51
|
Yu M, Chen Y, Wang Z, Ding X. pHLIP(Var7)-P1AP suppresses tumor cell proliferation in MDA-MB-231 triple-negative breast cancer by targeting protease activated receptor 1. Breast Cancer Res Treat 2020; 180:379-384. [PMID: 32034579 PMCID: PMC7066270 DOI: 10.1007/s10549-020-05560-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/31/2020] [Indexed: 01/17/2023]
Abstract
PURPOSE Protease-activated receptor 1 (PAR1) is a signaling protein ubiquitously present on the surface of tumor cells, and its homologous protein fragment, PAR1-activating peptide (P1AP), can inhibit protein signal transduction of PAR1/G in tumor cells. pH (Low) insertion peptide (pHLIP) can target the acidic tumor microenvironment (TME) and can be used as an excellent carrier to deliver P1AP to tumor cells for therapeutic purposes. METHODS PAR1 expression on the surface of MDA-MB-231 cells and human MCF10A mammary epithelial cells was observed. The binding between fluorescent-labeled pHLIP(Var7)-P1AP and MDA-MB-231 cells under different pH values was analyzed. The effect of pHLIP(Var7)-P1AP on the proliferation of MDA-MB-231 cells was analyzed under the conditions of pH 7.4 and 6.0. RESULTS PAR1 was highly expressed on the surface of MDA-MB-231 cells. In an acidic environment (pH 6.0 and 5.0), fluorescent-labeled pHLIP(Var7)-P1AP and MDA-MB-231 cells had a high binding ability, and the binding ability increased with the decrease in pH. In an acidic environment (pH 6.0), pHLIP(Var7)-P1AP significantly inhibited MDA-MB-231 cell proliferation. With 0.5 μg, 1 μg, 2 μg, 4 μg, and 8 μg of pHLIP(Var7)-P1AP, the cell proliferation inhibition rates were 3.39%, 5.27%, 14.29%, 22.14%, and 35.69%, respectively. CONCLUSION PAR1 was highly expressed on the surface of MDA-MB-231 cells. pHLIP(Var7)-P1AP can effectively target MDA-MB-231 cells in an acidic environment and inhibit the growth of MDA-MB-231 cells by inhibiting the signal transduction of PAR1/G protein.
Collapse
Affiliation(s)
- MingMing Yu
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Rd., Qingdao, 266100, China
| | - YueHua Chen
- Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - ZhenGuang Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Rd., Qingdao, 266100, China.
| | - XiaoDong Ding
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
52
|
Singhvi G, Rapalli VK, Nagpal S, Dubey SK, Saha RN. Nanocarriers as Potential Targeted Drug Delivery for Cancer Therapy. ENVIRONMENTAL CHEMISTRY FOR A SUSTAINABLE WORLD 2020. [DOI: 10.1007/978-3-030-29207-2_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
53
|
Cassim S, Pouyssegur J. Tumor Microenvironment: A Metabolic Player that Shapes the Immune Response. Int J Mol Sci 2019; 21:E157. [PMID: 31881671 PMCID: PMC6982275 DOI: 10.3390/ijms21010157] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023] Open
Abstract
Immune cells survey and patrol throughout the body and sometimes take residence in niche environments with distinct cellular subtypes and nutrients that may fluctuate from those in which they matured. Rooted in immune cell physiology are metabolic pathways and metabolites that not only deliver substrates and energy for growth and survival, but also instruct effector functions and cell differentiation. Unlike cancer cells, immune cells are not subject to a "Darwinian evolutionary pressure" that would allow them to adapt to developing tumors but are often irrevocably affected to local nutrient deprivation. Thus, immune cells must metabolically adapt to these changing conditions in order to perform their necessary functions. On the other hand, there is now a growing appreciation that metabolic changes occurring in cancer cells can impact on immune cell functionality and contribute to tumor immune evasion, and as such, there is a considerable and growing interest in developing techniques that target metabolism for immunotherapy. In this review, we discuss the metabolic plasticity displayed by innate and adaptive immune cells and highlight how tumor-derived lactate and tumor acidity restrict immunity. To our knowledge, this review outlines the most recent insights on how tumor microenvironment metabolically instructs immune responsiveness.
Collapse
Affiliation(s)
- Shamir Cassim
- Department of Medical Biology, Centre Scientifique de Monaco, CSM, 98000 Monaco, Monaco;
| | - Jacques Pouyssegur
- Department of Medical Biology, Centre Scientifique de Monaco, CSM, 98000 Monaco, Monaco;
- University Côte d’Azur, IRCAN, CNRS, Centre A. Lacassagne, 06189 Nice, France
| |
Collapse
|
54
|
Mononuclear but Not Polymorphonuclear Phagocyte Depletion Increases Circulation Times and Improves Mammary Tumor-Homing Efficiency of Donor Bone Marrow-Derived Monocytes. Cancers (Basel) 2019; 11:cancers11111752. [PMID: 31717301 PMCID: PMC6896201 DOI: 10.3390/cancers11111752] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/29/2019] [Accepted: 11/06/2019] [Indexed: 01/05/2023] Open
Abstract
Tumor associated macrophages are an essential part of the tumor microenvironment. Consequently, bone marrow-derived monocytes (BMDMs) are continuously recruited to tumors and are therefore seen as ideal delivery vehicles with tumor-targeting properties. By using immune cell depleting agents and macroscopic in vivo fluorescence imaging, we demonstrated that removal of endogenous monocytes and macrophages (but not neutrophils) leads to an increased tumor accumulation of exogenously administered BMDMs. By means of intravital microscopy (IVM), we confirmed our macroscopic findings on a cellular level and visualized in real time the migration of the donor BMDMs in the tumors of living animals. Moreover, IVM also revealed that clodronate-mediated depletion drastically increases the circulation time of the exogenously administered BMDMs. In summary, these new insights illustrate that impairment of the mononuclear phagocyte system increases the circulation time and tumor accumulation of donor BMDMs.
Collapse
|
55
|
Yu Q, Huang T, Liu C, Zhao M, Xie M, Li G, Liu S, Huang W, Zhao Q. Oxygen self-sufficient NIR-activatable liposomes for tumor hypoxia regulation and photodynamic therapy. Chem Sci 2019; 10:9091-9098. [PMID: 31827751 PMCID: PMC6889832 DOI: 10.1039/c9sc03161h] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/01/2019] [Indexed: 01/25/2023] Open
Abstract
The inherent hypoxic environment in tumors severely resists the efficacy of photodynamic therapy. To address this problem, herein, the strategy of using oxygen self-sufficient liposomes (denoted as CaO2/B1/NH4HCO3 lipo), which contained aza-BODIPY dye (B1) and CaO2 nanoparticles in the hydrophobic layer and NH4HCO3 in the hydrophilic cavity, was presented to overcome hypoxia-associated photodynamic resistance. Under near-infrared (NIR) irradiation, NIR-absorbable B1 was activated to induce hyperthermia and further triggered the decomposition of NH4HCO3. Subsequently, with the aid of NH4HCO3 and CaO2 nanoparticles, oxygen was rapidly and self-sufficiently generated, during which clean by-products were produced. Furthermore, the increased amount of oxygen promoted the singlet oxygen production in the presence of B1, which served as a photosensitizer because of the heavy atom effect. The oxygen self-sufficient system improved the anticancer efficiency and alleviated the hypoxic environment in vivo, which demonstrated a valuable attempt to regulate intratumoral hypoxia and overcome the limitation of current photodynamic therapy systems. To our knowledge, this highlights the first example of using NIR light to activate CaO2 nanoparticle-containing liposomes for the modulation of the hypoxic environment in tumors.
Collapse
Affiliation(s)
- Qi Yu
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
| | - Tianci Huang
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
| | - Chao Liu
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
| | - Menglong Zhao
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
| | - Mingjuan Xie
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
| | - Guo Li
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
| | - Shujuan Liu
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
| | - Wei Huang
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
- Shaanxi Institute of Flexible Electronics (SIFE) , Northwestern Polytechnical University (NPU) , Xi'an 710072 , Shaanxi , P. R. China .
| | - Qiang Zhao
- Key Laboratory for Organic Electronics and Information Displays , Jiangsu Key Laboratory for Biosensors , Institute of Advanced Materials (IAM) , Nanjing University of Posts and Telecommunications (NUPT) , Nanjing 210023 , P. R. China .
| |
Collapse
|
56
|
Houghton PJ, Kurmasheva RT. Challenges and Opportunities for Childhood Cancer Drug Development. Pharmacol Rev 2019; 71:671-697. [PMID: 31558580 PMCID: PMC6768308 DOI: 10.1124/pr.118.016972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer in children is rare with approximately 15,700 new cases diagnosed in the United States annually. Through use of multimodality therapy (surgery, radiation therapy, and aggressive chemotherapy), 70% of patients will be "cured" of their disease, and 5-year event-free survival exceeds 80%. However, for patients surviving their malignancy, therapy-related long-term adverse effects are severe, with an estimated 50% having chronic life-threatening toxicities related to therapy in their fourth or fifth decade of life. While overall intensive therapy with cytotoxic agents continues to reduce cancer-related mortality, new understanding of the molecular etiology of many childhood cancers offers an opportunity to redirect efforts to develop effective, less genotoxic therapeutic options, including agents that target oncogenic drivers directly, and the potential for use of agents that target the tumor microenvironment and immune-directed therapies. However, for many high-risk cancers, significant challenges remain.
Collapse
Affiliation(s)
- Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| |
Collapse
|
57
|
Huai Y, Hossen MN, Wilhelm S, Bhattacharya R, Mukherjee P. Nanoparticle Interactions with the Tumor Microenvironment. Bioconjug Chem 2019; 30:2247-2263. [PMID: 31408324 PMCID: PMC6892461 DOI: 10.1021/acs.bioconjchem.9b00448] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Compared to normal tissues, the tumor microenvironment (TME) has a number of aberrant characteristics including hypoxia, acidosis, and vascular abnormalities. Many researchers have sought to exploit these anomalous features of the TME to develop anticancer therapies, and several nanoparticle-based cancer therapeutics have resulted. In this Review, we discuss the composition and pathophysiology of the TME, introduce nanoparticles (NPs) used in cancer therapy, and address the interaction between the TME and NPs. Finally, we outline both the potential problems that affect TME-based nanotherapy and potential strategies to overcome these challenges.
