751
|
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
|
752
|
Zhang H, Zhang W, Zhou Y, Jiang Y, Li S. Dual Functional Mesoporous Silicon Nanoparticles Enhance the Radiosensitivity of VPA in Glioblastoma. Transl Oncol 2017; 10:229-240. [PMID: 28193559 PMCID: PMC5304234 DOI: 10.1016/j.tranon.2016.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 01/17/2023] Open
Abstract
Radiotherapy is a critical strategy and standard adjuvant approach to glioblastoma treatment. One of the major challenges facing radiotherapy is to minimize radiation damage to normal tissue without compromising therapeutic effects on cancer cells. Various agents and numerous approaches have been developed to improve the therapeutic index of radiotherapy. Among them, radiosensitizers have attracted much attention because they selectively increase susceptibility of cancer cells to radiation and thus enhance biological effectiveness of radiotherapy. However, clinical translation of radiosensitizers has been severely limited by their potential toxicity to normal tissue. Recent advances in nanomedicine offer an opportunity to overcome this hindrance. In this study, a dual functional mesoporous silica nanoparticle (MSN) formulation of the valproic acid (VPA) radiosensitizer was developed, which specifically recognized folic acid-overexpressing cancer cells and released VPA conditionally in acidic turmeric microenvironment. The efficacy of this targeted and pH-responsive VPA nanocarrier was evaluated as compared to VPA treatment approach in two cell lines: rat glioma cells C6 and human glioma U87. Compared to VPA treatment, targeted VPA-MSNs not only potentiated the toxic effects of radiation and led to a higher rate of cell death but also enhanced inhibition on clonogenic assay. More interestingly, these effects were further accentuated by VPA-MSNs at low pH values. Western blot analysis showed that the effects were mediated via enhanced apoptosis-inducing effects. Our results suggest that the adjunctive use of VPA-MSNs may enhance the effectiveness of radiotherapy in glioma treatment by lowering the radiation doses required to kill cancer cells and thereby minimize collateral damage to healthy adjacent tissue.
Collapse
Affiliation(s)
- Hailong Zhang
- Drug Innovation, The Second Hospital of Shandong University, Jinan, PR China
| | - Wei Zhang
- Cancer Centre, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, PR China
| | - Yong Zhou
- Cancer Centre, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, PR China
| | - Yuhua Jiang
- Drug Innovation, The Second Hospital of Shandong University, Jinan, PR China
- Cancer Centre, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, PR China
| | - Shupeng Li
- Drug Innovation, The Second Hospital of Shandong University, Jinan, PR China
- Cancer Centre, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, PR China
| |
Collapse
|
753
|
Yan B, Qin H, Huang C, Li C, Chen Q, Xing D. Single-wavelength excited photoacoustic-fluorescence microscopy for in vivo pH mapping. OPTICS LETTERS 2017; 42:1253-1256. [PMID: 28362742 DOI: 10.1364/ol.42.001253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The balance between radiative and non-radiative decay is affected by various factors at the excitation state. A simultaneous acquisition of the fluorescence and photoacoustic signals from an appropriate probe provides an efficient, high-resolution means to monitor such a balance in a biological target, and thus may render its physiological information. Acidity plays an important role in tissue physiology. Here, we report an integrated photoacoustic-fluorescence microscopy (PA-FLM) for high-resolution (<3 μm) image mapping of interstitial pH by detecting the shift in the signal balance of a pH-sensitive probe. The hypothesis and the technical feasibility are validated with an in vivo tumor model. The results show that the technique can effectively monitor pH changes within the range of biological acidity and are independent of the excitation source fluctuation and local probe concentration. We thus propose that, with further research and selection of proper probes, PA-FLM may provide a potential alternative for monitoring tissue physiology.
Collapse
|
754
|
Kelbauskas L, Glenn H, Anderson C, Messner J, Lee KB, Song G, Houkal J, Su F, Zhang L, Tian Y, Wang H, Bussey K, Johnson RH, Meldrum DR. A platform for high-throughput bioenergy production phenotype characterization in single cells. Sci Rep 2017; 7:45399. [PMID: 28349963 PMCID: PMC5368665 DOI: 10.1038/srep45399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/23/2017] [Indexed: 02/06/2023] Open
Abstract
Driven by an increasing number of studies demonstrating its relevance to a broad variety of disease states, the bioenergy production phenotype has been widely characterized at the bulk sample level. Its cell-to-cell variability, a key player associated with cancer cell survival and recurrence, however, remains poorly understood due to ensemble averaging of the current approaches. We present a technology platform for performing oxygen consumption and extracellular acidification measurements of several hundreds to 1,000 individual cells per assay, while offering simultaneous analysis of cellular communication effects on the energy production phenotype. The platform comprises two major components: a tandem optical sensor for combined oxygen and pH detection, and a microwell device for isolation and analysis of single and few cells in hermetically sealed sub-nanoliter chambers. Our approach revealed subpopulations of cells with aberrant energy production profiles and enables determination of cellular response variability to electron transfer chain inhibitors and ion uncouplers.
Collapse
Affiliation(s)
- Laimonas Kelbauskas
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Honor Glenn
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Clifford Anderson
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Jacob Messner
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Kristen B. Lee
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Ganquan Song
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Jeff Houkal
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Fengyu Su
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Liqiang Zhang
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Yanqing Tian
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Hong Wang
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Kimberly Bussey
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Roger H. Johnson
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| | - Deirdre R. Meldrum
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, 1001S. McAllister Ave., Tempe, AZ 85287, USA
| |
Collapse
|
755
|
Temperature induces significant changes in both glycolytic reserve and mitochondrial spare respiratory capacity in colorectal cancer cell lines. Exp Cell Res 2017; 354:112-121. [PMID: 28342898 DOI: 10.1016/j.yexcr.2017.03.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 02/27/2017] [Accepted: 03/21/2017] [Indexed: 12/21/2022]
Abstract
Thermotherapy, as a method of treating cancer, has recently attracted considerable attention from basic and clinical investigators. A number of studies and clinical trials have shown that thermotherapy can be successfully used as a therapeutic approach for various cancers. However, the effects of temperature on cancer bioenergetics have not been studied in detail with a real time, microplate based, label-free detection approach. This study investigates how changes in temperature affect the bioenergetics characteristics (mitochondrial function and glycolysis) of three colorectal cancer (CRC) cell lines utilizing the Seahorse XF96 technology. Experiments were performed at 32°C, 37°C and 42°C using assay medium conditions and equipment settings adjusted to produce equal oxygen and pH levels ubiquitously at the beginning of all experiments. The results suggest that temperature significantly changes multiple components of glycolytic and mitochondrial function of all cell lines tested. Under hypothermia conditions (32°C), the extracellular acidification rates (ECAR) of CRC cells were significantly lower compared to the same basal ECAR levels measured at 37°C. Mitochondrial stress test for SW480 cells at 37°C vs 42°C demonstrated increased proton leak while all other OCR components remained unchanged (similar results were detected also for the patient-derived xenograft cells Pt.93). Interestingly, the FCCP dose response at 37°C vs 42°C show significant shifts in profiles, suggesting that single dose FCCP experiments might not be sufficient to characterize the mitochondrial metabolic potential when comparing groups, conditions or treatments. These findings provide valuable insights for the metabolic and bioenergetic changes of CRC cells under hypo- and hyperthermia conditions that could potentially lead to development of better targeted and personalized strategies for patients undergoing combined thermotherapy with chemotherapy.
Collapse
|
756
|
Agrawal AK, Aqil F, Jeyabalan J, Spencer WA, Beck J, Gachuki BW, Alhakeem SS, Oben K, Munagala R, Bondada S, Gupta RC. Milk-derived exosomes for oral delivery of paclitaxel. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1627-1636. [PMID: 28300659 DOI: 10.1016/j.nano.2017.03.001] [Citation(s) in RCA: 379] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/13/2017] [Accepted: 03/03/2017] [Indexed: 12/11/2022]
Abstract
In this report milk-derived exosomes have been investigated for oral delivery of the chemotherapeutic drug paclitaxel (PAC) as an alternative to conventional i.v. therapy for improved efficacy and reduced toxicity. PAC-loaded exosomes (ExoPAC) were found to have a particle size of ~108 nm, a narrow particle size distribution (PDI ~0.190), zeta potential (~ -7 mV) and a practical loading efficiency of ~8%. Exosomes and ExoPAC exhibited excellent stability in the presence of simulated-gastrointestinal fluids, and during the storage at -80 °C. A sustained release of PAC was also observed up to 48 h in vitro using PBS (pH 6.8). Importantly, ExoPAC delivered orally showed significant tumor growth inhibition (60%; P<0.001) against human lung tumor xenografts in nude mice. Treatment with i.p. PAC at the same dose as ExoPAC, however, showed modest but statistically insignificant inhibition (31%). Moreover, ExoPAC demonstrated remarkably lower systemic and immunologic toxicities as compared to i.v. PAC.
