-HK PDT activated dendritic cells and significantly prevented tumor growth and lung metastasis by increasing the infiltration of cytotoxic CD8+ T lymphocytes within tumors as evidenced by in vivo optical and single-photon emission computed tomography (SPECT)/CT imaging. These results demonstrate that tumor-targeted PDT using GO(HPPH)-PEG-HK could effectively ablate primary tumors and destroy residual tumor cells, thereby preventing distant metastasis by activating host antitumor immunity and suppressing tumor relapse by stimulation of immunological memory.
Collapse
Affiliation(s)
- Xinhe Yu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Duo Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Liquan Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Jianhao Lai
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Chenran Zhang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Yang Zhao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| | - Lijun Zhong
- Medical and Healthy Analytical Center, Peking University , Beijing 100191, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
- Medical and Healthy Analytical Center, Peking University , Beijing 100191, China
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Zhaofei Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center , Beijing 100191, China
| |
Collapse
|
57
|
Combination of a chemopreventive agent and paclitaxel in CD44-targeted hybrid nanoparticles for breast cancer treatment. Arch Pharm Res 2017; 40:1420-1432. [PMID: 29027133 DOI: 10.1007/s12272-017-0968-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 10/06/2017] [Indexed: 01/20/2023]
Abstract
The CD44 receptor, which is upregulated in many cancer cells, provides a selective cellular surface for targeted drug delivery systems. We developed a hybrid nanocarrier for the CD44-targeted delivery of ibuprofen (IBU) and paclitaxel (PTX). The solid lipid nanoparticles (SLNs) were prepared by a hot-melt oil/water emulsion technique and then coated with hyaluronic acid (HA) by electrostatic interactions. The final SLN were spherical with a hydrodynamic diameter (Z) of 72.16 ± 2.9 nm, polydispersity index (PDI) of 0.276 ± 0.009, and zeta potential (ZP) of 28.20 ± 0.69 mV. Similarly, SLN coated with HA (SLN-HA) exhibited acceptable physical properties (Z 169.3 ± 0.55 nm, PDI 0.285 ± 0.004, and ZP - 10.5 ± 0.15 mV). Cell viability assays showed that the combination of IBU, a chemopreventive agent, and PTX exerted a synergistic inhibitory effect on the proliferation of cancer cells (CI < 1.0). Additionally, our observations indicated that both SLN and SLN-HA enhanced apoptosis and cellular uptake compared to the cocktail of free drugs. HA indicated its affinity for cancer cells through the improvement of cellular uptake and induction of apoptosis. These results clearly indicated that these nanoparticle systems hold great promise for drug delivery in breast cancer treatment.
Collapse
|
58
|
Sha S, Yuan D, Liu Y, Han B, Zhong N. Targeting long non-coding RNA DANCR inhibits triple negative breast cancer progression. Biol Open 2017; 6:1310-1316. [PMID: 28760736 PMCID: PMC5612229 DOI: 10.1242/bio.023135] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancer (TNBC) is non-responsive to conventional anti-hormonal and Her2-targeted therapies, making it necessary to identify new molecular targets for therapy. Long non-coding RNA anti-differentiation ncRNA (lncRNA DANCR) was identified participating in carcinogenesis of hepatocellular carcinoma, but its expression and potential role in TNBC progression is still unclear. In the present study, our results showed that DANCR expression was increased in TNBC tissues compared with the adjacent normal tissues using quantitative real-time PCR (qRT-PCR) in 63 TNBC specimens. Patients with higher DANCR expression correlated with worse TNM stages as well as a shorter overall survival (OS) using Kaplan–Meier analysis. When the endogenous DANCR was knocked-down via specific siRNA, cell proliferation and invasion were decreased obviously in the MDA-MB-231 cells. In vivo xenograft experiments showed that knockdown of the DANCR in MDA-MB-231 cells reduced the tumor growth significantly. Furthermore, a compendium of TNBC cancer stem cell markers such as CD44, ABCG2 transporter and aldehyde dehydrogenase (ALDH1) were greatly downregulated in the MDA-MB-231 cells with DANCR knockdown. Molecular mechanistic studies revealed that knockdown of DANCR was associated with increased binding of EZH2 on the promoters of CD44 and ABCG2, and concomitant reduction of expression of these genes suggested that they may be DANCR targets in TNBC. Thus, our study demonstrated that targeting DANCR expression might be a viable therapeutic approach to treat triple negative breast cancer. Summary: Targeting LncRNA DANCR obstructs triple negative breast cancer progression by down-regulating cancer stem cell marker CD44 and ABCG2.