Collapse
Affiliation(s)
- Yanyan Huai
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Md Nazir Hossen
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Stefan Wilhelm
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73072, United States
| | - Resham Bhattacharya
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Priyabrata Mukherjee
- peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
58
|
Intratumor Heterogeneity in Interstitial Fluid Pressure in Cervical and Pancreatic Carcinoma Xenografts. Transl Oncol 2019; 12:1079-1085. [PMID: 31174058 PMCID: PMC6556493 DOI: 10.1016/j.tranon.2019.05.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 12/22/2022] Open
Abstract
Preclinical studies have suggested that interstitial fluid pressure (IFP) is uniformly elevated in the central region of tumors, whereas clinical studies have revealed that IFP may vary among different measurement sites in the tumor center. IFP measurements are technically difficult, and it has been claimed that the intratumor heterogeneity in IFP reported for human tumors is due to technical problems. The main purpose of this study was to determine conclusively whether IFP may be heterogeneously elevated in the central tumor region, and if so, to reveal possible mechanisms and possible consequences. Tumors of two xenograft models were included in the study: HL-16 cervical carcinoma and Panc-1 pancreatic carcinoma. IFP was measured with Millar SPC 320 catheters in two positions in each tumor and related to tumor histology or the metastatic status of the host mouse. Some tumors of both models showed significant intratumor heterogeneity in IFP, and this heterogeneity was associated with a compartmentalized histological appearance (i.e., the tissue was divided into compartments separated by thick connective tissue bands) in HL-16 tumors and with a dense collagen-I-rich extracellular matrix in Panc-1 tumors, suggesting that these connective tissue structures prevented efficient interstitial convection. Furthermore, some tumors of both models developed lymph node metastases, and of the two IFP values measured in each tumor, only the higher value was significantly higher in metastatic than in non-metastatic tumors, suggesting that metastatic propensity was determined by the tumor region having the highest IFP.
Collapse
|
59
|
Kim A, Ma JY. Piceatannol-3-O-β-D-glucopyranoside (PG) exhibits in vitro anti-metastatic and anti-angiogenic activities in HT1080 malignant fibrosarcoma cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:95-104. [PMID: 30668328 DOI: 10.1016/j.phymed.2018.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Several components isolated from rhubarb, the root of Rheum undulatum L., including emodin, rhein, rhaponticin, and piceatannol, have been reported to induce cell death and inhibit metastasis in various types of cancer. Recently, piceatannol-3-O-β-D-glucopyranoside (PG) isolated from rhubarb was demonstrated to improve vascular dysfunction by inhibiting arginase activity. PURPOSE In this study, we examined the anti-cancer activities of PG, including effects on the proliferation, metastasis, and angiogenesis of endothelial and malignant cancer cells. RESULTS We found that PG did not affect the proliferation of human fibrosarcoma (HT1080) and human umbilical vein endothelial cells (HUVECs) at treatments up to 100 μM. However, PG efficiently suppressed the metastatic ability of HT1080 cells, as determined by scratch wound migration, transwell migration/invasion assay, and three-dimensional (3D) spheroid invasion assay. PG significantly suppressed the phorbol 12-myristate 13-acetate (PMA)-induced increase of matrix metalloproteinase (MMP)-9 expression as well as gelatinolytic MMP-9 activity, which are essential for cancer metastasis. In addition, PG treatment reduced the production of proangiogenic factors in HT1080 cells under normoxic and hypoxic conditions and suppressed hypoxia-induced activation of the hypoxia-inducible factor (HIF)-1α pathway. We also found that HUVEC angiogenic activity, including migration and tubular structure formation, were significantly reduced by PG treatment. Moreover, in an in ovo chick chorioallantoic membrane assay, spontaneous and vascular endothelial growth factor (VEGF)-induced vessel formation were significantly inhibited by PG treatment. CONCLUSION These results collectively indicate that PG has potent anti-metastatic and anti-angiogenic activities with no cytotoxicity. Thus, PG may be useful to limit the hyperplasia of malignant tumors and the spread of cancer to distant secondary organs.
Collapse
Affiliation(s)
- Aeyung Kim
- Clinical Medicine Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
| | - Jin Yeul Ma
- Korean-Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea.
| |
Collapse
|
60
|
Li Y, Du L, Wu C, Yu B, Zhang H, An F. Peptide Sequence-Dominated Enzyme-Responsive Nanoplatform for Anticancer Drug Delivery. Curr Top Med Chem 2019; 19:74-97. [PMID: 30686257 DOI: 10.2174/1568026619666190125144621] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/06/2018] [Accepted: 11/23/2018] [Indexed: 02/08/2023]
Abstract
Enzymatic dysregulation in tumor and intracellular microenvironments has made this property
a tremendously promising responsive element for efficient diagnostics, carrier targeting, and drug
release. When combined with nanotechnology, enzyme-responsive drug delivery systems (DDSs) have
achieved substantial advancements. In the first part of this tutorial review, changes in tumor and intracellular
microenvironmental factors, particularly the enzymatic index, are described. Subsequently, the
peptide sequences of various enzyme-triggered nanomaterials are summarized for their uses in various
drug delivery applications. Then, some other enzyme responsive nanostructures are discussed. Finally,
the future opportunities and challenges are discussed. In brief, this review can provide inspiration and
impetus for exploiting more promising internal enzyme stimuli-responsive nanoDDSs for targeted tumor
diagnosis and treatment.
Collapse
Affiliation(s)
- Yanan Li
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Liping Du
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Chunsheng Wu
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hui Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| |
Collapse
|
61
|
Wiley SZ, Sriram K, Salmerón C, Insel PA. GPR68: An Emerging Drug Target in Cancer. Int J Mol Sci 2019; 20:E559. [PMID: 30696114 PMCID: PMC6386835 DOI: 10.3390/ijms20030559] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 12/19/2022] Open
Abstract
GPR68 (or ovarian cancer G protein-coupled receptor 1, OGR1) is a proton-sensing G-protein-coupled receptor (GPCR) that responds to extracellular acidity and regulates a variety of cellular functions. Acidosis is considered a defining hallmark of the tumor microenvironment (TME). GPR68 expression is highly upregulated in numerous types of cancer. Emerging evidence has revealed that GPR68 may play crucial roles in tumor biology, including tumorigenesis, tumor growth, and metastasis. This review summarizes current knowledge regarding GPR68-its expression, regulation, signaling pathways, physiological roles, and functions it regulates in human cancers (including prostate, colon and pancreatic cancer, melanoma, medulloblastoma, and myelodysplastic syndrome). The findings provide evidence for GPR68 as a potentially novel therapeutic target but in addition, we note challenges in developing drugs that target GPR68.
Collapse
Affiliation(s)
- Shu Z Wiley
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Krishna Sriram
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Cristina Salmerón
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Paul A Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
62
|
Gwee CP, Khoo CH, Yeap SK, Tan GC, Cheah YK. Targeted inactivation of Salmonella Agona metabolic genes by group II introns and in vivo assessment of pathogenicity and anti-tumour activity in mouse model. PeerJ 2019; 7:e5989. [PMID: 30671294 PMCID: PMC6339473 DOI: 10.7717/peerj.5989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
The fight against cancer has been a never-ending battle. Limitations of conventional therapies include lack of selectivity, poor penetration and highly toxic to the host. Using genetically modified bacteria as a tumour therapy agent has gained the interest of scientist from the past few decades. Low virulence and highly tolerability of Salmonella spp. in animals and humans make it as the most studied pathogen with regards to anti-tumour therapy. The present study aims to construct a genetically modified S. Agona auxotroph as an anti-tumour agent. LeuB and ArgD metabolic genes in ΔSopBΔSopD double knockout S. Agona were successfully knocked out using a Targetron gene knockout system. The knockout was confirmed by colony PCR and the strains were characterized in vitro and in vivo. The knockout of metabolic genes causes significant growth defect in M9 minimal media. Quadruple knockout ΔSopBΔSopDΔLeuBΔArgD (BDLA) exhibited lowest virulence among all of the strains in all parameters including bacterial load, immunity profile and histopathology studies. In vivo anti-tumour study on colorectal tumour bearing-BALB/c mice revealed that all strains of S. Agona were able to suppress the growth of the large solid tumour as compared with negative control and ΔLeuBΔArgD (LA) and BDLA auxotroph showed better efficacy. Interestingly, higher level of tumour growth suppression was noticed in large tumour. However, multiple administration of bacteria dosage did not increase the tumour suppression efficacy. In this study, the virulence of BDLA knockout strain was slightly reduced and tumour growth suppression efficacy was successfully enhanced, which provide a valuable starting point for the development of S. Agona as anti-tumour agent.
Collapse
Affiliation(s)
- Chin Piaw Gwee
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Chai Hoon Khoo
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Selangor, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Yoke Kqueen Cheah
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
63
|
Li J, Chen H, Zeng L, Rees TW, Xiong K, Chen Y, Ji L, Chao H. Mitochondria-targeting cyclometalated iridium(iii) complexes for tumor hypoxic imaging and therapy. Inorg Chem Front 2019. [DOI: 10.1039/c9qi00081j] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The organometallic anthraquinone iridium(iii) complexes display an efficient turn-on phosphorescence response to hypoxia. The complexes can induce cell apoptosis in HeLa cells via mitochondrial dysfunction and caspase-3 activation making them excellent candidates as theranostic agents for hypoxic cancer cells.