Collapse
Affiliation(s)
- Ashish K Agrawal
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY
| | - Farrukh Aqil
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY; Department of Medicine, University of Louisville, Louisville, KY
| | | | | | | | - Beth W Gachuki
- Department of Microbiology, Immunology & Molecular Genetics, and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Sara S Alhakeem
- Department of Microbiology, Immunology & Molecular Genetics, and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Karine Oben
- Department of Microbiology, Immunology & Molecular Genetics, and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Radha Munagala
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY; Department of Medicine, University of Louisville, Louisville, KY
| | - Subbarao Bondada
- Department of Microbiology, Immunology & Molecular Genetics, and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Ramesh C Gupta
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY; 3P Biotechnologies, Inc., Louisville, KY; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY.
| |
Collapse
|
757
|
Hu X, Chao M, Wu H. Central role of lactate and proton in cancer cell resistance to glucose deprivation and its clinical translation. Signal Transduct Target Ther 2017; 2:16047. [PMID: 29263910 PMCID: PMC5661620 DOI: 10.1038/sigtrans.2016.47] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/21/2016] [Accepted: 12/26/2016] [Indexed: 12/14/2022] Open
Abstract
Targeting common weaknesses of cancer is an important strategy for cancer therapy. Glucose is a nutrient that maintains essential cellular metabolism, supporting cancer cell survival, growth and proliferation. Depriving glucose rapidly kills cancer cells. Most cancer cells possess a feature called Warburg effect, which refers to that cancer cells even with ample oxygen exhibit an exceptionally high glycolysis rate and convert most incoming glucose to lactate. Although it is recognized that Warburg effect confers growth advantage to cancer cells when glucose supply is sufficient, this feature could be considered as a fatal weakness of cancer cells when glucose supply is a problem. As glucose supply in many solid tumors is poor, and as most cancer cells have exceptionally high glycolytic capacity, maximizing cancer cell glycolysis rate would possibly exhaust intratumoral glucose, leading cancer cell to death. Lactate and proton are two common factors in solid tumors, they jointly protect cancer cells against glucose deprivation, and they are also powerful regulators dictating glucose metabolic phenotypes of cancer cells. Disrupting the joint action of lactate and proton, for example, by means of bicarbonate infusion into tumor, could maximize cancer cell glycolytic rate to rapidly use up glucose, expose their vulnerability to glucose deprivation and ultimately kill cancer cells. A pilot clinical study demonstrated that this approach achieved a remarkable improvement in local control of large and huge hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xun Hu
- Cancer Institute (a Key Laboratory For Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Chao
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wu
- Cancer Institute (a Key Laboratory For Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
758
|
Qiu H, Min Y, Rodgers Z, Zhang L, Wang AZ. Nanomedicine approaches to improve cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9. [PMID: 28296286 DOI: 10.1002/wnan.1456] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/28/2016] [Accepted: 12/17/2016] [Indexed: 01/10/2023]
Abstract
Significant advances have been made in the field of cancer immunotherapy by orchestrating the body's immune system to eradicate cancer cells. However, safety and efficacy concerns stemming from the systemic delivery of immunomodulatory compounds limits cancer immunotherapies expansion and application. In this context, nanotechnology presents a number of advantages, such as targeted delivery to immune cells, enhanced clinical outcomes, and reduced adverse events, which may aid in the delivery of cancer vaccines and immunomodulatory agents. With this in mind, a diverse range of nanomaterials with different physicochemical characteristics have been developed to stimulate the immune system and battle cancer. In this review, we will focus on some recent developments and the potential advantages of utilizing nanotechnology within the field of cancer immunotherapy. WIREs Nanomed Nanobiotechnol 2017, 9:e1456. doi: 10.1002/wnan.1456 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Hui Qiu
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuanzeng Min
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Zach Rodgers
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Longzhen Zhang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
759
|
Giammona A, Mangiapane LR, Di Franco S, Benfante A, Todaro M, Stassi G. Innovative Therapeutic Strategies Targeting Colorectal Cancer Stem Cells. CURRENT COLORECTAL CANCER REPORTS 2017. [DOI: 10.1007/s11888-017-0353-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
760
|
John S, Sivakumar KC, Mishra R. Extracellular Proton Concentrations Impacts LN229 Glioblastoma Tumor Cell Fate via Differential Modulation of Surface Lipids. Front Oncol 2017; 7:20. [PMID: 28299282 PMCID: PMC5331044 DOI: 10.3389/fonc.2017.00020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/02/2017] [Indexed: 12/13/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer with marginal survival rates. GBM extracellular acidosis can profoundly impact its cell fate heterogeneities and progression. However, the molecules and mechanisms that enable GBM tumor cells acid adaptation and consequent cell fate competencies are weakly understood. Since extracellular proton concentrations (pHe) directly intercept the tumor cell plasma membrane, surface lipids must play a crucial role in pHe-dependent tumor cell fate dynamics. Hence, a more detailed insight into the finely tuned pH-dependent modulation of surface lipids is required to generate strategies that can inhibit or surpass tumor cell acid adaptation, thereby forcing the eradication of heterogeneous oncogenic niches, without affecting the normal cells. Results By using image-based single cell analysis and physicochemical techniques, we made a small-scale survey of the effects of pH ranges (physiological: pHe 7.4, low: 6.2, and very low: 3.4) on LN229 glioblastoma cell line surface remodeling and analyzed the consequent cell fate heterogeneities with relevant molecular targets and behavioral assays. Through this basic study, we uncovered that the extracellular proton concentration (1) modulates surface cholesterol-driven cell fate dynamics and (2) induces ‘differential clustering’ of surface resident GM3 glycosphingolipid which together coordinates the proliferation, migration, survival, and death reprogramming via distinct effects on the tumor cell biomechanical homeostasis. A novel synergy of anti-GM3 antibody and cyclophilin A inhibitor was found to mimic the very low pHe-mediated GM3 supraclustered conformation that elevated the surface rigidity and mechano-remodeled the tumor cell into a differentiated phenotype which eventually succumbed to the anoikis type of cell death, thereby eradicating the tumorigenic niches. Conclusion and significance This work presents an initial insight into the physicochemical capacities of extracellular protons in the generation of glioblastoma tumor cell heterogeneities and cell death via the crucial interplay of surface lipids and their conformational changes. Hence, monitoring of proton–cholesterol–GM3 correlations in vivo through diagnostic imaging and in vitro in clinical samples may assist better tumor staging and prognosis. The emerged insights have further led to the translation of a ‘pH-dependent mechanisms of oncogenesis control’ into the surface targeted anti-GBM therapeutics.
Collapse
Affiliation(s)
- Sebastian John
- Disease Biology Program, Department of Neurobiology and Genetics, Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , India
| | - K C Sivakumar
- Distributed Information Sub-Centre, Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , India
| | - Rashmi Mishra
- Disease Biology Program, Department of Neurobiology and Genetics, Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , India
| |
Collapse
|
761
|
Noguchi F, Inui S, Fedele C, Shackleton M, Itami S. Calcium-Dependent Enhancement by Extracellular Acidity of the Cytotoxicity of Mitochondrial Inhibitors against Melanoma. Mol Cancer Ther 2017; 16:936-947. [DOI: 10.1158/1535-7163.mct-15-0235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 04/30/2015] [Accepted: 02/08/2017] [Indexed: 11/16/2022]
|
762
|
Pautu V, Leonetti D, Lepeltier E, Clere N, Passirani C. Nanomedicine as a potent strategy in melanoma tumor microenvironment. Pharmacol Res 2017; 126:31-53. [PMID: 28223185 DOI: 10.1016/j.phrs.2017.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 12/19/2022]
Abstract
Melanoma originated from melanocytes is the most aggressive type of skin cancer. Despite considerable progresses in clinical treatment with the discovery of BRAF or MEK inhibitors and monoclonal antibodies, the durability of response to treatment is often limited to the development of acquired resistance and systemic toxicity. The limited success of conventional treatment highlights the importance of understanding the role of melanoma tumor microenvironment in tumor developement and drug resistance. Nanoparticles represent a promising strategy for the development of new cancer treatments able to improve the bioavailability of drugs and increase their penetration by targeting specifically tumors cells and/or tumor environment. In this review, we will discuss the main influence of tumor microenvironment in melanoma growth and treatment outcome. Furthermore, third generation loaded nanotechnologies represent an exciting tool for detection, treatment, and escape from possible mechanism of resistance mediated by tumor microenvironment, and will be highlighted in this review.
Collapse
Affiliation(s)
- Vincent Pautu
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | | | - Elise Lepeltier
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | - Nicolas Clere
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | - Catherine Passirani
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France.
| |
Collapse
|
763
|
White KA, Grillo-Hill BK, Barber DL. Cancer cell behaviors mediated by dysregulated pH dynamics at a glance. J Cell Sci 2017; 130:663-669. [PMID: 28202602 PMCID: PMC5339414 DOI: 10.1242/jcs.195297] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysregulated pH is a common characteristic of cancer cells, as they have an increased intracellular pH (pHi) and a decreased extracellular pH (pHe) compared with normal cells. Recent work has expanded our knowledge of how dysregulated pH dynamics influences cancer cell behaviors, including proliferation, metastasis, metabolic adaptation and tumorigenesis. Emerging data suggest that the dysregulated pH of cancers enables these specific cell behaviors by altering the structure and function of selective pH-sensitive proteins, termed pH sensors. Recent findings also show that, by blocking pHi increases, cancer cell behaviors can be attenuated. This suggests ion transporter inhibition as an effective therapeutic approach, either singly or in combination with targeted therapies. In this Cell Science at a Glance article and accompanying poster, we highlight the interconnected roles of dysregulated pH dynamics in cancer initiation, progression and adaptation.
Collapse
Affiliation(s)
- Katharine A White
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Bree K Grillo-Hill
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
764
|
Kolosenko I, Avnet S, Baldini N, Viklund J, De Milito A. Therapeutic implications of tumor interstitial acidification. Semin Cancer Biol 2017; 43:119-133. [PMID: 28188829 DOI: 10.1016/j.semcancer.2017.01.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 01/25/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
Interstitial acidification is a hallmark of solid tumor tissues resulting from the combination of different factors, including cellular buffering systems, defective tissue perfusion and high rates of cellular metabolism. Besides contributing to tumor pathogenesis and promoting tumor progression, tumor acidosis constitutes an important intrinsic and extrinsic mechanism modulating therapy sensitivity and drug resistance. In fact, pharmacological properties of anticancer drugs can be affected not only by tissue structure and organization but also by the distribution of the interstitial tumor pH. The acidic tumor environment is believed to create a chemical barrier that limits the effects and activity of many anticancer drugs. In this review article we will discuss the general protumorigenic effects of acidosis, the role of tumor acidosis in the modulation of therapeutic efficacy and potential strategies to overcome pH-dependent therapy-resistance.