Collapse
Affiliation(s)
- Sha Sha
- Department of Medical Genetics, Peking University Health Science Center, Beijing, 100038 China.,Department of Medical Science, Xizang Minzu University, Xianyang, Shaanxi Province, 712082 China
| | - Dongya Yuan
- Department of Immunology and Microbiology, Xizang Minzu University, Xianyang, Shaanxi Province, 712082 China
| | - Yuejun Liu
- Department of General Surgery, Haifushan Hospital, Weifang, Shandong, 262605 China
| | - Baosan Han
- Department of General Surgery, Shanghai Jiaotong University Xinhua Hospital, Shanghai, 200240 China
| | - Nanbert Zhong
- Department of Medical Genetics, Peking University Health Science Center, Beijing, 100038 China
| |
Collapse
|
59
|
Railkar R, Krane LS, Li QQ, Sanford T, Siddiqui MR, Haines D, Vourganti S, Brancato SJ, Choyke PL, Kobayashi H, Agarwal PK. Epidermal Growth Factor Receptor (EGFR)-targeted Photoimmunotherapy (PIT) for the Treatment of EGFR-expressing Bladder Cancer. Mol Cancer Ther 2017; 16:2201-2214. [PMID: 28619755 DOI: 10.1158/1535-7163.mct-16-0924] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/05/2017] [Accepted: 06/09/2017] [Indexed: 12/12/2022]
Abstract
The use of light as a means of therapy for bladder cancer has a long history but has been hampered by a lack of tumor specificity and therefore, damage to the normal bladder mucosa. Here, we describe a targeted form of phototherapy called photoimmunotherapy (PIT), which targets EGFR-expressing bladder cancer. Anti-EGFR antibody panitumumab was labeled with the photoabsorber (PA), IRDye 700Dx (IR700), to create a panitumumab-IR700 antibody-PA conjugate that is activated by near-infrared radiation (NIR). Bladder cancer tissue microarray (TMA) and bladder cancer cell lines were analyzed for expression of EGFR. Mechanism of PIT-induced cell death was studied using proliferation assays, transmission electron microscopy (TEM), and production of reactive oxygen species. Finally, the in vivo effect was studied in xenografts. EGFR staining of TMAs showed that while most bladder cancers have expression of EGFR to a varying degree, squamous cell carcinomas (SCC) have the highest expression of EGFR. Panitumumab-IR700 activated by NIR light rapidly killed UMUC-5 cells, a bladder SCC line. Panitumumab alone, panitumumab-IR700 without NIR, or NIR alone had no effect on cells. TEM demonstrated that cell death is due to necrosis. Singlet oxygen species contributed toward cell death. NIR-PIT with panitumumab-IR700 reduced growth compared with only panitumumab-IR700-treated UMUC-5 xenograft tumors. PIT is a new targeted treatment for bladder cancer. Panitumumab-IR700-induced PIT selectively kills EGFR-expressing bladder cancer cells in vitro and in vivo and therefore warrants further therapeutic studies in orthotopic xenografts of bladder cancer and ultimately in patients. Mol Cancer Ther; 16(10); 2201-14. ©2017 AACR.
Collapse
Affiliation(s)
- Reema Railkar
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - L Spencer Krane
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Q Quentin Li
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Thomas Sanford
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Mohammad Rashid Siddiqui
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Diana Haines
- Pathology Section, Pathology/Histotechnology Laboratory, Leidos Biomedical Research, Inc. Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Srinivas Vourganti
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Sam J Brancato
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Piyush K Agarwal
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
60
|
Hartmans E, Linssen MD, Sikkens C, Levens A, Witjes MJ, van Dam GM, Nagengast WB. Tyrosine kinase inhibitor induced growth factor receptor upregulation enhances the efficacy of near-infrared targeted photodynamic therapy in esophageal adenocarcinoma cell lines. Oncotarget 2017; 8:29846-29856. [PMID: 28415738 PMCID: PMC5444708 DOI: 10.18632/oncotarget.16165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/27/2017] [Indexed: 12/12/2022] Open
Abstract
Esophageal carcinoma (EC) is a global health problem, with disappointing 5-year survival rates of only 15-25%. Near-infrared targeted photodynamic therapy (NIR-tPDT) is a novel strategy in which cancer-targeted phototoxicity is able to selectively treat malignant cells. In this in vitro report we demonstrate the applicability of antibody-based NIR-tPDT in esophageal adenocarcinoma (EAC), using the phototoxic compounds cetuximab-IRDye700DX and trastuzumab-IRDye700DX, targeting respectively epidermal growth factor receptor 1 (EGFR) and 2 (HER2). Furthermore, we demonstrate that NIR-tPDT can be made more effective by tyrosine kinase inhibitor (TKI) induced growth receptor upregulation. Together, these results unveil a novel strategy for non-invasive EAC treatment, and by pretreatment-induced receptor upregulation its future clinical application may be optimized.