Collapse
Affiliation(s)
- Jia Li
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- P. R. China
| | - Hongmin Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- P. R. China
| | - Leli Zeng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- P. R. China
| | - Thomas W. Rees
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- P. R. China
| | - Kai Xiong
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- P. R. China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- P. R. China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- P. R. China
| |
Collapse
|
64
|
Abouelmagd SA, Ellah NHA, Hamid BNAE. Temperature and pH dual-stimuli responsive polymeric carriers for drug delivery. STIMULI RESPONSIVE POLYMERIC NANOCARRIERS FOR DRUG DELIVERY APPLICATIONS 2019:87-109. [DOI: 10.1016/b978-0-08-101995-5.00003-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
65
|
Kim A, Ma JY. Isoliquiritin Apioside Suppresses in vitro Invasiveness and Angiogenesis of Cancer Cells and Endothelial Cells. Front Pharmacol 2018; 9:1455. [PMID: 30618749 PMCID: PMC6295464 DOI: 10.3389/fphar.2018.01455] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/28/2018] [Indexed: 12/23/2022] Open
Abstract
Several components isolated from Glycyrrhizae radix rhizome (GR), including glycyrrhizin, liquiritin, and liquiritigenin, have been shown to induce cancer cell death and inhibit cancer metastasis. Isoliquiritin apioside (ISLA), a component isolated from GR, has been effective for treating tetanic contraction and genotoxicity. However, the effects of ISLA on the metastasis and angiogenesis of malignant cancer cells and endothelial cells (ECs) have not been reported. In this study, we found that up to 100 μM ISLA did not affect cell proliferation but efficiently suppressed the metastatic ability of HT1080 cells, as assessed by scratch-wound migration, Transwell® migration, scratch-wound invasion, Transwell® invasion, and three-dimensional spheroid invasion. ISLA significantly decreased phorbol 12-myristate 13-acetate (PMA)-induced increases in matrix metalloproteinase (MMP) activities and suppressed PMA-induced activation of mitogen-activated protein kinase as well as NF-κB, which are involved in cancer metastasis. In addition, ILSA treatment reduced the production of pro-angiogenic factors in HT1080 cells, including MMP-9, placental growth factor, and vascular endothelial growth factor under normoxia as well as hypoxia conditions, by impairing the hypoxia-inducible factor-1α pathway. We also found that the abilities of human umbilical vein ECs to migrate across the Transwell® and to form tube-like structures were significantly reduced by ISLA treatment. Moreover, using the chorioallantoic membrane assay, vessel formation with or without vascular endothelial growth factor was significantly suppressed by ISLA. These results suggested that ISLA possesses anti-metastatic and anti-angiogenic abilities in malignant cancer cells and ECs, with no cytotoxicity. ISLA may therefore be a safe and effective lead compound to develop anti-cancer drug for limiting the spread of primary tumors to distant organs to form secondary tumors.
Collapse
Affiliation(s)
- Aeyung Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, South Korea
| | - Jin Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, South Korea
| |
Collapse
|
66
|
Yang Y, Zhu W, Feng L, Chao Y, Yi X, Dong Z, Yang K, Tan W, Liu Z, Chen M. G-Quadruplex-Based Nanoscale Coordination Polymers to Modulate Tumor Hypoxia and Achieve Nuclear-Targeted Drug Delivery for Enhanced Photodynamic Therapy. NANO LETTERS 2018; 18:6867-6875. [PMID: 30303384 DOI: 10.1021/acs.nanolett.8b02732] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Photodynamic therapy (PDT) is a light-triggered therapy used to kill cancer cells by producing reactive oxygen species (ROS). Herein, a new kind of DNA nanostructure based on the coordination between calcium ions (Ca2+) and AS1411 DNA G quadruplexes to form nanoscale coordination polymers (NCPs) is developed via a simple method. Both chlorine e6 (Ce6), a photosensitizer, and hemin, an iron-containing porphyrin, can be inserted into the G-quadruplex structure in the obtained NCPs. With further polyethylene glycol (PEG) modification, we obtain Ca-AS1411/Ce6/hemin@pHis-PEG (CACH-PEG) NCP nanostructure that enables the intranuclear transport of photosensitizer Ce6 to generate ROS inside cell nuclei that are the most vulnerable to ROS. Meanwhile, the inhibition of antiapoptotic protein B-cell lymphoma 2 (Bcl-2) expression by AS1411 allows for greatly improved PDT-induced cell apoptosis. Furthermore, the catalase-mimicking DNAzyme function of G-quadruplexes and hemin in those NCPs could decompose tumor endogenous H2O2 to in situ generate oxygen so as to further enhance PDT by overcoming the hypoxia-associated resistance. This work develops a simple yet general method with which to fabricate DNA-based NCPs and presents an interesting concept of a nanoscale drug-delivery system that could achieve the intranuclear delivery of photosensitizers, the down-regulation of anti-apoptotic proteins, and the modulation of the unfavorable tumor microenvironment simultaneously for improved cancer therapy.
Collapse
Affiliation(s)
- Yu Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences , University of Macau , Avenida da Universidade , Taipa , Macau , China
| | - Wenjun Zhu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM) , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM) , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Yu Chao
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM) , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Xuan Yi
- School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X) , Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Soochow University , Suzhou , Jiangsu 215123 , China
| | - Ziliang Dong
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM) , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Kai Yang
- School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X) , Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Soochow University , Suzhou , Jiangsu 215123 , China
| | - Weihong Tan
- Center for Research at Bio/Nano Interface, Department of Chemistry, Department of Physiology and Functional Genomics, Health Cancer Center, UF Genetics Institute, McKnight Brain Institute , University of Florida , Gainesville , Florida 32611 , United States
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM) , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences , University of Macau , Avenida da Universidade , Taipa , Macau , China
| |
Collapse
|
67
|
Targeting the invincible barrier for drug delivery in solid cancers: interstitial fluid pressure. Oncotarget 2018; 9:35723-35725. [PMID: 30515264 PMCID: PMC6254664 DOI: 10.18632/oncotarget.26267] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/06/2018] [Indexed: 12/17/2022] Open
Abstract
Although a number of new systemic therapeutic options in patients with advanced solid cancers have emerged due to the improved knowledge of molecular dysregulation in cancers, the durable, long-term, objective responses infrequently occur. This editorial article highlights the major limitation of current systemic therapy due to an inefficient drug delivery. While several mechanisms contributing to cancer drug resistance have been described, the common key barrier among solid cancers is the unique tumor microenvironment that causes the high interstitial fluid pressure (IFP). We discussed the mechanism causing an elevated IFP and how it interferes with drug delivery. To target the high IFP, we demonstrated the novel approach using gold nanoparticle carrying recombinant human tumor necrosis factor (TNF), a vascular disrupting agent, that preferentially and specifically targets tumors while the systemic toxicity is markedly reduced. The addition of cytotoxic agent by either directly conjugating to the gold nanoparticle or by systemic administration following gold nanoparticle carrying TNF resulted in significantly reduced tumor burden and increased survival in multiple mouse models with primary and metastatic endocrine cancer and pancreatic ductal carcinoma. A clinical trial in patients with advanced solid cancers is warranted based on the promising results in preclinical studies.
Collapse
|
68
|
Zhang X, Ding K, Wang J, Li X, Zhao P. Chemoresistance caused by the microenvironment of glioblastoma and the corresponding solutions. Biomed Pharmacother 2018; 109:39-46. [PMID: 30391707 DOI: 10.1016/j.biopha.2018.10.063] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary human brain tumor. Although comprehensive therapies combining radiotherapy and chemotherapy after surgery can prolong survival, the prognosis is still poor with a median survival of only 14.6 months. Chemoresistance is one of the major causes of relapse as well as poor survival in glioma patients. Therefore, novel strategies to overcome chemoresistance are desperately needed for improved treatment of human GBM. Recent studies have demonstrated that the tumor microenvironment plays a critical role in the chemoresistance of various tumor types, which makes it a suitable target in anti-cancer therapies, as well as a valuable biomarker for prognostic purposes. This review focuses on chemoresistance in GBM induced by stromal cells, including the endothelium of blood vessels, astrocytes, and myeloid cells, as well as non-cellular factors in the tumor microenvironment. Corresponding therapies are discussed, including progressive strategies involving 3-dimensional models integrating engineering as well as biological advances.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Kaikai Ding
- Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, PR China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Peng Zhao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China.
| |
Collapse
|
69
|
Simonsen TG, Lund KV, Hompland T, Kristensen GB, Rofstad EK. DCE-MRI–Derived Measures of Tumor Hypoxia and Interstitial Fluid Pressure Predict Outcomes in Cervical Carcinoma. Int J Radiat Oncol Biol Phys 2018; 102:1193-1201. [DOI: 10.1016/j.ijrobp.2018.04.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/05/2018] [Accepted: 04/12/2018] [Indexed: 12/25/2022]
|
70
|
Samykutty A, Grizzle WE, Fouts BL, McNally MW, Chuong P, Thomas A, Chiba A, Otali D, Woloszynska A, Said N, Frederick PJ, Jasinski J, Liu J, McNally LR. Optoacoustic imaging identifies ovarian cancer using a microenvironment targeted theranostic wormhole mesoporous silica nanoparticle. Biomaterials 2018; 182:114-126. [PMID: 30118979 PMCID: PMC6289590 DOI: 10.1016/j.biomaterials.2018.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/12/2022]
Abstract
At the intersection of the newly emerging fields of optoacoustic imaging and theranostic nanomedicine, promising clinical progress can be made in dismal prognosis of ovarian cancer. An acidic pH targeted wormhole mesoporous silica nanoparticle (V7-RUBY) was developed to serve as a novel tumor specific theranostic nanoparticle detectable using multispectral optoacoustic tomographic (MSOT) imaging. We report the synthesis of a small, < 40 nm, biocompatible asymmetric wormhole pore mesoporous silica core particle that has both large loading capacity and favorable release kinetics combined with tumor-specific targeting and gatekeeping. V7-RUBY exploits the acidic tumor microenvironment for tumor-specific targeting and tumor-specific release. In vitro, treatment with V7-RUBY containing either paclitaxel or carboplatin resulted in increased cell death at pH 6.6 in comparison to drug alone (p < 0.0001). In orthotopic ovarian xenograft mouse models, V7-RUBY containing IR780 was specifically detected within the tumor 7X and 4X higher than the liver and >10X higher than in the kidney using both multispectral optoacoustic tomography (MSOT) imaging with secondary confirmation using near infrared fluorescence imaging (p < 0.0004). The V7-RUBY system carrying a cargo of either contrast agent or an anti-neoplastic drug has the potential to become a theranostic nanoparticle which can improve both diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Abhilash Samykutty
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27013, USA
| | - William E Grizzle
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Benjamin L Fouts
- Department of Chemistry, Earlham College, Indianapolis, IN, 27013, USA
| | - Molly W McNally
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27013, USA
| | - Phillip Chuong
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Alexandra Thomas
- Department of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27013, USA
| | - Akiko Chiba
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27013, USA
| | - Dennis Otali
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Anna Woloszynska
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Neveen Said
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27013, USA
| | - Peter J Frederick
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Jacek Jasinski
- Conn Center Materials Characterization, University of Louisville, Louisville, KY 40202, USA
| | - Jie Liu
- Department of Forest Materials, North Carolina State University, Raleigh, NC 27695, USA
| | - Lacey R McNally
- Department of Bioengineering, Wake Forest School of Medicine, Winston-Salem, North Carolina 27013, USA; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27013, USA.