Collapse
Affiliation(s)
- Iryna Kolosenko
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Sofia Avnet
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Angelo De Milito
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
765
|
Wenger KJ, Hattingen E, Franz K, Steinbach JP, Bähr O, Pilatus U. Intracellular pH measured by 31 P-MR-spectroscopy might predict site of progression in recurrent glioblastoma under antiangiogenic therapy. J Magn Reson Imaging 2017; 46:1200-1208. [PMID: 28165649 DOI: 10.1002/jmri.25619] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/15/2016] [Indexed: 01/05/2023] Open
Abstract
PURPOSE In solid tumors, changes in the expression/activity of plasma membrane ion transporters facilitate proton efflux and enable tumor cells to maintain a higher intracellular pH (pHi ), while the microenvironment (pHe ) is commonly more acidic. This supports various tumor-promoting mechanisms. We propose that these changes in pH take place before a magnetic resonance imaging (MRI)-detectable brain tumor recurrence occurs. MATERIALS AND METHODS We enrolled 66 patients with recurrent glioblastoma, treated with bevacizumab. Patients received a baseline and 8-week follow-up MRI including 1 H/31 P MRSI (spectroscopy) on a 3T clinical scanner, until progressive disease according to Response Assessment in Neuro-Oncology (RANO) criteria occurred. Fourteen patients showed a distant or diffuse tumor recurrence (subsequent tumor) during treatment and were therefore selected for further evaluation. At the site of the subsequent tumor, an area of interest for MRSI voxel selection was retrospectively defined on radiographically unaffected baseline MRI sequences. RESULTS Before treatment, pHi in the area of interest (subsequent tumor) was significantly higher than pHi of the contralateral normal-appearing tissue (control; P < 0.001). It decreased at the time of best response (P = 0.06), followed by a significant increase at progression (P = 0.03; baseline mean: 7.06, median: 7.068, SD: 0.032; best response mean: 7.044, median: 7.036, SD: 0.025; progression mean: 7.08, median: 7.095, SD 0.035). Until progression, the subsequent tumor was not detectable on standard MRI sequences. The area of existing tumor responded similar, but changes were not significant (decrease P = 0.22; increase P = 0.28). CONCLUSION Elevated pHi in radiographically unaffected tissue at baseline might precede MRI-detectable progression in patients with recurrent glioblastoma treated with bevacizumab. LEVEL OF EVIDENCE 2 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2017;46:1200-1208.
Collapse
Affiliation(s)
- Katharina J Wenger
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Kea Franz
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Joachim P Steinbach
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Bähr
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrich Pilatus
- Institute of Neuroradiology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| |
Collapse
|
766
|
Preparation of dual-responsive hybrid fluorescent nano probe based on graphene oxide and boronic acid/BODIPY-conjugated polymer for cell imaging. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:1064-1071. [DOI: 10.1016/j.msec.2016.11.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/04/2016] [Accepted: 11/07/2016] [Indexed: 12/25/2022]
|
767
|
Melzer C, von der Ohe J, Lehnert H, Ungefroren H, Hass R. Cancer stem cell niche models and contribution by mesenchymal stroma/stem cells. Mol Cancer 2017; 16:28. [PMID: 28148265 PMCID: PMC5286787 DOI: 10.1186/s12943-017-0595-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/18/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The initiation and progression of malignant tumors is driven by distinct subsets of tumor-initiating or cancer stem-like cells (CSCs) which develop therapy/apoptosis resistance and self-renewal capacity. In order to be able to eradicate these CSCs with novel classes of anti-cancer therapeutics, a better understanding of their biology and clinically-relevant traits is mandatory. MAIN BODY Several requirements and functions of a CSC niche physiology are combined with current concepts for CSC generation such as development in a hierarchical tumor model, by stochastic processes, or via a retrodifferentiation program. Moreover, progressive adaptation of endothelial cells and recruited immune and stromal cells to the tumor site substantially contribute to generate a tumor growth-permissive environment resembling a CSC niche. Particular emphasis is put on the pivotal role of multipotent mesenchymal stroma/stem cells (MSCs) in supporting CSC development by various kinds of interaction and cell fusion to form hybrid tumor cells. CONCLUSION A better knowledge of CSC niche physiology may increase the chances that cancer stemness-depleting interventions ultimately result in arrest of tumor growth and metastasis.
Collapse
Affiliation(s)
- Catharina Melzer
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Medical University Hannover, Carl-Neuberg-Str. 1, D – 30625 Hannover, Germany
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Medical University Hannover, Carl-Neuberg-Str. 1, D – 30625 Hannover, Germany
| | - Hendrik Lehnert
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
- Department of General, Visceral-, Thoracic-Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, Kiel, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Medical University Hannover, Carl-Neuberg-Str. 1, D – 30625 Hannover, Germany
| |
Collapse
|
768
|
Targeting pH regulating proteins for cancer therapy-Progress and limitations. Semin Cancer Biol 2017; 43:66-73. [PMID: 28137473 DOI: 10.1016/j.semcancer.2017.01.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/24/2017] [Indexed: 12/21/2022]
Abstract
Tumour acidity induced by metabolic alterations and incomplete vascularisation sets cancer cells apart from normal cellular physiology. This distinguishing tumour characteristic has been an area of intense study, as cellular pH (pHi) disturbances disrupt protein function and therefore multiple cellular processes. Tumour cells effectively utilise pHi regulating machinery present in normal cells with enhancements provided by additional oncogenic or hypoxia induced protein modifications. This overall improvement of pH regulation enables maintenance of an alkaline pHi in the continued presence of external acidification (pHe). Considerable experimentation has revealed targets that successfully disrupt tumour pHi regulation in efforts to develop novel means to weaken or kill tumour cells. However, redundancy in these pH-regulating proteins, which include Na+/H+ exchangers (NHEs), carbonic anhydrases (CAs), Na+/HCO3- co-transporters (NBCs) and monocarboxylate transporters (MCTs) has prevented effective disruption of tumour pHi when individual protein targeting is performed. Here we synthesise recent advances in understanding both normoxic and hypoxic pH regulating mechanisms in tumour cells with an ultimate focus on the disruption of tumour growth, survival and metastasis. Interactions between tumour acidity and other cell types are also proving to be important in understanding therapeutic applications such as immune therapy. Promising therapeutic developments regarding pH manipulation along with current limitations are highlighted to provide a framework for future research directives.
Collapse
|
769
|
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
|
770
|
Cui L, Her S, Dunne M, Borst GR, De Souza R, Bristow RG, Jaffray DA, Allen C. Significant Radiation Enhancement Effects by Gold Nanoparticles in Combination with Cisplatin in Triple Negative Breast Cancer Cells and Tumor Xenografts. Radiat Res 2017; 187:147-160. [PMID: 28085639 DOI: 10.1667/rr14578.1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Gold nanoparticles (AuNPs) and cisplatin have been explored in concomitant chemoradiotherapy, wherein they elicit their effects by distinct and overlapping mechanisms. Cisplatin is one of the most frequently utilized radiosensitizers in the clinical setting; however, the therapeutic window of cisplatin-aided chemoradiotherapy is limited by its toxicity. The goal of this study was to determine whether AuNPs contribute to improving the treatment response when combined with fractionated cisplatin-based chemoradiation in both in vitro and in vivo models of triple-negative breast cancer (MDA-MB-231Luc+). Cellular-targeting AuNPs with receptor-mediated endocytosis (AuNP-RME) in vitro at a noncytotoxic concentration (0.5 mg/ml) or cisplatin at IC25 (12 μM) demonstrated dose enhancement factors (DEFs) of 1.25 and 1.14, respectively; the combination of AuNP-RME and cisplatin resulted in a significant DEF of 1.39 in vitro. Transmission electron microscopy (TEM) images showed effective cellular uptake of AuNPs at tumor sites 24 h after intratumoral infusion. Computed tomography (CT) images demonstrated that the intratumoral levels of gold remained stable up to 120 h after infusion. AuNPs (0.5 mg gold per tumor) demonstrated a radiation enhancement effect that was equivalent to three doses of cisplatin at IC25 (4 mg/kg), but did not induce intrinsic toxicity or increased radiotoxicity. Results from this study suggest that AuNPs are the true radiosensitizer in these settings. Importantly, AuNPs enhance the treatment response when combined with cisplatin-based fractionated chemoradiation. This combination of AuNPs and cisplatin provides a promising approach to improving the therapeutic ratio of fractionated radiotherapy.
Collapse
Affiliation(s)
- Lei Cui
- Departments of a Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy
| | - Sohyoung Her
- Departments of a Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy
| | - Michael Dunne
- Departments of a Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy
| | - Gerben R Borst
- d Department of Radiation Oncology, the Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| | - Raquel De Souza
- Departments of a Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy
| | - Robert G Bristow
- b Radiation Oncology and Medical Biophysics and.,e Ontario Cancer Institute.,f STTARR Innovation Centre, Radiation Medicine Program
| | - David A Jaffray
- b Radiation Oncology and Medical Biophysics and.,c Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,f STTARR Innovation Centre, Radiation Medicine Program.,g TECHNA Institute and.,h Department of Radiation Physics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Christine Allen
- Departments of a Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy.,c Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,f STTARR Innovation Centre, Radiation Medicine Program
| |
Collapse
|
771
|
The Effect of Surface Charges on the Cellular Uptake of Liposomes Investigated by Live Cell Imaging. Pharm Res 2017; 34:704-717. [DOI: 10.1007/s11095-017-2097-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/04/2017] [Indexed: 12/22/2022]
|
772
|
Simmons A, Burrage PM, Nicolau DV, Lakhani SR, Burrage K. Environmental factors in breast cancer invasion: a mathematical modelling review. Pathology 2017; 49:172-180. [PMID: 28081961 DOI: 10.1016/j.pathol.2016.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/07/2016] [Accepted: 11/13/2016] [Indexed: 12/17/2022]
Abstract
This review presents a brief overview of breast cancer, focussing on its heterogeneity and the role of mathematical modelling and simulation in teasing apart the underlying biophysical processes. Following a brief overview of the main known pathophysiological features of ductal carcinoma, attention is paid to differential equation-based models (both deterministic and stochastic), agent-based modelling, multi-scale modelling, lattice-based models and image-driven modelling. A number of vignettes are presented where these modelling approaches have elucidated novel aspects of breast cancer dynamics, and we conclude by offering some perspectives on the role mathematical modelling can play in understanding breast cancer development, invasion and treatment therapies.