Collapse
Affiliation(s)
- Elmire Hartmans
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre, Groningen, The Netherlands
| | - Matthijs D. Linssen
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre, Groningen, The Netherlands
| | - Claire Sikkens
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre, Groningen, The Netherlands
| | - Afra Levens
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre, Groningen, The Netherlands
| | - Max J.H. Witjes
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Centre, Groningen, The Netherlands
| | - Gooitzen M. van Dam
- Department of Surgery, Nuclear Medicine and Molecular imaging and Intensive Care, University of Groningen, University Medical Centre, Groningen, The Netherlands
| | - Wouter B. Nagengast
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre, Groningen, The Netherlands
| |
Collapse
|
61
|
Wickens JM, Alsaab HO, Kesharwani P, Bhise K, Amin MCIM, Tekade RK, Gupta U, Iyer AK. Recent advances in hyaluronic acid-decorated nanocarriers for targeted cancer therapy. Drug Discov Today 2017; 22:665-680. [PMID: 28017836 PMCID: PMC5413407 DOI: 10.1016/j.drudis.2016.12.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/28/2016] [Accepted: 12/15/2016] [Indexed: 12/16/2022]
Abstract
The cluster-determinant 44 (CD44) receptor has a high affinity for hyaluronic acid (HA) binding and is a desirable receptor for active targeting based on its overexpression in cancer cells compared with normal body cells. The nanocarrier affinity can be increased by conjugating drug-loaded carriers with HA, allowing enhanced cancer cell uptake via the HA-CD44 receptor-mediated endocytosis pathway. In this review, we discuss recent advances in HA-based nanocarriers and micelles for cancer therapy. In vitro and in vivo experiments have repeatedly indicated HA-based nanocarriers to be a target-specific drug and gene delivery platform with great promise for future applications in clinical cancer therapy.
Collapse
Affiliation(s)
- Jennifer M Wickens
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; Department of Chemistry, Lake Superior State University, 680 W. Easterday Avenue, Sault Ste. Marie, MI 49783, USA
| | - Hashem O Alsaab
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA
| | - Prashant Kesharwani
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; The International Medical University, School of Pharmacy, Department of Pharmaceutical Technology, Jalan Jalil Perkasa 19, 57000 Kuala Lumpur, Malaysia
| | - Ketki Bhise
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education and Research (NIPER), Sarkhej - Gandhinagar Highway, Thaltej, Ahmedabad 380054, Gujarat, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
62
|
Prasad CP, Chaurasiya SK, Guilmain W, Andersson T. WNT5A signaling impairs breast cancer cell migration and invasion via mechanisms independent of the epithelial-mesenchymal transition. J Exp Clin Cancer Res 2016; 35:144. [PMID: 27623766 PMCID: PMC5022188 DOI: 10.1186/s13046-016-0421-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND WNT5A (-/-) mammary tissue has been shown to exhibit increased ductal elongation, suggesting elevated mammary cell migration. Increased epithelial cell migration/invasion has often but not always been linked to the epithelial-mesenchymal transition (EMT). In the current study, we investigated the loss of WNT5A in HB2 human mammary epithelial cells and hypothesized that this loss increased their invasion via the EMT. Based on these results, we postulated that suppression of breast cancer cell migration and invasion by WNT5A is due to EMT reversal. METHODS WNT5A was transiently knocked down using specific siRNAs, whereas WNT5A signaling was induced in MDA-MB468 and MDA-MB231 breast cancer cells by stably transfecting cells with WNT5A or treating them with recombinant WNT5A (rWNT5A). Changes in EMT markers, CD44, pAKT and AKT expression were assessed using Western blotting and immunofluorescence. The physiological relevance of altered WNT5A signaling was assessed using migration and invasion assays. RESULTS WNT5A knockdown in HB2 mammary epithelial cells resulted in EMT-like changes and increased invasiveness, and these changes were partially reversed by the addition of rWNT5A. These data suggest that WNT5A might inhibit breast cancer cell migration and invasion by a similar EMT reversal. Contrary to our expectations, we did not observe any changes in the EMT status of breast cancer cells, either after treatment with rWNT5A or stable transfection with a WNT5A plasmid, despite the parallel WNT5A-induced inhibition of migration and invasion. Instead, we found that WNT5A signaling impaired CD44 expression and its downstream signaling via AKT. Moreover, knocking down CD44 in breast cancer cells using siRNA impaired cell migration and invasion. CONCLUSIONS WNT5A bi-directionally regulates EMT in mammary epithelial cells, thereby affecting their migration and invasion. However, the ability of WNT5A to inhibit breast cancer cell migration and invasion is an EMT-independent mechanism that, at least in part, can be explained by decreased CD44 expression.
Collapse
Affiliation(s)
- Chandra Prakash Prasad
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, SE-20502, Malmö, Sweden.
| | - Shivendra Kumar Chaurasiya
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, SE-20502, Malmö, Sweden.,Present Address: Department of Applied Microbiology, School of Biological Sciences, Dr HS Gour Central University, Sagar, Madhya Pradesh, India
| | - William Guilmain
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, SE-20502, Malmö, Sweden
| | - Tommy Andersson
- Cell and Experimental Pathology, Department of Translational Medicine, Lund University, Clinical Research Centre, Skåne University Hospital, SE-20502, Malmö, Sweden
| |
Collapse
|