| |
Collapse
|
71
|
Souho T, Lamboni L, Xiao L, Yang G. Cancer hallmarks and malignancy features: Gateway for improved targeted drug delivery. Biotechnol Adv 2018; 36:1928-1945. [DOI: 10.1016/j.biotechadv.2018.08.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/22/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022]
|
72
|
Fernandes C, Suares D, Yergeri MC. Tumor Microenvironment Targeted Nanotherapy. Front Pharmacol 2018; 9:1230. [PMID: 30429787 PMCID: PMC6220447 DOI: 10.3389/fphar.2018.01230] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Recent developments in nanotechnology have brought new approaches to cancer diagnosis and therapy. While enhanced permeability and retention effect promotes nano-chemotherapeutics extravasation, the abnormal tumor vasculature, high interstitial pressure and dense stroma structure limit homogeneous intratumoral distribution of nano-chemotherapeutics and compromise their imaging and therapeutic effect. Moreover, heterogeneous distribution of nano-chemotherapeutics in non-tumor-stroma cells damages the non-tumor cells, and interferes with tumor-stroma crosstalk. This can lead not only to inhibition of tumor progression, but can also paradoxically induce acquired resistance and facilitate tumor cell proliferation and metastasis. Overall, the tumor microenvironment plays a vital role in regulating nano-chemotherapeutics distribution and their biological effects. In this review, the barriers in tumor microenvironment, its consequential effects on nano-chemotherapeutics, considerations to improve nano-chemotherapeutics delivery and combinatory strategies to overcome acquired resistance induced by tumor microenvironment have been summarized. The various strategies viz., nanotechnology based approach as well as ligand-mediated, redox-responsive, and enzyme-mediated based combinatorial nanoapproaches have been discussed in this review.
Collapse
Affiliation(s)
| | | | - Mayur C Yergeri
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies - NMIMS, Mumbai, India
| |
Collapse
|
73
|
Parashar P, Tripathi CB, Arya M, Kanoujia J, Singh M, Yadav A, Kumar A, Guleria A, Saraf SA. Biotinylated naringenin intensified anticancer effect of gefitinib in urethane-induced lung cancer in rats: favourable modulation of apoptotic regulators and serum metabolomics. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S598-S610. [DOI: 10.1080/21691401.2018.1505738] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Poonam Parashar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Chandra Bhushan Tripathi
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Malti Arya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Jovita Kanoujia
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Mahendra Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Abhishek Yadav
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Amit Kumar
- Centre of Biomedical Research, Lucknow, India
| | | | - Shubhini A. Saraf
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| |
Collapse
|
74
|
Klein D. The Tumor Vascular Endothelium as Decision Maker in Cancer Therapy. Front Oncol 2018; 8:367. [PMID: 30250827 PMCID: PMC6139307 DOI: 10.3389/fonc.2018.00367] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
Genetic and pathophysiologic criteria prearrange the uncontrolled growth of neoplastic cells that in turn initiates new vessel formation, which is prerequisite for further tumor growth and progression. This first endothelial lining is patchy, disordered in structure and thus, angiogenic tumor vessels were proven to be functionally inferior. As a result, tumors were characterized by areas with an apparent oversupply in addition to areas with an undersupply of vessels, which complicates an efficient administration of intravenous drugs in cancer therapy and might even lower the response e.g. of radiotherapy (RT) because of the inefficient oxygen supply. In addition to the vascular dysfunction, tumor blood vessels contribute to the tumor escape from immunity by the lack of response to inflammatory activation (endothelial anergy) and by repression of leukocyte adhesion molecule expression. However, tumor vessels can remodel by the association with and integration of pericytes and smooth muscle cells which stabilize these immature vessels resulting in normalization of the vascular structures. This normalization of the tumor vascular bed could improve the efficiency of previously established therapeutic approaches, such as chemo- or radiotherapy by a more homogenous drug and oxygen distribution, and/or by overcoming endothelial anergy. This review highlights the current investigations that take advantage of a proper vascular function for improving cancer therapy with a special focus on the endothelial-immune system interplay.
Collapse
Affiliation(s)
- Diana Klein
- Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
75
|
Paramita P, Subramaniam VD, Murugesan R, Gopinath M, Ramachandran I, Ramalingam S, Sun XF, Banerjee A, Marotta F, Pathak S. Evaluation of potential anti-cancer activity of cationic liposomal nanoformulated Lycopodium clavatum in colon cancer cells. IET Nanobiotechnol 2018; 12:727-732. [PMID: 30104445 PMCID: PMC8675948 DOI: 10.1049/iet-nbt.2017.0106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 02/12/2018] [Accepted: 02/25/2018] [Indexed: 02/05/2023] Open
Abstract
Research dealing with early diagnosis and efficient treatment in colon cancer to improve patient's survival is still under investigation. Chemotherapeutic agent result in high systemic toxicity due to their non-specific actions on DNA repair and/or cell replication. Traditional medicine such as Lycopodium clavatum (LC) has been claimed to have therapeutic potentials against cancer. The present study focuses on targeted drug delivery of cationic liposomal nanoformulated LC (CL-LC) in colon cancer cells (HCT15) and comparing the efficacy with an anti-colon cancer drug, 7-ethyl-10-hydroxy-camptothecin (SN38) along with its nanoformulated form (CL-SN38). The colloidal suspension of LC was made using thin film hydration method. The drugs were characterised using ultraviolet, dynamic light scattering, scanning electron microscopy, energy, dispersive X-ray spectroscopy. Invitro drug release showed kinetics of 49 and 89% of SN38 and LC, whereas CL-SN38 and CL-LC showed 73 and 74% of sustained drug release, respectively. Studies on morphological changes, cell viability, cytotoxicity, apoptosis, cancer-associated gene expression analysis of Bcl-2, Bax, p53 by real-time polymerase chain reaction and western blot analysis of Bad and p53 protein were performed. Nanoformulated LC significantly inhibited growth and increased the apoptosis of colon cancer cells indicating its potential anti-cancer activity against colon cancer cells.
Collapse
Affiliation(s)
- Pragyan Paramita
- Department of Medical Biotechnology, Chettinad Academy of Research and Education, Kelambakkam 603 103, Tamil Nadu, India
| | - Vimala Devi Subramaniam
- Department of Medical Biotechnology, Chettinad Academy of Research and Education, Kelambakkam 603 103, Tamil Nadu, India
| | - Ramachandran Murugesan
- Department of Medical Biotechnology, Chettinad Academy of Research and Education, Kelambakkam 603 103, Tamil Nadu, India
| | - Madhumala Gopinath
- Department of Medical Biotechnology, Chettinad Academy of Research and Education, Kelambakkam 603 103, Tamil Nadu, India
| | - Ilangovan Ramachandran
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600 113, Tamil Nadu, India
| | - Satish Ramalingam
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram 603 203, Tamil Nadu, India
| | - Xiao Feng Sun
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Antara Banerjee
- Department of Medical Biotechnology, Chettinad Academy of Research and Education, Kelambakkam 603 103, Tamil Nadu, India
| | | | - Surajit Pathak
- Department of Medical Biotechnology, Chettinad Academy of Research and Education, Kelambakkam 603 103, Tamil Nadu, India.
| |
Collapse
|
76
|
Kim A, Ma JY. Rhaponticin decreases the metastatic and angiogenic abilities of cancer cells via suppression of the HIF‑1α pathway. Int J Oncol 2018; 53:1160-1170. [PMID: 30015877 PMCID: PMC6065401 DOI: 10.3892/ijo.2018.4479] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
Rhaponticin (RA; 3′5-dihydroxy-4′-methoxystilbene 3-O-β-D-glucopyranoside) is a component isolated from various medicinal herbs including Rheum undulatum L. RA has been reported to be an effective treatment for allergy, diabetes, thrombosis, liver steatosis, lung fibrosis and colitis. In addition, RA effectively inhibits tumor growth and induces apoptosis; however, the effects of RA, at non-cytotoxic doses, on the metastasis and angiogenesis of malignant cancer cells have, to be the best of our knowledge, not been identified. In the present study, it was identified that RA suppressed the metastatic potential of MDA-MB231 breast cancer cells, including colony formation, migration and invasion. Human umbilical vein endothelial cells (HUVECs) treated with RA exhibited a decreased ability to form tube-like networks and to migrate across a Transwell membrane, when compared with RA-untreated HUVECs. Using the chick chorioallantoic membrane assay, RA treatment significantly suppressed spontaneous and vascular endothelial growth factor (VEGF)-induced angiogenesis. Furthermore, RA inhibited the production of pro-angiogenic factors, including matrix metalloproteinase (MMP)-9, pentraxin-3, interleukin-8, VEGF and placental growth factor under normoxic and hypoxic conditions, and suppressed the phorbol 12-myristate 13-acetate-induced increase in the gelatinolytic MMP-9 activity and MMP-9 expression in HT1080 cells. RA also significantly inhibited the hypoxia-inducible factor (HIF)-1α pathway, leading to decreased HIF-1α accumulation and HIF-1α nuclear expression under hypoxia. These results indicated that RA exhibits potent anti-metastatic and anti-angiogenic activities with no cytotoxicity via suppression of the HIF-1α signaling pathway. Thus, RA may control malignant cancer cells by inhibiting the spread from primary tumors and expansion to distant organs.