Collapse
Affiliation(s)
- Alex Simmons
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia
| | - Pamela M Burrage
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia
| | - Dan V Nicolau
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia; Mathematical Institute, University of Oxford, Oxford, United Kingdom; Molecular Sense Ltd, Oxford, United Kingdom
| | - Sunil R Lakhani
- The University of Queensland, Centre for Clinical Research and School of Medicine and Pathology Queensland, The Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia
| | - Kevin Burrage
- School of Mathematical Sciences, and ARC Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Gardens Point, Brisbane, Qld, Australia; Department of Computer Science, University of Oxford, United Kingdom.
| |
Collapse
|
773
|
Zhao M, Liu Q, Gong Y, Xu X, Zhang C, Liu X, Zhang C, Guo H, Zhang X, Gong Y, Shao C. GSH-dependent antioxidant defense contributes to the acclimation of colon cancer cells to acidic microenvironment. Cell Cycle 2017; 15:1125-33. [PMID: 26950675 PMCID: PMC4889284 DOI: 10.1080/15384101.2016.1158374] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Due to increased glycolysis and poor local perfusion, solid tumors are usually immersed in an acidic microenvironment. While extracellular acidosis is cytotoxic, cancer cells eventually become acclimated to it. While previous studies have addressed the acute effect of acidosis on cancer cells, little is known about how cancer cells survive chronic acidosis. In this study we exposed colorectal cancer (CRC) cells (HCT15, HCT116 and LoVo) to acidic pH (pH 6.5) continuously for over three months and obtained CRC cells that become acclimated to acidic pH, designated as CRC-acidosis-acclimated or CRC-AA. We unexpectedly found that while acute exposure to low pH resulted in an increase in the level of intracellular reactive oxygen species (ROS), CRC-AA cells exhibited a significantly reduced level of ROS when compared to ancestor cells. CRC-AA cells were found to maintain a higher level of reduced glutathione, via the upregulation of CD44 and glutathione reductase (GSR), among others, than their ancestor cells. Importantly, CRC-AA cells were more sensitive to agents that deplete GSH. Moreover, downregulation of GSR by RNA interference was more deleterious to CRC-AA cells than to control cells. Together, our results demonstrate a critical role of glutathione-dependent antioxidant defense in acclimation of CRC cells to acidic extracellular pH.
Collapse
Affiliation(s)
- Minnan Zhao
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Qiao Liu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Yanchao Gong
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiuhua Xu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Chen Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiaojie Liu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Caibo Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Haiyang Guo
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiyu Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Yaoqin Gong
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Changshun Shao
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China.,b Department of Genetics/Human Genetics Institute of New Jersey , Rutgers University , Piscataway , NJ , USA
| |
Collapse
|
774
|
Yu H, Wang Y, Yang H, Peng K, Zhang X. Injectable self-healing hydrogels formed via thiol/disulfide exchange of thiol functionalized F127 and dithiolane modified PEG. J Mater Chem B 2017; 5:4121-4127. [DOI: 10.1039/c7tb00746a] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
An injectable thermo-responsive hydrogel with excellent mechanical properties which can self-heal under mildly acidic to basic conditions was prepared.
Collapse
Affiliation(s)
- Hansen Yu
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Yanan Wang
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Haiyang Yang
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Kang Peng
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Xingyuan Zhang
- CAS Key Laboratory of Soft Matter Chemistry
- School of Chemistry and Materials Science
- University of Science and Technology of China
- Hefei
- P. R. China
| |
Collapse
|
775
|
Zhao Q, Lin Y, Han N, Li X, Geng H, Wang X, Cui Y, Wang S. Mesoporous carbon nanomaterials in drug delivery and biomedical application. Drug Deliv 2017; 24:94-107. [PMID: 29124979 PMCID: PMC8812584 DOI: 10.1080/10717544.2017.1399300] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent development of nano-technology provides highly efficient and versatile treatment methods to achieve better therapeutic efficacy and lower side effects of malignant cancer. The exploration of drug delivery systems (DDSs) based on nano-material shows great promise in translating nano-technology to clinical use to benefit patients. As an emerging inorganic nanomaterial, mesoporous carbon nanomaterials (MCNs) possess both the mesoporous structure and the carbonaceous composition, endowing them with superior nature compared with mesoporous silica nanomaterials and other carbon-based materials, such as carbon nanotube, graphene and fullerene. In this review, we highlighted the cutting-edge progress of carbon nanomaterials as drug delivery systems (DDSs), including immediate/sustained drug delivery systems and controlled/targeted drug delivery systems. In addition, several representative biomedical applications of mesoporous carbon such as (1) photo-chemo synergistic therapy; (2) delivery of therapeutic biomolecule and (3) in vivo bioimaging are discussed and integrated. Finally, potential challenges and outlook for future development of mesoporous carbon in biomedical fields have been discussed in detail.
Collapse
Affiliation(s)
- Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Yuanzhe Lin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Ning Han
- School of Chinese Materia Medica, Department of Chinese Medicinal Pharmaceutics, Beijing university of Chinese Medicine, Beijing, PR China
| | - Xian Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Hongjian Geng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Xiudan Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Yu Cui
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| |
Collapse
|
776
|
Hypoxia-Related Tumor Acidosis Affects MicroRNA Expression Pattern in Prostate and Breast Tumor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 977:119-124. [DOI: 10.1007/978-3-319-55231-6_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
777
|
Abstract
Physiological characteristics of diseases bring about both challenges and opportunities for targeted drug delivery. Various drug delivery platforms have been devised ranging from macro- to micro- and further into the nanoscopic scale in the past decades. Recently, the favorable physicochemical properties of nanomaterials, including long circulation, robust tissue and cell penetration attract broad interest, leading to extensive studies for therapeutic benefits. Accumulated knowledge about the physiological barriers that affect the in vivo fate of nanomedicine has led to more rational guidelines for tailoring the nanocarriers, such as size, shape, charge, and surface ligands. Meanwhile, progresses in material chemistry and molecular pharmaceutics generate a panel of physiological stimuli-responsive modules that are equipped into the formulations to prepare “smart” drug delivery systems. The capability of harnessing physiological traits of diseased tissues to control the accumulation of or drug release from nanomedicine has further improved the controlled drug release profiles with a precise manner. Successful clinical translation of a few nano-formulations has excited the collaborative efforts from the research community, pharmaceutical industry, and the public towards a promising future of smart drug delivery.
Collapse
Affiliation(s)
- Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Wenyan Ji
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Grace Wright
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
778
|
Qiao W, Lan X, Tsoi JKH, Chen Z, Su RYX, Yeung KK, Matinlinna JP. Biomimetic hollow mesoporous hydroxyapatite microsphere with controlled morphology, entrapment efficiency and degradability for cancer therapy. RSC Adv 2017. [DOI: 10.1039/c7ra09204k] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hollow mesoporous carbonated hydroxyapatite microspheres produced via sodium dodecyl sulfate assisted precipitation can target cancer cells through acidic dependent release of loaded CDDP.
Collapse
Affiliation(s)
- Wei Qiao
- Dental Materials Science, Applied Oral Sciences
- Faculty of Dentistry
- The University of Hong Kong
- Hong Kong S.A.R
- P. R. China
| | - Xinmiao Lan
- Department of Oral and Maxillofacial Surgery
- Faculty of Dentistry
- The University of Hong Kong
- Hong Kong S.A.R
- P. R. China
| | - James K. H. Tsoi
- Dental Materials Science, Applied Oral Sciences
- Faculty of Dentistry
- The University of Hong Kong
- Hong Kong S.A.R
- P. R. China
| | - Zhuofan Chen
- Department of Oral Implantology
- Hospital of Stomatology
- Guanghua School of Stomatology
- Institute of Stomatological Research
- Sun Yat-sen University
| | - Richard Y. X. Su
- Department of Oral and Maxillofacial Surgery
- Faculty of Dentistry
- The University of Hong Kong
- Hong Kong S.A.R
- P. R. China
| | - Kelvin W. K. Yeung
- Department of Orthopaedics and Traumatology
- Li Ka Shing Faculty of Medicine
- The University of Hong Kong
- Hong Kong S.A.R
- P. R. China
| | - Jukka P. Matinlinna
- Dental Materials Science, Applied Oral Sciences
- Faculty of Dentistry
- The University of Hong Kong
- Hong Kong S.A.R
- P. R. China
| |
Collapse
|
779
|
Recouvreux MV, Commisso C. Macropinocytosis: A Metabolic Adaptation to Nutrient Stress in Cancer. Front Endocrinol (Lausanne) 2017; 8:261. [PMID: 29085336 PMCID: PMC5649207 DOI: 10.3389/fendo.2017.00261] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/20/2017] [Indexed: 12/28/2022] Open
Abstract
Oncogenic mutations, such as Ras mutations, drive not only enhanced proliferation but also the metabolic adaptations that confer to cancer cells the ability to sustain cell growth in a harsh tumor microenvironment. These adaptations might represent metabolic vulnerabilities that can be exploited to develop novel and more efficient cancer therapies. Macropinocytosis is an evolutionarily conserved endocytic pathway that permits the internalization of extracellular fluid via large endocytic vesicles known as macropinosomes. Recently, macropinocytosis has been determined to function as a nutrient-scavenging pathway in Ras-driven cancer cells. Macropinocytic uptake of extracellular proteins, and their further degradation within endolysosomes, provides the much-needed amino acids that fuel cancer cell metabolism and tumor growth. Here, we review the molecular mechanisms that govern the process of macropinocytosis, as well as discuss recent work that provides evidence of the important role of macropinocytosis as a nutrient supply pathway in cancer cells.
Collapse
Affiliation(s)
- Maria Victoria Recouvreux
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Cosimo Commisso
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
- *Correspondence: Cosimo Commisso,
| |
Collapse
|
780
|
Molas Saborit J, Caubet A, Brissos RF, Korrodi-Gregório L, Pérez-Tomás R, Martínez M, Gamez P. pH-Driven preparation of two related platinum(ii) complexes exhibiting distinct cytotoxic properties. Dalton Trans 2017; 46:11214-11222. [DOI: 10.1039/c7dt02127e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
A distinct cytotoxic/DNA interaction behaviour has been observed for a pair of aqua/hydroxido/chlorido equilibrium related PtII complex structures. The equilibrium is shifted between {PtII(NN)} and {PtII(NNO)} cores as a function of pCl and pH.