Collapse
Affiliation(s)
- Aeyung Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701‑300, Republic of Korea
| | - Jin Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701‑300, Republic of Korea
| |
Collapse
|
77
|
Carter LM, Poty S, Sharma SK, Lewis JS. Preclinical optimization of antibody-based radiopharmaceuticals for cancer imaging and radionuclide therapy-Model, vector, and radionuclide selection. J Labelled Comp Radiopharm 2018; 61:611-635. [PMID: 29412489 PMCID: PMC6081268 DOI: 10.1002/jlcr.3612] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/19/2017] [Accepted: 01/16/2018] [Indexed: 12/25/2022]
Abstract
Intact antibodies and their truncated counterparts (eg, Fab, scFv fragments) are generally exquisitely specific and selective vectors, enabling recognition of individual cancer-associated molecular phenotypes against a complex and dynamic biomolecular background. Complementary alignment of these advantages with unique properties of radionuclides is a defining paradigm in both radioimmunoimaging and radioimmunotherapy, which remain some of the most adept and promising tools for cancer diagnosis and treatment. This review discusses how translational potency can be maximized through rational selection of antibody-nuclide couples for radioimmunoimaging/therapy in preclinical models.
Collapse
Affiliation(s)
- Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sophie Poty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Departments of Radiology and Pharmacology, Weill Cornell Medical College, New York, New York, USA
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
78
|
El-Sayed NS, Miyake T, Shirazi AN, Park SE, Clark J, Buchholz S, Parang K, Tiwari R. Design, Synthesis, and Evaluation of Homochiral Peptides Containing Arginine and Histidine as Molecular Transporters. Molecules 2018; 23:1590. [PMID: 29966296 PMCID: PMC6100079 DOI: 10.3390/molecules23071590] [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: 05/03/2018] [Revised: 06/20/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022] Open
Abstract
Linear (HR)n and cyclic [HR]n peptides (n = 4,5) containing alternate arginine and histidine residues were synthesized. The peptides showed 0⁻15% cytotoxicity at 5⁻100 µM in human ovarian adenocarcinoma (SK-OV-3) cells while they exhibited 0⁻12% toxicity in human leukemia cancer cell line (CCRF-CEM). Among all peptides, cyclic [HR]₄ peptide was able to improve the delivery of a cell impermeable fluorescence-labeled phosphopeptide by two-fold. Fatty acids of different alkyl chain length were attached at the N-terminal of the linear peptide (HR)₄ to improve the molecular transporter property. Addition of fatty acyl chains was expected to help with the permeation of the peptides through the cell membrane. Thus, we synthesized seven fatty acyl derivatives of the linear (HR)₄ peptide. The peptides were synthesized using Fmoc/tBu solid phase peptide chemistry, purified by reverse-phase high-performance liquid chromatography (RP-HPLC) and characterized by matrix-assisted laser desorption/ionization (MALDI) spectrometry. The fatty acyl peptides containing C₈, C12, C14, and C18 alkyl chain did not show cytotoxicity on SK-OV-3 or CCRF-CEM cell lines up to 50 µM concentration; however, at higher concentration (100 µM), they showed mild cytotoxicity. For example, C16-(HR)₄ was also found to reduce the proliferation of SK-OV-3 cells by 11% at 50 µM and C20-(HR)₄ showed mild toxicity at 10 µM, reducing the proliferation of SK-OV-3 cells by 21%. Increase in the length of alkyl chain showed cytotoxicity to the cell lines. C20-(HR)₄ peptide showed better efficiency in translocation of F′-GpYEEI to SK-OV-3 than the phosphopeptide alone. Further investigation of C20-(HR)₄ peptide efficacy showed that the peptide could deliver doxorubicin and epirubicin into SK-OV-3 and also improved the drug antiproliferative ability. These studies provided insights into understanding the structural requirements for optimal cellular delivery of the fatty acyl-(HR)₄ peptide conjugates.
Collapse
Affiliation(s)
- Naglaa Salem El-Sayed
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
- Cellulose and Paper Department, National Research Center, Dokki, Cairo 12622, Egypt.
| | - Taryn Miyake
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| | - Amir N Shirazi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| | - Shang Eun Park
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| | - Jimmy Clark
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| | - Stephani Buchholz
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| | - Rakesh Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA.
| |
Collapse
|
79
|
The homeostasis-maintaining metabolites from bacterial stress response to bacteriophage infection suppress tumor metastasis. Oncogene 2018; 37:5766-5779. [PMID: 29925861 DOI: 10.1038/s41388-018-0376-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/26/2018] [Accepted: 05/29/2018] [Indexed: 12/18/2022]
Abstract
The antiviral metabolites from bacterial stress response to bacteriophage infection can maintain homeostasis of host cells, while metabolism disorder is a remarkable characteristic of tumorigenesis. In the aspect of metabolic homeostasis, therefore, the antiviral homeostasis-maintaining metabolites of bacteria may possess anti-tumor activity. However, this issue has not been addressed. Here we show that the homeostasis-challenged maintaining metabolites from deep-sea bacteriophage-challenged thermophile can suppress tumor metastasis. The results indicated that the metabolic profiles of the bacteriophage GVE2-infected and virus-free thermophile Geobacillus sp. E263 from a deep-sea hydrothermal vent were remarkably different. Thirteen metabolites were significantly elevated and two metabolites were downregulated in thermophile stress response to GVE2 infection. As an example, the upregulated L-norleucine was characterized. The data showed that L-norleucine had antiviral activity in thermophile. Furthermore, the in vitro and in vivo assays revealed that L-norleucine, as well as its derivative, significantly suppressed metastasis of gastric and breast cancer cells. L-norleucine interacted with hnRNPA2/B1 protein to inhibit the expressions of Twist1 and Snail, two inhibitors of E-cadherin, and promote the E-cadherin expression, leading to the inhibition of tumor metastasis. Therefore, our study presented that antiviral homeostasis-maintaining metabolites of microbes might be a promising source for anti-tumor drugs.
Collapse
|
80
|
Chen Y, Bian X, Aliru M, Deorukhkar AA, Ekpenyong O, Liang S, John J, Ma J, Gao X, Schwartz J, Singh P, Ye Y, Krishnan S, Xie H. Hypoxia-targeted gold nanorods for cancer photothermal therapy. Oncotarget 2018; 9:26556-26571. [PMID: 29899876 PMCID: PMC5995181 DOI: 10.18632/oncotarget.25492] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/08/2018] [Indexed: 11/25/2022] Open
Abstract
Tumor hypoxia is a well-recognized driver of resistance to traditional cancer therapies such as chemotherapy and radiation therapy. We describe development of a new nanoconstruct composed of gold nanorods (GNRs) conjugated to carbonic anhydrase IX (CAIX) antibody that specifically binds to CAIX, a biomarker of hypoxia, to facilitate targeting tumor hypoxic areas for focused photothermal ablation. Physicochemical characterization studies confirmed the size, shape, monodispersity, surface charge, and serum stability of the GNRs. Enzyme-linked immunosorbent assays and cellular binding and uptake studies confirmed successful conjugation of antibody to the GNRs and specificity for CAIX. Near-infrared irradiation of CAIX-overexpressing cells treated with GNR/anti-CAIX resulted in significantly higher cell death than cells treated with control GNRs. In vivo biodistribution studies using hyperspectral imaging and inductively coupled plasma mass spectrometry confirmed intravenous administration results not only in greater accumulation of GNR/anti-CAIX in tumors than control GNRs but also greater penetration into hypoxic areas of tumors. Near-infrared ablation of these tumors showed no tumor regression in the sham-treated group, regression but recurrence in the non-targeted-GNR group, and complete tumor regression in the targeted-GNR group. GNR/anti-CAIX nanoconstructs show promise as hypoxia targeting and photothermal ablation agents for cancer treatment.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Xiaomei Bian
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Maureen Aliru
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Amit A Deorukhkar
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Oscar Ekpenyong
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Su Liang
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Jyothy John
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Jing Ma
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Xiuqing Gao
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - Jon Schwartz
- Nanospectra Biosciences, Inc., Houston, Texas, USA
| | - Pankaj Singh
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Yuanqing Ye
- Department of Epidemiology, Division of OVP, Cancer Prevention and Population Science, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Sunil Krishnan
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Huan Xie
- Department of Pharmaceutical and Environmental Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| |
Collapse
|
81
|
Tang J, Howard CB, Mahler SM, Thurecht KJ, Huang L, Xu ZP. Enhanced delivery of siRNA to triple negative breast cancer cells in vitro and in vivo through functionalizing lipid-coated calcium phosphate nanoparticles with dual target ligands. NANOSCALE 2018; 10:4258-4266. [PMID: 29436549 DOI: 10.1039/c7nr08644j] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The conjugation of ligands to nanoparticle platforms for the target delivery of therapeutic agents to the tumor tissue is one of the promising anti-cancer strategies. However, conventional nanoparticle platforms are not so effective in terms of the selectivity and transfection efficiency. In this study, we designed and developed a dual-target drug/gene delivery system based on lipid-coated calcium phosphate (LCP) nanoparticles (NPs) for significantly enhanced siRNA cellular uptake and transfection efficiency. LCP NPs loaded with therapeutic siRNA were conjugated with a controlled number of folic acid and/or EGFR-specific single chain fragment antibody (ABX-EGF scFv). The uptake of ABX-EGF scFv-modified (LCP-scFv) and folic acid-modified LCP NPs (LCP-FA) by human breast tumor cells (MDA-MB-468) was significantly higher with an optimal ligand density on each NP surface (LCP-125scFv and LCP-100FA). Co-conjugation with sub-optimal dual ligands (50 FA and 75 ABX-EGF scFv) per LCP NP (LCP-50FA-75scFv) further enhanced the cellular uptake. More significantly, much more NPs were delivered to the MDA-MB-468 tumor tissue in the nude mouse model when LCP-50FA-75scFv NPs were used. Therefore, the new dual-ligand LCP NPs may be a valuable targeting system for human breast cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia.
| | | | | | | | | | | |
Collapse
|
82
|
Differentiated embryo chondrocyte plays a crucial role in DNA damage response via transcriptional regulation under hypoxic conditions. PLoS One 2018; 13:e0192136. [PMID: 29466367 PMCID: PMC5821451 DOI: 10.1371/journal.pone.0192136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 01/17/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor hypoxia contributes to a biologically aggressive phenotype and therapeutic resistance. Recent studies have revealed that hypoxia reduces expression of several DNA damage recognition and repair (DRR) genes via both hypoxia-inducible factor (HIF)-independent and -dependent pathways, and this induced genomic instability in cancer cells. We show here that one of the HIF-target genes—differentiated embryo chondrocyte (DEC)—plays a role in DNA damage response via transcriptional repression. Comprehensive gene expression and database analyses have revealed systemic repression of DNA-DRR genes in cancer and non-cancer cells under hypoxic conditions. Hypoxic repression in typical cases was confirmed by quantitative RT-PCR and promoter reporter experiments, and knockdown experiments indicated the critical role of DEC2 in such repression. Assessment of histone H2AX phosphorylation revealed that recognition and repair of DNA double-strand breaks (DSBs) induced by bleomycin or γ-ray irradiation were attenuated; moreover, Cleaved Caspase-3 levels were decreased with pre-conditioning under hypoxia: opposing phenomena were ascertained by knockdown of DEC2. Finally, pre-conditioning under hypoxia decreased the sensitivity of cancer cells to DSBs, and knockdown of DEC2 increased γ-ray sensitivity. These data imply that a critical reduction of DNA-DRR occurs via DEC-dependent transcriptional repression and suggest that DEC is a potential molecular target for anti-cancer strategies.
Collapse
|
83
|
Tumor Microenvironment and Metabolism. Int J Mol Sci 2017; 18:ijms18122729. [PMID: 29258182 PMCID: PMC5751330 DOI: 10.3390/ijms18122729] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 11/30/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023] Open
|
84
|
Xie F, Ding RL, He WF, Liu ZJL, Fu SZ, Wu JB, Yang LL, Lin S, Wen QL. In vivo antitumor effect of endostatin-loaded chitosan nanoparticles combined with paclitaxel on Lewis lung carcinoma. Drug Deliv 2017; 24:1410-1418. [PMID: 28933203 PMCID: PMC8241112 DOI: 10.1080/10717544.2017.1378938] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/05/2017] [Accepted: 09/09/2017] [Indexed: 11/11/2022] Open
Abstract
The purpose of this study was to prepare endostatin-loaded chitosan nanoparticles (ES-NPs) and evaluate their antitumor effect when combined with paclitaxel (PTX) on Lewis lung carcinoma (LLC) mouse xenografts. ES-NPs were prepared by ionic cross-linking. Characterization of the ES-NPs included size distribution, drug-loading efficiency (DL), and encapsulation efficiency (EE). An in vitro release test was also used to determine the release behavior of the ES-NPs. A subcutaneous LC xenograft model of C57BL/6J mice was established. The mice were randomly divided into six groups: control (0.9% NaCl), ES, PTX, ES-NPs, ES + PTX, and ES-NPs + PTX. The tumor volume was dynamically measured for the duration of the experiment. Immunohistochemistry was performed to determine the Ki-67 and microvascular density (MVD) in each group. Serum vascular endothelial growth factor (VEGF) and ES levels were determined by enzyme-linked immunosorbent assay (ELISA). ES-NPs were successfully synthesized and had suitable size distribution and high EE. The NPs were homogenously spherical and exhibited an ideal release profile in vitro. In vivo, tumor growth was significantly inhibited in the ES-NPs + PTX group. The tumor inhibitory rate was significantly higher in the ES-NPs + PTX group than in the other groups (p < .05). The results of the immunohistochemical assay and ELISA confirmed that ES-NPs combined with PTX had a strong antiangiogenic effect. ES-NPs can overcome the shortcomings of free ES, such as short retention time in circulation, which enhances the antitumor effect of ES. The antitumor effect was more pronounced when treatment included PTX and ES-loaded NPs.
Collapse
Affiliation(s)
- Fang Xie
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Rui-Lin Ding
- Experiment and Training Center, Sichuan College of Traditional Chinese Medicine, Mianyang, Sichuan, China
| | - Wen-Feng He
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zong-Jun-Lin Liu
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Shao-Zhi Fu
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing-Bo Wu
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ling-Lin Yang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Sheng Lin
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qing-Lian Wen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
85
|
Kumari P, Jain S, Ghosh B, Zorin V, Biswas S. Polylactide-Based Block Copolymeric Micelles Loaded with Chlorin e6 for Photodynamic Therapy: In Vitro Evaluation in Monolayer and 3D Spheroid Models. Mol Pharm 2017; 14:3789-3800. [DOI: 10.1021/acs.molpharmaceut.7b00548] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Preeti Kumari
- Department of Pharmacy, Birla Institute of Technology & Science—Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad 500078, Telangana, India
| | - Shreya Jain
- Department of Pharmacy, Birla Institute of Technology & Science—Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad 500078, Telangana, India
| | - Balaram Ghosh
- Department of Pharmacy, Birla Institute of Technology & Science—Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad 500078, Telangana, India
| | - Vladimir Zorin
- Department
of Biophysics, Belarusian State University, 220030 Minsk, Belarus
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science—Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad 500078, Telangana, India
| |
Collapse
|
86
|
Yao C, Tu Y, Ding L, Li C, Wang J, Fang H, Huang Y, Zhang K, Lu Q, Wu M, Wang Y. Tumor Cell-Specific Nuclear Targeting of Functionalized Graphene Quantum Dots In Vivo. Bioconjug Chem 2017; 28:2608-2619. [PMID: 28903003 DOI: 10.1021/acs.bioconjchem.7b00466] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Specific targeting of tumor tissues is essential for tumor imaging and therapeutics but remains challenging. Here, we report an unprecedented method using synthetic sulfonic-graphene quantum dots (sulfonic-GQDs) to exactly target the cancer cell nuclei in vivo without any bio- ligand modification, with no intervention in cells of normal tissues. The key factor for such selectivity is the high interstitial fluid pressure (IFP) in tumor tissues, which allows the penetration of sulfonic-GQDs into the plasma membrane of tumor cells. In vitro, the sulfonic-GQDs are repelled out of the cell membrane because of the repulsive force between negatively charged sulfonic-GQDs and the cell membranes which contributes to the low distribution in normal tissues in vivo. However, the plasma membrane-crossing process can be activated by incubating cells in ultrathin film culture medium because of the attachment of sulfonic-GQDs on cell memebranes. Molecular dynamics simulations demonstrated that, once transported across the plasma membrane, the negatively charged functional groups of these GQDs will leave the membrane with a self-cleaning function retaining a small enough size to achieve penetration through the nuclear membrane into the nucleus. Our study showed that IFP is a previously unrecognized mechanism for specific targeting of tumor cell nuclei and suggested that sulfonic-GQDs may be developed into novel tools for tumor-specific imaging and therapeutics.
Collapse
Affiliation(s)
- Chenjie Yao
- Institute of Nano-chemistry and Nano-biology, Shanghai University , Shanghai 200444, P.R. China
| | - Yusong Tu
- College of Physics Science and Technology, Yangzhou University , Jiangsu 225009, P.R. China
| | - Lin Ding
- Institute of Nano-chemistry and Nano-biology, Shanghai University , Shanghai 200444, P.R. China
| | - Chenchen Li
- Institute of Nano-chemistry and Nano-biology, Shanghai University , Shanghai 200444, P.R. China
| | - Jiao Wang
- School of Life Science, Shanghai University , Shanghai 200444, P.R. China
| | - Haiping Fang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences , Shanghai 201800, P. R. China
| | - Yanan Huang
- Institute of Nano-chemistry and Nano-biology, Shanghai University , Shanghai 200444, P.R. China
| | - Kangkang Zhang
- Institute of Nano-chemistry and Nano-biology, Shanghai University , Shanghai 200444, P.R. China
| | - Quan Lu
- Program in Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health , Boston, Massachusetts 02115, United States
| | - Minghong Wu
- Institute of Nano-chemistry and Nano-biology, Shanghai University , Shanghai 200444, P.R. China
| | - Yanli Wang
- Institute of Nano-chemistry and Nano-biology, Shanghai University , Shanghai 200444, P.R. China.,Program in Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health , Boston, Massachusetts 02115, United States
| |
Collapse
|
87
|
Rausch LK, Netzer NC, Hoegel J, Pramsohler S. The Linkage between Breast Cancer, Hypoxia, and Adipose Tissue. Front Oncol 2017; 7:211. [PMID: 28993797 PMCID: PMC5622311 DOI: 10.3389/fonc.2017.00211] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/28/2017] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE The development of breast cancer cells is linked to hypoxia. The hypoxia-induced factor HIF-1α influences metastasis through neovascularization. Hypoxia seems to decrease the responsiveness to hormonal treatment due to loss of estrogen receptors (ERs). Obesity is discussed to increase hypoxia in adipocytes, which promotes a favorable environment for tumor cells in mammary fat tissue, whereas, tumor cells profit from good oxygen supply and are influenced by its deprivation as target regions within tumors show. This review gives an overview of the current state on research of hypoxia and breast cancer in human adipose tissue. METHODS A systematic literature search was conducted on PubMed (2000-2016) by applying hypoxia and/or adipocytes and breast cancer as keywords. Review articles were excluded as well as languages other than English or German. There was no restriction regarding the study design or type of breast cancer. A total of 35 papers were found. Eight studies were excluded due to missing at least two of the three keywords. One paper was removed due to Russian language, and one was dismissed due to lack of adherence. Seven papers were identified as reviews. After applying exclusion criteria, 18 articles were eligible for inclusion. RESULTS Two articles describe the impairment of mammary epithelial cell polarization through hypoxic preconditioning. A high amount of adipocytes enhances cancer progression due to the increased expression of HIF-1α which causes the loss of ER α protein as stated in four articles. Four articles analyzed that increased activation of HIF's induces a series of transcriptions resulting in tumor angiogenesis. HIF inhibition, especially when combined with cytotoxic chemotherapy, holds strong potential for tumor suppression as stated in further four articles. In two articles there is evidence of a strong connection between hypoxia, oxidative stress and a poor prognosis for breast cancer via HIF regulated pathways. Acute hypoxia seems to normalize the microenvironment in breast cancer tissue and has proven to affect tumor growth positively as covered in two articles. CONCLUSION This review indicates that the development of breast cancer is influenced by hypoxia. A high amount of adipocytes enhances cancer progression due to the increased expression of HIF-1α.