Collapse
Affiliation(s)
- Jordi Molas Saborit
- Department of Inorganic and Organic Chemistry
- Inorganic Chemistry Section
- University of Barcelona
- 08028 Barcelona
- Spain
| | - Amparo Caubet
- Department of Inorganic and Organic Chemistry
- Inorganic Chemistry Section
- University of Barcelona
- 08028 Barcelona
- Spain
| | - Rosa F. Brissos
- Department of Inorganic and Organic Chemistry
- Inorganic Chemistry Section
- University of Barcelona
- 08028 Barcelona
- Spain
| | - Luís Korrodi-Gregório
- Department of Pathology and Experimental Therapeutics
- Faculty of Medicine
- University of Barcelona
- 08907 L'Hospitalet de Llobregat
- Spain
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics
- Faculty of Medicine
- University of Barcelona
- 08907 L'Hospitalet de Llobregat
- Spain
| | - Manuel Martínez
- Department of Inorganic and Organic Chemistry
- Inorganic Chemistry Section
- University of Barcelona
- 08028 Barcelona
- Spain
| | - Patrick Gamez
- Department of Inorganic and Organic Chemistry
- Inorganic Chemistry Section
- University of Barcelona
- 08028 Barcelona
- Spain
| |
Collapse
|
781
|
Seetharaman G, Kallar AR, Vijayan VM, Muthu J, Selvam S. Design, preparation and characterization of pH-responsive prodrug micelles with hydrolyzable anhydride linkages for controlled drug delivery. J Colloid Interface Sci 2016; 492:61-72. [PMID: 28068545 DOI: 10.1016/j.jcis.2016.12.070] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 01/08/2023]
Abstract
We report a new prodrug micelle-based approach in which a model hydrophobic non-steroidal anti-inflammatory drug (NSAID), ibuprofen (Ibu), is tethered to amphiphilic methoxy polyethylene glycol-polypropylene fumarate (mPEG-PPF) diblock copolymer via hydrolytic anhydride linkages for potential controlled release applications of NSAIDs. Synthesized mPEG-PPF-Ibu polymer drug conjugates (PDCs) demonstrated high drug conjugation efficiency (∼90%) and self-assembled to form micellar nanostructures in aqueous medium with critical micelle concentrations ranging between 16 and 30μg/mL. The entrapment efficiency of Ibu in prepared PDC micelles was as high as 18% (w/w). Crosslinking of prodrug micelles with N,N'-dimethylaminoethyl methacrylate conferred pH-responsive characteristics. pH-responsive PDC micelles averaged 100nm in size at pH 7.4 and exhibited concomitant changes in size upon incubation in physiologically relevant mildly acidic conditions. Ibu release was observed to increase with increasing acidic conditions and could be controlled by varying the amount of crosslinker used. Furthermore, the prepared mPEG-PPF-based micelles demonstrated excellent cytocompatibility and cellular internalization in vitro. More importantly, PDC micelles exerted anti-inflammatory effects by significantly decreasing monosodium urate crystal-induced prostaglandin E2 levels in rabbit synoviocyte cultures in vitro. Cumulatively, our results indicate that this new prodrug micelle approach is promising for NSAID-based therapies in the treatment of arthritis and cancer.
Collapse
Affiliation(s)
- Girija Seetharaman
- Polymer Science Division, BMT Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695012, Kerala, India
| | - Adarsh R Kallar
- Polymer Science Division, BMT Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695012, Kerala, India
| | - Vineeth M Vijayan
- Polymer Science Division, BMT Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695012, Kerala, India
| | - Jayabalan Muthu
- Polymer Science Division, BMT Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695012, Kerala, India
| | - Shivaram Selvam
- Polymer Science Division, BMT Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695012, Kerala, India.
| |
Collapse
|
782
|
Faes S, Uldry E, Planche A, Santoro T, Pythoud C, Demartines N, Dormond O. Acidic pH reduces VEGF-mediated endothelial cell responses by downregulation of VEGFR-2; relevance for anti-angiogenic therapies. Oncotarget 2016; 7:86026-86038. [PMID: 27852069 PMCID: PMC5349894 DOI: 10.18632/oncotarget.13323] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 11/06/2016] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic treatments targeting the vascular endothelial growth factor or its receptors have shown clinical benefits. However, impact on long-term survival remains limited. Solid tumors display an acidic microenvironment that profoundly influences their biology. Consequences of acidity on endothelial cells and anti-angiogenic therapies remain poorly characterized and hence are the focus of this study. We found that exposing endothelial cells to acidic extracellular pH resulted in reduced cell proliferation and migration. Also, whereas VEGF increased endothelial cell proliferation and survival at pH 7.4, it had no effect at pH 6.4. Furthermore, in acidic conditions, stimulation of endothelial cells with VEGF did not result in activation of downstream signaling pathways such as AKT. At a molecular level, acidity significantly decreased the expression of VEGFR-2 by endothelial cells. Consequently, anti-angiogenic therapies that target VEGFR-2 such as sunitinib and sorafenib failed to block endothelial cell proliferation in acidic conditions. In vivo, neutralizing tumor acidity with sodium bicarbonate increased the percentage of endothelial cells expressing VEGFR-2 in tumor xenografts. Furthermore, combining sodium bicarbonate with sunitinib provided stronger anti-cancer activity than either treatment alone. Histological analysis showed that sunitinib had a stronger anti-angiogenic effect when combined with sodium bicarbonate. Overall, our results show that endothelial cells prosper independently of VEGF in acidic conditions partly as a consequence of decreased VEGFR-2 expression. They further suggest that strategies aiming to raise intratumoral pH can improve the efficacy of anti-VEGF treatments.
Collapse
Affiliation(s)
- Seraina Faes
- Department of Visceral Surgery, University Hospital of Lausanne, Switzerland
| | - Emilie Uldry
- Department of Visceral Surgery, University Hospital of Lausanne, Switzerland
| | - Anne Planche
- Department of Visceral Surgery, University Hospital of Lausanne, Switzerland
| | - Tania Santoro
- Department of Visceral Surgery, University Hospital of Lausanne, Switzerland
| | - Catherine Pythoud
- Department of Visceral Surgery, University Hospital of Lausanne, Switzerland
| | - Nicolas Demartines
- Department of Visceral Surgery, University Hospital of Lausanne, Switzerland
| | - Olivier Dormond
- Department of Visceral Surgery, University Hospital of Lausanne, Switzerland
| |
Collapse
|
783
|
Ascorbic acid, but not dehydroascorbic acid increases intracellular vitamin C content to decrease Hypoxia Inducible Factor -1 alpha activity and reduce malignant potential in human melanoma. Biomed Pharmacother 2016; 86:502-513. [PMID: 28012930 DOI: 10.1016/j.biopha.2016.12.056] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/09/2016] [Accepted: 12/14/2016] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Accumulation of hypoxia inducible factor-1 alpha (HIF-1α) in malignant tissue is known to contribute to oncogenic progression and is inversely associated with patient survival. Ascorbic acid (AA) depletion in malignant tissue may contribute to aberrant normoxic activity of HIF-1α. While AA supplementation has been shown to attenuate HIF-1α function in malignant melanoma, the use of dehydroascorbic acid (DHA) as a therapeutic means to increase intracellular AA and modulate HIF-1α function is yet to be evaluated. Here we compared the ability of AA and DHA to increase intracellular vitamin C content and decrease the malignant potential of human melanoma by reducing the activity of HIF-1α. METHODS HIF-1α protein accumulation was evaluated by western blot and transcriptional activity was evaluated by reporter gene assay using a HIF-1 HRE-luciferase plasmid. Protein expressions and subcellular localizations of vitamin C transporters were evaluated by western blot and confocal imaging. Intracellular vitamin C content following AA, ascorbate 2-phosphate (A2P), or DHA supplementation was determined using a vitamin C assay. Malignant potential was accessed using a 3D spheroid Matrigel invasion assay. Data was analyzed by One or Two-way ANOVA with Tukey's multiple comparisons test as appropriate with p<0.05 considered significant. RESULTS Melanoma cells expressed both sodium dependent vitamin C (SVCT) and glucose (GLUT) transporters for AA and DHA transport respectively, however advanced melanomas responded favorably to AA, but not DHA. Physiological glucose conditions significantly impaired intracellular vitamin C accumulation following DHA treatment. Consequently, A2P and AA, but not DHA treated cells demonstrated lower HIF-1α protein expression and activity, and reduced malignant potential. The ability of AA to regulate HIF-1α was dependent on SVCT2 function and SVCT2 was not significantly inhibited at pH representative of the tumor microenvironment. CONCLUSIONS The use of ascorbic acid as an adjuvant cancer therapy remains under investigated. While AA and A2P were capable of modulating HIF-1α protein accumulation/activity, DHA supplementation resulted in minimal intracellular vitamin C activity with decreased ability to inhibit HIF-1α activity and malignant potential in advanced melanoma. Restoring AA dependent regulation of HIF-1α in malignant cells may prove beneficial in reducing chemotherapy resistance and improving treatment outcomes.
Collapse
|
784
|
Bainer R, Frankenberger C, Rabe D, An G, Gilad Y, Rosner MR. Gene expression in local stroma reflects breast tumor states and predicts patient outcome. Sci Rep 2016; 6:39240. [PMID: 27982086 PMCID: PMC5159815 DOI: 10.1038/srep39240] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/21/2016] [Indexed: 02/04/2023] Open
Abstract
The surrounding microenvironment has been implicated in the progression of breast tumors to metastasis. However, the degree to which metastatic breast tumors locally reprogram stromal cells as they disrupt tissue boundaries is not well understood. We used species-specific RNA sequencing in a mouse xenograft model to determine how the metastasis suppressor RKIP influences transcription in a panel of paired tumor and stroma tissues. We find that gene expression in metastatic breast tumors is pervasively correlated with gene expression in local stroma of both mouse xenografts and human patients. Changes in stromal gene expression elicited by tumors better predicts subtype and patient survival than tumor gene expression, and genes with coordinated expression in both tissues predict metastasis-free survival. These observations support the use of stroma-based strategies for the diagnosis and prognosis of breast cancer.
Collapse
Affiliation(s)
- Russell Bainer
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Casey Frankenberger
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Daniel Rabe
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Gary An
- Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | - Yoav Gilad
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
785
|
Rat Aquaporin-5 Is pH-Gated Induced by Phosphorylation and Is Implicated in Oxidative Stress. Int J Mol Sci 2016; 17:ijms17122090. [PMID: 27983600 PMCID: PMC5187890 DOI: 10.3390/ijms17122090] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/25/2016] [Accepted: 12/06/2016] [Indexed: 12/02/2022] Open
Abstract
Aquaporin-5 (AQP5) is a membrane water channel widely distributed in human tissues that was found up-regulated in different tumors and considered implicated in carcinogenesis in different organs and systems. Despite its wide distribution pattern and physiological importance, AQP5 short-term regulation was not reported and mechanisms underlying its involvement in cancer are not well defined. In this work, we expressed rat AQP5 in yeast and investigated mechanisms of gating, as well as AQP5’s ability to facilitate H2O2 plasma membrane diffusion. We found that AQP5 can be gated by extracellular pH in a phosphorylation-dependent manner, with higher activity at physiological pH 7.4. Moreover, similar to other mammalian AQPs, AQP5 is able to increase extracellular H2O2 influx and to affect oxidative cell response with dual effects: whereas in acute oxidative stress conditions AQP5 induces an initial higher sensitivity, in chronic stress AQP5 expressing cells show improved cell survival and resistance. Our findings support the involvement of AQP5 in oxidative stress and suggest AQP5 modulation by phosphorylation as a novel tool for therapeutics.