Collapse
Affiliation(s)
- Linda K Rausch
- Hermann Buhl Institute for Hypoxia and Sleep Medicine Research, Bad Aibling, Germany.,Department of Sports Science, University Innsbruck, Innsbruck, Austria
| | - Nikolaus C Netzer
- Hermann Buhl Institute for Hypoxia and Sleep Medicine Research, Bad Aibling, Germany.,Department of Sports Science, University Innsbruck, Innsbruck, Austria.,Division of Sports Medicine and Rehabilitation, Department of Medicine, University Ulm, Ulm, Germany
| | - Josef Hoegel
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Stephan Pramsohler
- Hermann Buhl Institute for Hypoxia and Sleep Medicine Research, Bad Aibling, Germany
| |
Collapse
|
88
|
A Biomimetic Microfluidic Tumor Microenvironment Platform Mimicking the EPR Effect for Rapid Screening of Drug Delivery Systems. Sci Rep 2017; 7:9359. [PMID: 28839211 PMCID: PMC5571192 DOI: 10.1038/s41598-017-09815-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/31/2017] [Indexed: 01/18/2023] Open
Abstract
Real-time monitoring of tumor drug delivery in vivo is a daunting challenge due to the heterogeneity and complexity of the tumor microenvironment. In this study, we developed a biomimetic microfluidic tumor microenvironment (bMTM) comprising co-culture of tumor and endothelial cells in a 3D environment. The platform consists of a vascular compartment featuring a network of vessels cultured with endothelial cells forming a complete lumen under shear flow in communication with 3D solid tumors cultured in a tumor compartment. Endothelial cell permeability to both small dye molecules and large liposomal drug carriers were quantified using fluorescence microscopy. Endothelial cell intercellular junction formation was characterized by immunostaining. Endothelial cell permeability significantly increased in the presence of either tumor cell conditioned media (TCM) or tumor cells. The magnitude of this increase in permeability was significantly higher in the presence of metastatic breast tumor cells as compared to non-metastatic ones. Immunostaining revealed impaired endothelial cell-cell junctions in the presence of either metastatic TCM or metastatic tumor cells. Our findings indicate that the bMTM platform mimics the tumor microenvironment including the EPR effect. This platform has a significant potential in applications such as cell-cell/cell-drug carrier interaction studies and rapid screening of cancer drug therapeutics/carriers.
Collapse
|
89
|
Hui TH, Tang YH, Yan Z, Yip TC, Fong HW, Cho WC, Ngan KC, Shum HC, Lin Y. Cadherin- and Rigidity-Dependent Growth of Lung Cancer Cells in a Partially Confined Microenvironment. ACS Biomater Sci Eng 2017; 4:446-455. [PMID: 33418735 DOI: 10.1021/acsbiomaterials.7b00130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During tumor development, cancer cells constantly confront different types of extracellular barriers. However, fundamental questions like whether tumor cells will continue to grow against confinement or away from it and what key factors govern this process remain poorly understood. To address these issues, here we examined the growth dynamics of human lung epithelial carcinoma A549 cells partially confined in micrometer-sized cylindrical pores with precisely controlled wall stiffness. It was found that, after reaching confluency, the cell monolayer enclosed by a compliant wall was able to keep growing and pushing the boundary, eventually leading to a markedly enlarged pore. In contrast, a much reduced in-plane growth and elevated strain level among cells were observed when the confining wall becomes stiff. Furthermore, under such circumstance, cells switched their growth from within the monolayer to along the out-of-plane direction, resulting in cell stacking. We showed that these observations can be well explained by a simple model taking into account the deformability of the wall and the threshold stress for inhibiting cell growth. Interestingly, cadherins were found to play an important role in the proliferation and stress buildup within the cell monolayer by aggregating at cell-cell junctions. The stiff confinement led to an elevated expression level of cadherins. Furthermore, inhibition of N-cadherin resulted in a significantly suppressed cell growth under the same confining conditions.
Collapse
Affiliation(s)
- T H Hui
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.,HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Kejizhong second Rd., Hi-Tech Industrial Park, Nanshan District, Shenzhen, Guangdong, China
| | - Y H Tang
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Z Yan
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.,HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Kejizhong second Rd., Hi-Tech Industrial Park, Nanshan District, Shenzhen, Guangdong, China
| | - T C Yip
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong SAR, China
| | - H W Fong
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong SAR, China
| | - W C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong SAR, China
| | - K C Ngan
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong SAR, China
| | - H C Shum
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.,HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Kejizhong second Rd., Hi-Tech Industrial Park, Nanshan District, Shenzhen, Guangdong, China
| | - Y Lin
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.,HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Kejizhong second Rd., Hi-Tech Industrial Park, Nanshan District, Shenzhen, Guangdong, China
| |
Collapse
|
90
|
Wei Y, Liao R, Mahmood AA, Xu H, Zhou Q. pH-responsive pHLIP (pH low insertion peptide) nanoclusters of superparamagnetic iron oxide nanoparticles as a tumor-selective MRI contrast agent. Acta Biomater 2017; 55:194-203. [PMID: 28363789 DOI: 10.1016/j.actbio.2017.03.046] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/20/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022]
Abstract
Superparamagnetic iron oxide nanoparticles (SPION) are contrast agents used for noninvasive tumor magnetic resonance imaging (MRI). SPION with active targeting by tumor-specific ligands can effectively enhance the MRI sensitivity and specificity of tumors. However, the challenge remains when the tumor specific markers are yet to be determined, especially in the case of early tumor detection. In this study, the effectiveness of pH-responsive SPION via a pH low insertion peptide (pHLIP) to target tumor acidic microenvironments was investigated. Polylysine polymers were first successfully modified with pHLIP to have the pH-responsive capability. SPION pHLIP nanoclusters of 64, 82, 103, and 121nm size were then assembled by the pH-responsive polymers in a size-controlled manner. The pH-responsive SPION nanoclusters of the 64nm size exhibited the most effective pH-responsive retention in cells and tumor selective imaging in MRI. More importantly, the unique contrast enhancement of tumor inner core by the pH-responsive SPION in three different tumor models demonstrated the clinical potential to target tumor acidic microenvironment through pHLIP for tumor early detection and diagnosis by MRI. STATEMENT OF SIGNIFICANCE Detection and diagnosis of tumors at early stage are critical for the improvement of the survival rate of cancer patients. However, the challenge remains when the tumor specific markers are yet to be determined, especially in early tumor detection. pH low insertion peptide (pHLIP) has been used as a specific ligand to target the tumor acidic microenvironment for tumors at early and metastatic stages. Superparamagnetic iron nanoparticles (SPION) are contrast enhancing agents used in the noninvasive magnetic resonance imaging for tumors. This research has demonstrated that pH-responsive pHLIP nanoclusters of SPION were able to target different tumors and facilitate the noninvasive diagnosis of tumors by MRI.
Collapse
Affiliation(s)
- Yushuang Wei
- Department of Nanomedicine & Biopharmaceuticals, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Rufang Liao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Abdulrahman Ahmed Mahmood
- Department of Nanomedicine & Biopharmaceuticals, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Qibing Zhou
- Department of Nanomedicine & Biopharmaceuticals, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
91
|
Brocker C, Kim H, Smith D, Barua S. Heteromer Nanostars by Spontaneous Self-Assembly. NANOMATERIALS 2017; 7:nano7060127. [PMID: 28561759 PMCID: PMC5485774 DOI: 10.3390/nano7060127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/17/2017] [Accepted: 05/23/2017] [Indexed: 11/16/2022]
Abstract
Heteromer star-shaped nanoparticles have the potential to carry out therapeutic agents, improve intracellular uptake, and safely release drugs after prolonged periods of residence at the diseased site. A one-step seed mediation process was employed using polylactide-co-glycolic acid (PLGA), polyvinyl alcohol (PVA), silver nitrate, and tetrakis(hydroxymethyl)phosphonium chloride (THPC). Mixing these reagents followed by UV irradiation successfully produced heteromer nanostars containing a number of arm chains attached to a single core with a high yield. The release of THPC from heteromer nanostars was tested for its potential use for breast cancer treatment. The nanostars present a unique geometrical design exhibiting a significant intracellular uptake by breast cancer cells but low cytotoxicity that potentiates its efficacy as drug carriers.
Collapse
Affiliation(s)
- Caitlin Brocker
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO 65409, USA.
| | - Hannah Kim
- Department of Biological Sciences, Missouri University of Science and Technology, 143 Schrenk Hall, 400 W. 11th St., Rolla, MO 65409, USA.
| | - Daniel Smith
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO 65409, USA.
| | - Sutapa Barua
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO 65409, USA.
| |
Collapse
|
92
|
Gul HI, Yamali C, Yesilyurt F, Sakagami H, Kucukoglu K, Gulcin I, Gul M, Supuran CT. Microwave-assisted synthesis and bioevaluation of new sulfonamides. J Enzyme Inhib Med Chem 2017; 32:369-374. [PMID: 28260401 PMCID: PMC6009867 DOI: 10.1080/14756366.2016.1254207] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In this study, 4-[5-(4-hydroxyphenyl)-3-aryl-4,5-dihydro-1H-pyrazol-1-yl]benzenesulfonamide derivatives (8-14) were synthesized for the first time by microwave irradiation and their chemical structures were confirmed by 1H NMR, 13C NMR and HRMS. Cytotoxic activities and inhibitory effects on carbonic anhydrase I and II isoenzymes of the compounds were investigated. The compounds 9 (PSE = 4.2), 12 (PSE = 4.1) and 13 (PSE = 3.9) with the highest potency selectivity expression (PSE) values in cytotoxicity experiments and the compounds 13 (Ki = 3.73 ± 0.91 nM toward hCA I) and 14 (Ki = 3.85 ± 0.57 nM toward hCA II) with the lowest Ki values in CA inhibition studies can be considered as leader compounds for further studies.
Collapse
Affiliation(s)
- Halise Inci Gul
- a Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Ataturk University , Erzurum , Turkey
| | - Cem Yamali
- a Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Ataturk University , Erzurum , Turkey
| | - Fatma Yesilyurt
- a Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Ataturk University , Erzurum , Turkey
| | - Hiroshi Sakagami
- b Division of Pharmacology , Meikai University School of Dentistry , Sakado , Saitama , Japan
| | - Kaan Kucukoglu
- a Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Ataturk University , Erzurum , Turkey
| | - Ilhami Gulcin
- c Department of Chemistry, Faculty of Science , Ataturk University , Erzurum , Turkey.,d Department of Zoology, College of Science , King Saud University , Riyadh , Saudi Arabia
| | - Mustafa Gul
- e Department of Physiology, Faculty of Medicine , Ataturk University , Erzurum , Turkey
| | - Claudiu T Supuran
- f Neurofarba Departmente Laboratorio di Chimica Bioinorganica , Universita degli Studi di Firenze , Sesto Fiorentino , Florence , Italy
| |
Collapse
|
93
|
Cao Z, Wang X, Cheng X, Wang J, Tang R. In vitro and in vivo antitumor study of folic acid-conjugated carboxymethyl chitosan and phenylboronic acid–based nanoparticles. INT J POLYM MATER PO 2017. [DOI: 10.1080/00914037.2016.1252346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Zhipeng Cao
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| | - Xu Cheng
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| | - Jun Wang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| |
Collapse
|
94
|
Linking tumor glycolysis and immune evasion in cancer: Emerging concepts and therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2017; 1868:212-220. [PMID: 28400131 DOI: 10.1016/j.bbcan.2017.04.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 03/30/2017] [Accepted: 04/06/2017] [Indexed: 12/17/2022]
Abstract
Metabolic reprogramming and immune evasion are two hallmarks of cancer. Metabolic reprogramming is exemplified by cancer's propensity to utilize glucose at an exponential rate which in turn is linked with "aerobic glycolysis", popularly known as the "Warburg effect". Tumor glycolysis is pivotal for the efficient management of cellular bioenergetics and uninterrupted cancer growth. Mounting evidence suggests that tumor glycolysis also plays a key role in instigating immunosuppressive networks that are critical for cancer cells to escape immune surveillance ("immune evasion"). Recent data show that induction of cellular stress or metabolic dysregulation sensitize cancer cells to antitumor immune cells implying that metabolic reprogramming and immune evasion harmonize during cancer progression. However, the molecular link between these two hallmarks of cancer remains obscure. In this review the molecular intricacies of tumor glycolysis that facilitate immune evasion has been discussed in the light of recent research to explore immunotherapeutic potential of targeting cancer metabolism.
Collapse
|
95
|
Zhou Z, Lu ZR. Molecular imaging of the tumor microenvironment. Adv Drug Deliv Rev 2017; 113:24-48. [PMID: 27497513 DOI: 10.1016/j.addr.2016.07.012] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment plays a critical role in tumor initiation, progression, metastasis, and resistance to therapy. It is different from normal tissue in the extracellular matrix, vascular and lymphatic networks, as well as physiologic conditions. Molecular imaging of the tumor microenvironment provides a better understanding of its function in cancer biology, and thus allowing for the design of new diagnostics and therapeutics for early cancer diagnosis and treatment. The clinical translation of cancer molecular imaging is often hampered by the high cost of commercialization of targeted imaging agents as well as the limited clinical applications and small market size of some of the agents. Because many different cancer types share similar tumor microenvironment features, the ability to target these biomarkers has the potential to provide clinically translatable molecular imaging technologies for a spectrum of cancers and broad clinical applications. There has been significant progress in targeting the tumor microenvironment for cancer molecular imaging. In this review, we summarize the principles and strategies of recent advances made in molecular imaging of the tumor microenvironment, using various imaging modalities for early detection and diagnosis of cancer.
Collapse
|
96
|
Foster JC, Radzinski SC, Zou X, Finkielstein CV, Matson JB. H 2S-Releasing Polymer Micelles for Studying Selective Cell Toxicity. Mol Pharm 2017; 14:1300-1306. [PMID: 28300411 DOI: 10.1021/acs.molpharmaceut.6b01117] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
We report the preparation of S-aroylthiooxime (SATO) functionalized amphiphilic block copolymer micelles that release hydrogen sulfide (H2S), a gaseous signaling molecule of relevance to various physiological and pathological conditions. The micelles release H2S in response to cysteine with a half-life of 3.3 h, which is substantially slower than a related small molecule SATO. Exogenous administration of H2S impacts growth and proliferation of cancer cells; however, the limited control over H2S generation from inorganic sulfide sources results in conflicting reports. Therefore, we compare the cellular cytotoxicity of SATO-functionalized micelles, which release H2S in a sustained manner, to Na2S, which releases H2S in a single dose. Our results show that H2S-releasing micelles significantly reduce the survival of HCT116 colon cancer cells relative to Na2S, GYY4137, and a small molecule SATO, indicating that release kinetics may play an important role in determining toxicity of H2S toward cancer cells. Furthermore, H2S-releasing micelles are well tolerated by immortalized fibroblasts (NIH/3T3 cells), suggesting a selective toxicity of H2S toward cancer cells.
Collapse
Affiliation(s)
- Jeffrey C Foster
- Department of Chemistry, Macromolecules Innovation Institute, and Center for Drug Discovery, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - Scott C Radzinski
- Department of Chemistry, Macromolecules Innovation Institute, and Center for Drug Discovery, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - Xianlin Zou
- Department of Biological Sciences and Biocomplexity Institute, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - Carla V Finkielstein
- Department of Biological Sciences and Biocomplexity Institute, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - John B Matson
- Department of Chemistry, Macromolecules Innovation Institute, and Center for Drug Discovery, Virginia Tech , Blacksburg, Virginia 24061, United States
| |
Collapse
|
97
|
Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol 2017; 43:74-89. [PMID: 28267587 DOI: 10.1016/j.semcancer.2017.03.001] [Citation(s) in RCA: 402] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 12/21/2022]
Abstract
The link between cancer metabolism and immunosuppression, inflammation and immune escape has generated major interest in investigating the effects of low pH on tumor immunity. Indeed, microenvironmental acidity may differentially impact on diverse components of tumor immune surveillance, eventually contributing to immune escape and cancer progression. Although the molecular pathways underlying acidity-related immune dysfunctions are just emerging, initial evidence indicates that antitumor effectors such as T and NK cells tend to lose their function and undergo a state of mostly reversible anergy followed by apoptosis, when exposed to low pH environment. At opposite, immunosuppressive components such as myeloid cells and regulatory T cells are engaged by tumor acidity to sustain tumor growth while blocking antitumor immune responses. Local acidity could also profoundly influence bioactivity and distribution of antibodies, thus potentially interfering with the clinical efficacy of therapeutic antibodies including immune checkpoint inhibitors. Hence tumor acidity is a central regulator of cancer immunity that orchestrates both local and systemic immunosuppression and that may offer a broad panel of therapeutic targets. This review outlines the fundamental pathways of acidity-driven immune dysfunctions and sheds light on the potential strategies that could be envisaged to potentiate immune-mediated tumor control in cancer patients.
Collapse
|
98
|
Liang Y, Dong C, Zhang J, Deng L, Dong A. A reconstituted thermosensitive hydrogel system based on paclitaxel-loaded amphiphilic copolymer nanoparticles and antitumor efficacy. Drug Dev Ind Pharm 2017; 43:972-979. [DOI: 10.1080/03639045.2017.1287718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Yanqin Liang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chengxia Dong
- Department of Laboratory, People’s Hospital of Jiyang County, Shandong, China
| | - Jianhua Zhang
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Liandong Deng
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Anjie Dong
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
99
|
Integrins and Cell Metabolism: An Intimate Relationship Impacting Cancer. Int J Mol Sci 2017; 18:ijms18010189. [PMID: 28106780 PMCID: PMC5297821 DOI: 10.3390/ijms18010189] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/26/2016] [Accepted: 01/06/2017] [Indexed: 12/19/2022] Open
Abstract
Integrins are important regulators of cell survival, proliferation, adhesion and migration. Once activated, integrins establish a regulated link between the extracellular matrix and the cytoskeleton. Integrins have well-established functions in cancer, such as in controlling cell survival by engagement of many specific intracellular signaling pathways and in facilitating metastasis. Integrins and associated proteins are regulated by control of transcription, membrane traffic, and degradation, as well as by a number of post-translational modifications including glycosylation, allowing integrin function to be modulated to conform to various cellular needs and environmental conditions. In this review, we examine the control of integrin function by cell metabolism, and the impact of this regulation in cancer. Within this context, nutrient sufficiency or deprivation is sensed by a number of metabolic signaling pathways such as AMP-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR) and hypoxia-inducible factor (HIF) 1, which collectively control integrin function by a number of mechanisms. Moreover, metabolic flux through specific pathways also controls integrins, such as by control of integrin glycosylation, thus impacting integrin-dependent cell adhesion and migration. Integrins also control various metabolic signals and pathways, establishing the reciprocity of this regulation. As cancer cells exhibit substantial changes in metabolism, such as a shift to aerobic glycolysis, enhanced glucose utilization and a heightened dependence on specific amino acids, the reciprocal regulation of integrins and metabolism may provide important clues for more effective treatment of various cancers.
Collapse
|
100
|
Gupta A, Kaur CD, Saraf S, Saraf S. Targeting of herbal bioactives through folate receptors: a novel concept to enhance intracellular drug delivery in cancer therapy. J Recept Signal Transduct Res 2017; 37:314-323. [PMID: 28095746 DOI: 10.3109/10799893.2016.1147581] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Targeted drug delivery through folate receptor (FR) has emerged as a most biocompatible, target oriented, and non-immunogenic cargoes for the delivery of anticancer drugs. FRs are highly overexpressed in many tumor cells (like ovarian, lung, breast, kidney, brain, endometrial, and colon cancer), and targeting them through conjugates bearing specific ligand with encapsulated nanodrug moiety is undoubtedly, a promising approach toward tumor targeting. Folate, being an endogenous ligand, can be exploited well to affect various cellular events occurring during the progress of tumor, in a more natural and definite way. Thus, the aim of the review lies in summarizing the advancements taken place in the drug delivery system of different therapeutics through FRs and to refine its role as an endogenous ligand, in targeting of synthetic as well as natural bioactives. The review also provides an update on the patents received on the folate-based drug delivery system.
Collapse
Affiliation(s)
- Anshita Gupta
- a University Institute of Pharmacy, Pt. Ravishankar Shukla University , Raipur , Chhattisgarh , India
| | - Chanchal Deep Kaur
- a University Institute of Pharmacy, Pt. Ravishankar Shukla University , Raipur , Chhattisgarh , India
| | - Shailendra Saraf
- a University Institute of Pharmacy, Pt. Ravishankar Shukla University , Raipur , Chhattisgarh , India
| | - Swarnlata Saraf
- a University Institute of Pharmacy, Pt. Ravishankar Shukla University , Raipur , Chhattisgarh , India
| |
Collapse
|