Collapse
|
786
|
Koshkin V, Ailles LE, Liu G, Krylov SN. Metabolic Suppression of a Drug-Resistant Subpopulation in Cancer Spheroid Cells. J Cell Biochem 2016; 117:59-65. [PMID: 26054050 DOI: 10.1002/jcb.25247] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 05/29/2015] [Indexed: 12/15/2022]
Abstract
Inhibition of metabolic features which distinguish cancer cells from their non-malignant counterparts is a promising approach to cancer treatment. Energy support for drug extrusion in multidrug resistance (MDR) is a potential target for metabolic inhibition. Two major sources of ATP-based metabolic energy are partial (glycolysis) and complete (mitochondrial oxidative phosphorylation) oxidation of metabolic fuels. In cancer cells, the balance between them tends to be shifted toward glycolysis; this shift is considered to be characteristic of the cancer metabolic phenotype. Numerous earlier studies, conducted with cells cultured in a monolayer (2-D model), suggested inhibition of glycolytic ATP production as an efficient tool to suppress MDR in cancer cells. Yet, more recent work challenged the appropriateness of the 2-D model for such studies and suggested that a more clinically relevant approach would utilize a more advanced cellular model such as a 3-D model. Here, we show that the transition from the 2-D model (cultured monolayer) to a 3-D model (cultured spheroids) introduces essential changes into the concept of energetic suppression of MDR. The 3-D cell organization leads to the formation of a discrete cell subpopulation (not formed in the 2-D model) with elevated MDR transport capacity. This subpopulation has a specific metabolic phenotype (mixed glycolytic/oxidative MDR support) different from that of cells cultured in the 2-D model. Finally, the shift to the oxidative phenotype becomes greater when the spheroids are grown under conditions of lactic acidosis that are typical for solid tumors. The potential clinical significance of these findings is discussed.
Collapse
Affiliation(s)
- Vasilij Koshkin
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario, Canada, M3J 1P3
| | - Laurie E Ailles
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada, N5G 1L7
| | - Geoffrey Liu
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Hospital, Toronto, Ontario, Canada, M5G 2C4
| | - Sergey N Krylov
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario, Canada, M3J 1P3
| |
Collapse
|
787
|
Chen B, Liu J, Ho TT, Ding X, Mo YY. ERK-mediated NF-κB activation through ASIC1 in response to acidosis. Oncogenesis 2016; 5:e279. [PMID: 27941930 PMCID: PMC5177778 DOI: 10.1038/oncsis.2016.81] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/30/2016] [Accepted: 11/03/2016] [Indexed: 12/16/2022] Open
Abstract
Acidic microenvironment is a common feature of solid tumors. We have previously shown that neuron specific acid-sensing ion channel 1 (ASIC1) is expressed in breast cancer, and it is responsible for acidosis-induced cellular signaling through AKT, leading to nuclear factor-κB (NF-κB) activation, and cell invasion and metastasis. However, AKT is frequently activated in cancer. Thus, a key question is whether ASIC1-mediated cell signaling still takes place in the cancer cells carrying constitutively active AKT. In the present study, we show that among four prostate cancer cell lines tested, 22Rv1 cells express the highest level of phosphorylated AKT that is not impacted by acidosis. However, acidosis can still induce NF-κB activation during which extracellular signal-regulated kinase (ERK) serves as an alternative pathway for ASIC-mediated cell signaling. Inhibition of ERK by chemical inhibitors or small interfering RNAs suppresses the acidosis-induced NF-κB activity through regulation of the inhibitory subunit IκBα phosphorylation. Furthermore, suppression of ASIC1-mediated generation of reactive oxygen species (ROS) by ROS scavengers, such as glutathione or N-acetyl-cysteine causes a decrease in ERK phosphorylation and degradation of IκBα. Finally, ASIC1 is upregulated in a subset of prostate cancer cases and ASIC1 knockout by CRISPR/Cas9 significantly suppresses cell invasion, and castration resistance both in vitro and in vivo. Together, these results support the significance of ASIC1-ROS-ERK-IκBα-NF-κB axis in prostate tumorigenesis, especially in the constitutively active AKT background.
Collapse
Affiliation(s)
- B Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - J Liu
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Emergency Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - T-T Ho
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| | - X Ding
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Y-Y Mo
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Pharmacology/Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
788
|
Kareva I, Abou-Slaybi A, Dodd O, Dashevsky O, Klement GL. Normal Wound Healing and Tumor Angiogenesis as a Game of Competitive Inhibition. PLoS One 2016; 11:e0166655. [PMID: 27935954 PMCID: PMC5147849 DOI: 10.1371/journal.pone.0166655] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/01/2016] [Indexed: 12/20/2022] Open
Abstract
Both normal wound healing and tumor angiogenesis are mitigated by the sequential, carefully orchestrated release of growth stimulators and inhibitors. These regulators are released from platelet clots formed at the sites of activated endothelium in a temporally and spatially controlled manner, and the order of their release depends on their affinity to glycosaminoglycans (GAG) such as heparan sulfate (HS) within the extracellular matrix, and platelet open canallicular system. The formation of vessel sprouts, triggered by angiogenesis regulating factors with lowest affinities for heparan sulfate (e.g. VEGF), is followed by vessel-stabilizing PDGF-B or bFGF with medium affinity for HS, and by inhibitors such as PF-4 and TSP-1 with the highest affinities for HS. The invasive wound-like edge of growing tumors has an overabundance of angiogenesis stimulators, and we propose that their abundance out-competes angiogenesis inhibitors, effectively preventing inhibition of angiogenesis and vessel maturation. We evaluate this hypothesis using an experimentally motivated agent-based model, and propose a general theoretical framework for understanding mechanistic similarities and differences between the processes of normal wound healing and pathological angiogenesis from the point of view of competitive inhibition.
Collapse
Affiliation(s)
- Irina Kareva
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Mathematical, Computational and Modeling Sciences Center, Arizona State Univ, Tempe, Arizona, United States of America
| | - Abdo Abou-Slaybi
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Oliver Dodd
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Massachusetts Institute of Technology, Boston, Massachusetts, United States of America
| | - Olga Dashevsky
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Dept. of Medical Oncology, Dana−Farber Cancer Institute, Dept. of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Giannoula Lakka Klement
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Pediatric Hematology Oncology, Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Sackler School of Graduate Biomedical Sciences at Tufts University, Boston, Massachusetts, United States of America
- * E-mail: ,
| |
Collapse
|
789
|
Faes S, Duval AP, Planche A, Uldry E, Santoro T, Pythoud C, Stehle JC, Horlbeck J, Letovanec I, Riggi N, Demartines N, Dormond O. Acidic tumor microenvironment abrogates the efficacy of mTORC1 inhibitors. Mol Cancer 2016; 15:78. [PMID: 27919264 PMCID: PMC5139076 DOI: 10.1186/s12943-016-0562-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 11/28/2016] [Indexed: 11/24/2022] Open
Abstract
Background Blocking the mechanistic target of rapamycin complex-1 (mTORC1) with chemical inhibitors such as rapamycin has shown limited clinical efficacy in cancer. The tumor microenvironment is characterized by an acidic pH which interferes with cancer therapies. The consequences of acidity on the anti-cancer efficacy of mTORC1 inhibitors have not been characterized and are thus the focus of our study. Methods Cancer cell lines were treated with rapamycin in acidic or physiological conditions and cell proliferation was investigated. The effect of acidity on mTORC1 activity was determined by Western blot. The anticancer efficacy of rapamycin in combination with sodium bicarbonate to increase the intratumoral pH was tested in two different mouse models and compared to rapamycin treatment alone. Histological analysis was performed on tumor samples to evaluate proliferation, apoptosis and necrosis. Results Exposing cancer cells to acidic pH in vitro significantly reduced the anti-proliferative effect of rapamycin. At the molecular level, acidity significantly decreased mTORC1 activity, suggesting that cancer cell proliferation is independent of mTORC1 in acidic conditions. In contrast, the activation of mitogen-activated protein kinase (MAPK) or AKT were not affected by acidity, and blocking MAPK or AKT with a chemical inhibitor maintained an anti-proliferative effect at low pH. In tumor mouse models, the use of sodium bicarbonate increased mTORC1 activity in cancer cells and potentiated the anti-cancer efficacy of rapamycin. Combining sodium bicarbonate with rapamycin resulted in increased tumor necrosis, increased cancer cell apoptosis and decreased cancer cell proliferation as compared to single treatment. Conclusions Taken together, these results emphasize the inefficacy of mTORC1 inhibitors in acidic conditions. They further highlight the potential of combining sodium bicarbonate with mTORC1 inhibitors to improve their anti-tumoral efficacy.
Collapse
Affiliation(s)
- Seraina Faes
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Adrian P Duval
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland.,Current Address: Swiss Institute of Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland
| | - Anne Planche
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Emilie Uldry
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Tania Santoro
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Catherine Pythoud
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Jean-Christophe Stehle
- Mouse Pathology Facility, Lausanne University Hospital CHUV and University of Lausanne, Lausanne, Switzerland
| | - Janine Horlbeck
- Mouse Pathology Facility, Lausanne University Hospital CHUV and University of Lausanne, Lausanne, Switzerland
| | - Igor Letovanec
- Institute of Pathology, Lausanne University Hospital CHUV and University of Lausanne, Lausanne, Switzerland
| | - Nicolo Riggi
- Institute of Pathology, Lausanne University Hospital CHUV and University of Lausanne, Lausanne, Switzerland
| | - Nicolas Demartines
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Olivier Dormond
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland.
| |
Collapse
|
790
|
Martin JD, Fukumura D, Duda DG, Boucher Y, Jain RK. Reengineering the Tumor Microenvironment to Alleviate Hypoxia and Overcome Cancer Heterogeneity. Cold Spring Harb Perspect Med 2016; 6:a027094. [PMID: 27663981 PMCID: PMC5131751 DOI: 10.1101/cshperspect.a027094] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Solid tumors consist of cancer cells and stromal cells, including resident and transiting immune cells-all ensconced in an extracellular matrix (ECM)-nourished by blood vessels and drained by lymphatic vessels. The microenvironment constituents are abnormal and heterogeneous in morphology, phenotype, and physiology. Such irregularities include an inefficient tumor vascular network comprised of leaky and compressed vessels, which impair blood flow and oxygen delivery. Low oxygenation in certain tumor regions-or focal hypoxia-is a mediator of cancer progression, metastasis, immunosuppression, and treatment resistance. Thus, repairing an abnormal and heterogeneous microenvironment-and hypoxia in particular-can significantly improve treatments of solid tumors. Here, we summarize two strategies to reengineer the tumor microenvironment (TME)-vessel normalization and decompression-that can alleviate hypoxia. In addition, we discuss how these two strategies alone and in combination with each other-or other therapeutic strategies-may overcome the challenges posed by cancer heterogeneity.
Collapse
Affiliation(s)
- John D Martin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Dan G Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Yves Boucher
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
791
|
Morandi A, Giannoni E, Chiarugi P. Nutrient Exploitation within the Tumor–Stroma Metabolic Crosstalk. Trends Cancer 2016; 2:736-746. [DOI: 10.1016/j.trecan.2016.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/31/2016] [Accepted: 11/01/2016] [Indexed: 01/01/2023]
|
792
|
Zhou Z, Song J, Nie L, Chen X. Reactive oxygen species generating systems meeting challenges of photodynamic cancer therapy. Chem Soc Rev 2016; 45:6597-6626. [PMID: 27722328 PMCID: PMC5118097 DOI: 10.1039/c6cs00271d] [Citation(s) in RCA: 1330] [Impact Index Per Article: 147.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The reactive oxygen species (ROS)-mediated mechanism is the major cause underlying the efficacy of photodynamic therapy (PDT). The PDT procedure is based on the cascade of synergistic effects between light, a photosensitizer (PS) and oxygen, which greatly favors the spatiotemporal control of the treatment. This procedure has also evoked several unresolved challenges at different levels including (i) the limited penetration depth of light, which restricts traditional PDT to superficial tumours; (ii) oxygen reliance does not allow PDT treatment of hypoxic tumours; (iii) light can complicate the phototherapeutic outcomes because of the concurrent heat generation; (iv) specific delivery of PSs to sub-cellular organelles for exerting effective toxicity remains an issue; and (v) side effects from undesirable white-light activation and self-catalysation of traditional PSs. Recent advances in nanotechnology and nanomedicine have provided new opportunities to develop ROS-generating systems through photodynamic or non-photodynamic procedures while tackling the challenges of the current PDT approaches. In this review, we summarize the current status and discuss the possible opportunities for ROS generation for cancer therapy. We hope this review will spur pre-clinical research and clinical practice for ROS-mediated tumour treatments.
Collapse
Affiliation(s)
- Zijian Zhou
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China. and Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Jibin Song
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Liming Nie
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
793
|
Díaz FE, Dantas E, Cabrera M, Benítez CA, Delpino MV, Duette G, Rubione J, Sanjuan N, Trevani AS, Geffner J. Fever-range hyperthermia improves the anti-apoptotic effect induced by low pH on human neutrophils promoting a proangiogenic profile. Cell Death Dis 2016; 7:e2437. [PMID: 27787523 PMCID: PMC5133997 DOI: 10.1038/cddis.2016.337] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/03/2016] [Accepted: 09/20/2016] [Indexed: 01/18/2023]
Abstract
Neutrophils have the shortest lifespan among leukocytes and usually die via apoptosis, limiting their deleterious potential. However, this tightly regulated cell death program can be modulated by pathogen-associated molecular patterns (PAMPs), danger-associated molecular pattern (DAMPs), and inflammatory cytokines. We have previously reported that low pH, a hallmark of inflammatory processes and solid tumors, moderately delays neutrophil apoptosis. Here we show that fever-range hyperthermia accelerates the rate of neutrophil apoptosis at neutral pH but markedly increases neutrophil survival induced by low pH. Interestingly, an opposite effect was observed in lymphocytes; hyperthermia plus low pH prevents lymphocyte activation and promotes the death of lymphocytes and lymphoid cell lines. Analysis of the mechanisms through which hyperthermia plus low pH increased neutrophil survival revealed that hyperthermia further decreases cytosolic pH induced by extracellular acidosis. The fact that two Na+/H+ exchanger inhibitors, 5-(N-ethyl-N-isopropyl) amiloride (EIPA) and amiloride, reproduced the effects induced by hyperthermia suggested that it prolongs neutrophil survival by inhibiting the Na+/H+ antiporter. The neutrophil anti-apoptotic effect induced by PAMPs, DAMPs, and inflammatory cytokines usually leads to the preservation of the major neutrophil effector functions such as phagocytosis and reactive oxygen species (ROS) production. In contrast, our data revealed that the anti-apoptotic effect induced by low pH and hyperthermia induced a functional profile characterized by a low phagocytic activity, an impairment in ROS production and a high ability to suppress T-cell activation and to produce the angiogenic factors VEGF, IL-8, and the matrix metallopeptidase 9 (MMP-9). These results suggest that acting together fever and local acidosis might drive the differentiation of neutrophils into a profile able to promote both cancer progression and tissue repair during the late phase of inflammation, two processes that are strongly dependent on the local production of angiogenic factors by infiltrating immune cells.
Collapse
Affiliation(s)
- Fernando Erra Díaz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ezequiel Dantas
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maia Cabrera
- Instituto de Investigaciones Farmacológicas (ININFA), CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Constanza A Benítez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María V Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriel Duette
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julia Rubione
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Norberto Sanjuan
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía S Trevani
- Instituto de Medicina Experimental (IMEX), CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
794
|
Paciotti GF, Zhao J, Cao S, Brodie PJ, Tamarkin L, Huhta M, Myer LD, Friedman J, Kingston DGI. Synthesis and Evaluation of Paclitaxel-Loaded Gold Nanoparticles for Tumor-Targeted Drug Delivery. Bioconjug Chem 2016; 27:2646-2657. [PMID: 27689882 DOI: 10.1021/acs.bioconjchem.6b00405] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The synthesis of a series of thiolated paclitaxel analogs is described as part of a novel nanomedicine program aimed at developing formulations of paclitaxel that will bind to gold nanoparticles for tumor targeted drug delivery. Preliminary evaluation of the new nanomedicine composed of 27 nm gold nanoparticles, tumor necrosis factor alpha (TNFα), thiolated polyethylene glycol (PEG-thiol), and one of several thiolated paclitaxel analogs is presented.
Collapse
Affiliation(s)
- Giulio F Paciotti
- CytImmune Sciences Inc. , 15010 Broschart Road, Rockville, Maryland 20850, United States
| | - Jielu Zhao
- Department of Chemistry and the Virginia Tech Center for Drug Discovery, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - Shugeng Cao
- Department of Chemistry and the Virginia Tech Center for Drug Discovery, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - Peggy J Brodie
- Department of Chemistry and the Virginia Tech Center for Drug Discovery, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - Lawrence Tamarkin
- CytImmune Sciences Inc. , 15010 Broschart Road, Rockville, Maryland 20850, United States
| | - Marja Huhta
- CytImmune Sciences Inc. , 15010 Broschart Road, Rockville, Maryland 20850, United States
| | - Lonnie D Myer
- CytImmune Sciences Inc. , 15010 Broschart Road, Rockville, Maryland 20850, United States
| | - Jay Friedman
- CytImmune Sciences Inc. , 15010 Broschart Road, Rockville, Maryland 20850, United States
| | - David G I Kingston
- Department of Chemistry and the Virginia Tech Center for Drug Discovery, Virginia Tech , Blacksburg, Virginia 24061, United States
| |
Collapse
|
795
|
Abstract
The highly regulated pH of cells and the less-regulated pH of the surrounding extracellular matrix (ECM) is the result of a delicate balance between metabolic processes and proton production, proton transportation, chemical buffering, and vascular removal of waste products. Malignant cells show a pronounced increase in metabolic processes where the 10- to 15-fold rise in glucose consumption is only the tip of the iceberg. Aerobic glycolysis (Warburg effect) is one of the hallmarks of cancer metabolism that implies excessive production of protons, which if stayed inside the cells would result in fatal intracellular acidosis (maintaining a strict acid-base balance is essential for the survival of eukaryotic cells). Malignant cells solve this problem by increasing mechanisms of proton transportation which expel the excess acidity. This allows cancer cells to keep a normal intracellular pH, or even overshooting this mechanism permits a slightly alkaline intracellular tendency. The proton excess expelled from malignant cells accumulates in the ECM, where chronic hypoxia and relative lack of enough blood vessels impede adequate proton clearance, thus creating an acidic microenvironment. This microenvironment is quite heterogeneous due to the tumor's metabolic heterogeneity and variable degrees of hypoxia inside the tumor mass. The acidic environment (plus other necessary cellular modifications) stimulates migration and invasion and finally intravasation of malignant cells which eventually may result in metastasis. Targeting tumor pH may go in two directions: 1) increasing extracellular pH which should result in less migration, invasion, and metastasis; and 2) decreasing intracellular pH which may result in acidic stress and apoptosis. Both objectives seem achievable at the present state of the art with repurposed drugs. This hypothesis analyzes the altered pH of tumors and its implications for progression and metastasis and also possible repurposed drug combinations targeting this vulnerable side of cancer development. It also analyzes the double-edged approach, which consists in pharmacologically increasing intracellular proton production and simultaneously decreasing proton extrusion creating intracellular acidity, acid stress, and eventual apoptosis.
Collapse
Affiliation(s)
- Tomas Koltai
- Obra Social del Personal de la, Industria Alimenticia, Filial Capital Federal, Republic of Argentina
| |
Collapse
|
796
|
Erdem M, Yalcin S, Gunduz U. Folic acid-conjugated polyethylene glycol-coated magnetic nanoparticles for doxorubicin delivery in cancer chemotherapy: Preparation, characterization and cytotoxicity on HeLa cell line. Hum Exp Toxicol 2016; 36:833-845. [PMID: 27758842 DOI: 10.1177/0960327116672910] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Conventional chemotherapy is the most valid method to cope with cancer; however, it has serious drawbacks such as decrease in production of blood cells or inflammation of the lining of the digestive tract. These side effects occur since generally the drugs used in chemotherapy are distributed evenly within the body of the patient and cannot distinguish the cancer cells from the healthy ones. In this study, folic acid (FA)-conjugated, polyethylene-coated magnetic nanoparticles (FA-MNPs), and doxorubicin (Dox)-loaded formulation (Dox-FA-MNPs) were prepared. The cytotoxicity of these nanoparticles on HeLa and Dox-resistant HeLa cells was investigated. Magnetic nanoparticles (MNPs), polyethylene glycol (PEG)-coated MNPs (PEG-MNPs), and FA-MNPs were successfully synthesized and characterized by several methods. Dox loading of FA-MNPs and release profile of Dox from the nanoparticles were studied. Cytotoxic effects of FA-MNPs and Dox-FA-MNPs on HeLa cells were analyzed. MNPs, PEG-MNPs, and FA-MNPs all had small sizes and supermagnetic behavior. High amounts of Dox could be loded onto the nanoparticles (290 μgmL-1). In 24 h, 15.7% of Dox was released from the Dox-FA-MNPs. The release was increased in acidic conditions (pH 4.1). Internalization studies showed that FA-MNPs and Dox-FA-MNPs were taken up efficiently by HeLa cells. The investigation of cytotoxicity of the particles indicated that 38-500 μgmL-1 Dox-FA-MNPs significantly decreased the proliferation of HeLa cells compared to FA-MNPs. Due to their size, magnetic properties, internalization, drug release, and cytotoxicity characteristics, the MNPs prepared in this study may have potential application as a drug delivery system in cancer chemotherapy.
Collapse
Affiliation(s)
- M Erdem
- 1 Department of Biology, Middle East Technical University, Ankara, Turkey
| | - S Yalcin
- 2 Department of Food Engineering, Ahi Evran University, Kirşehir, Turkey
| | - U Gunduz
- 3 Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
797
|
Ravichandran M, Oza G, Velumani S, Ramirez JT, Garcia-Sierra F, Andrade NB, Vera A, Leija L, Garza-Navarro MA. Plasmonic/Magnetic Multifunctional nanoplatform for Cancer Theranostics. Sci Rep 2016; 6:34874. [PMID: 27721391 PMCID: PMC5056510 DOI: 10.1038/srep34874] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/09/2016] [Indexed: 12/18/2022] Open
Abstract
A multifunctional magneto-plasmonic CoFe2O4@Au core-shell nanoparticle was developed by iterative-seeding based method. This nanocargo consists of a cobalt ferrite kernel as a core (Nk) and multiple layers of gold as a functionalizable active stratum, (named as Nk@A after fifth iteration). Nk@A helps in augmenting the physiological stability and enhancing surface plasmon resonance (SPR) property. The targeted delivery of Doxorubicin using Nk@A as a nanopayload is demonstrated in this report. The drug release profile followed first order rate kinetics optimally at pH 5.4, which is considered as an endosomal pH of cells. The cellular MR imaging showed that Nk@A is an efficient T2 contrast agent for both L6 (r2-118.08 mM-1s-1) and Hep2 (r2-217.24 mM-1s-1) cells. Microwave based magnetic hyperthermia studies exhibited an augmentation in the temperature due to the transformation of radiation energy into heat at 2.45 GHz. There was an enhancement in cancer cell cytotoxicity when hyperthermia combined with chemotherapy. Hence, this single nanoplatform can deliver 3-pronged theranostic applications viz., targeted drug-delivery, T2 MR imaging and hyperthermia.
Collapse
Affiliation(s)
- M. Ravichandran
- Program on Nanoscience and Nanotechnology, Av. 2508 National Polytechnic Institute, Gustavo A. Madero, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Goldie Oza
- Department of Genetics and Molecular Biology, Av. 2508 National Polytechnic Institute, Gustavo A. Madero, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - S. Velumani
- Department of Electrical Engineering, Av. 2508 National Polytechnic Institute, Gustavo A. Madero, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Jose Tapia Ramirez
- Department of Genetics and Molecular Biology, Av. 2508 National Polytechnic Institute, Gustavo A. Madero, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Francisco Garcia-Sierra
- Department of Cell Biology, Av. 2508 National Polytechnic Institute, Gustavo A. Madero, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - Norma Barragan Andrade
- Department of Cell Biology, Av. 2508 National Polytechnic Institute, Gustavo A. Madero, San Pedro Zacatenco, 07360 Mexico City, Mexico
| | - A. Vera
- Department of Electrical Engineering - Bioelectronics Section, CINVESTAV-IPN, Av. 2508 National Polytechnic Institute, Gustavo A. Madero, San Pedro Zacatenco, 07360 Mexico City
| | - L. Leija
- Department of Electrical Engineering - Bioelectronics Section, CINVESTAV-IPN, Av. 2508 National Polytechnic Institute, Gustavo A. Madero, San Pedro Zacatenco, 07360 Mexico City
| | - Marco A. Garza-Navarro
- Department of Mechanical and Electrical Engineering, Universidad Autonoma de Nuevo Leon, San Nicolás de Los Garza, Nuevo León, 66451 Mexico City, Mexico
| |
Collapse
|
798
|
Teo JY, Chin W, Ke X, Gao S, Liu S, Cheng W, Hedrick JL, Yang YY. pH and redox dual-responsive biodegradable polymeric micelles with high drug loading for effective anticancer drug delivery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:431-442. [PMID: 27720991 DOI: 10.1016/j.nano.2016.09.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/14/2016] [Accepted: 09/19/2016] [Indexed: 01/20/2023]
Abstract
Diblock copolymers of poly(ethylene glycol) (PEG) and biodegradable polycarbonate functionalized with GSH-sensitive disulfide bonds and pH-responsive carboxylic acid groups were synthesized via organocatalytic ring-opening polymerization of functional cyclic carbonates with PEG having different molecular weights as macroinitiators. These narrowly-dispersed polymers had predictable molecular weights, and were used to load doxorubicin (DOX) into micelles primarily through ionic interactions. The DOX-loaded micelles exhibited the requisite small particle size (<100 nm), narrow size distribution and high drug loading capacity. When exposed to endolysosomal pH of 5.0, drug release was accelerated by at least two-fold. The introduction of GSH further expedited DOX release. Effective DOX release enhanced cytotoxicity against cancer cells. More importantly, the DOX-loaded micelles with the optimized composition showed excellent antitumor efficacy in nude mice bearing BT-474 xenografts without inducing toxicity. These pH and redox dual-responsive micelles have the potential as delivery carriers to maximize the therapeutic effect of anticancer drugs.
Collapse
Affiliation(s)
- Jye Yng Teo
- Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore
| | - Willy Chin
- Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore
| | - Xiyu Ke
- Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore
| | - Shujun Gao
- Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore
| | - Shaoqiong Liu
- Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore
| | - Wei Cheng
- Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore
| | | | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore.
| |
Collapse
|
799
|
Si J, Shao S, Shen Y, Wang K. Macrophages as Active Nanocarriers for Targeted Early and Adjuvant Cancer Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:5108-5119. [PMID: 27560388 DOI: 10.1002/smll.201601282] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/22/2016] [Indexed: 05/18/2023]
Abstract
Taking advantage of the highly permeable vasculature and lack of lymphatic drainage in solid tumors (EPR effect), nanosized drug delivery systems or nanomedicines have been extensively explored for tumor-targeted drug delivery. However, in most clinical cases tumors such as the early stage tumors and post-surgery microscopic residual tumors have not yet developed such pathological EPR features, i.e., EPR-deficient. Therefore, nanomedicines may not be applicable for such these tumors. Macrophages by nature can actively home and extravasate through the tight vascular wall into tumors and migrate to their hypoxic regions, and possess perfect stealth ability for long blood circulation and impressive phagocytosis for drug loadings. Thus, nanomedicines loaded in macrophages would harness both merits and gain the active tumor homing capability independent of the EPR effect for treatments of the EPR-deficient tumors. Herein, the critical considerations, current progress, challenges and future prospects of macrophages as carriers for nanomedicines are summarized, aiming at rational design of EPR-independent tumor-targeting active nanomedicines for targeted early and adjuvant cancer chemotherapy.
Collapse
Affiliation(s)
- Jingxing Si
- Department of Respiratory Medicine, The Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Shiqun Shao
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Kai Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
800
|
Huang Y, Coman D, Herman P, Rao JU, Maritim S, Hyder F. Towards longitudinal mapping of extracellular pH in gliomas. NMR IN BIOMEDICINE 2016; 29:1364-1372. [PMID: 27472471 PMCID: PMC5035200 DOI: 10.1002/nbm.3578] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 06/06/2023]
Abstract
Biosensor imaging of redundant deviation in shifts (BIRDS), an ultrafast chemical shift imaging technique, requires infusion of paramagnetic probes such as 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetrakis methylene phosphonate (DOTP(8-) ) complexed with thulium (Tm(3+) ) ion (i.e. TmDOTP(5-) ), where the pH-sensitive resonances of hyperfine-shifted non-exchangeable protons contained within the paramagnetic probe are detected. While imaging extracellular pH (pHe ) with BIRDS meets an important cancer research need by mapping the intratumoral-peritumoral pHe gradient, the surgical intervention used to raise the probe's plasma concentration limits longitudinal scans on the same subject. Here we describe using probenecid (i.e. an organic anion transporter inhibitor) to temporarily restrict renal clearance of TmDOTP(5-) , thereby facilitating molecular imaging by BIRDS without surgical intervention. Co-infusion of probenecid with TmDOTP(5-) increased the probe's distribution into various organs, including the brain, compared with infusing TmDOTP(5-) alone. In vivo BIRDS data using the probenecid-TmDOTP(5-) co-infusion method in rats bearing RG2, 9 L, and U87 brain tumors showed intratumoral-peritumoral pHe gradients that were unaffected by the probe dose. This co-infusion method can be used for pHe mapping with BIRDS in preclinical models for tumor characterization and therapeutic monitoring, given the possibility of repeated scans with BIRDS (e.g. over days and even weeks) in the same subject. The longitudinal pHe readout by the probenecid-TmDOTP(5-) co-infusion method for BIRDS adds translational value in tumor assessment and treatment. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yuegao Huang
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA.
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA.
| | - Daniel Coman
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Peter Herman
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Jyotsna U Rao
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Samuel Maritim
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Fahmeed Hyder
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA.
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA.
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|