1
|
Kumari S, Sharma S, Advani D, Khosla A, Kumar P, Ambasta RK. Unboxing the molecular modalities of mutagens in cancer. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62111-62159. [PMID: 34611806 PMCID: PMC8492102 DOI: 10.1007/s11356-021-16726-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 09/22/2021] [Indexed: 04/16/2023]
Abstract
The etiology of the majority of human cancers is associated with a myriad of environmental causes, including physical, chemical, and biological factors. DNA damage induced by such mutagens is the initial step in the process of carcinogenesis resulting in the accumulation of mutations. Mutational events are considered the major triggers for introducing genetic and epigenetic insults such as DNA crosslinks, single- and double-strand DNA breaks, formation of DNA adducts, mismatched bases, modification in histones, DNA methylation, and microRNA alterations. However, DNA repair mechanisms are devoted to protect the DNA to ensure genetic stability, any aberrations in these calibrated mechanisms provoke cancer occurrence. Comprehensive knowledge of the type of mutagens and carcinogens and the influence of these agents in DNA damage and cancer induction is crucial to develop rational anticancer strategies. This review delineated the molecular mechanism of DNA damage and the repair pathways to provide a deep understanding of the molecular basis of mutagenicity and carcinogenicity. A relationship between DNA adduct formation and cancer incidence has also been summarized. The mechanistic basis of inflammatory response and oxidative damage triggered by mutagens in tumorigenesis has also been highlighted. We elucidated the interesting interplay between DNA damage response and immune system mechanisms. We addressed the current understanding of DNA repair targeted therapies and DNA damaging chemotherapeutic agents for cancer treatment and discussed how antiviral agents, anti-inflammatory drugs, and immunotherapeutic agents combined with traditional approaches lay the foundations for future cancer therapies.
Collapse
Affiliation(s)
- Smita Kumari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Sudhanshu Sharma
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Akanksha Khosla
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
2
|
Siddiqui I, Bilkey J, McKee TD, Serra S, Pintilie M, Do T, Xu J, Tsao MS, Gallinger S, Hill RP, Hedley DW, Dhani NC. Digital quantitative tissue image analysis of hypoxia in resected pancreatic ductal adenocarcinomas. Front Oncol 2022; 12:926497. [PMID: 35978831 PMCID: PMC9376475 DOI: 10.3389/fonc.2022.926497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTumor hypoxia is theorized to contribute to the aggressive biology of pancreatic ductal adenocarcinoma (PDAC). We previously reported that hypoxia correlated with rapid tumor growth and metastasis in patient-derived xenografts. Anticipating a prognostic relevance of hypoxia in patient tumors, we developed protocols for automated semi-quantitative image analysis to provide an objective, observer-independent measure of hypoxia. We further validated this method which can reproducibly estimate pimonidazole-detectable hypoxia in a high-through put manner.MethodsWe studied the performance of three automated image analysis platforms in scoring pimonidazole-detectable hypoxia in resected PDAC (n = 10) in a cohort of patients enrolled in PIMO-PANC. Multiple stained tumor sections were analyzed on three independent image-analysis platforms, Aperio Genie (AG), Definiens Tissue Studio (TS), and Definiens Developer (DD), which comprised of a customized rule set.ResultsThe output from Aperio Genie (AG) had good concordance with manual scoring, but the workflow was resource-intensive and not suited for high-throughput analysis. TS analysis had high levels of variability related to misclassification of cells class, while the customized rule set of DD had a high level of reliability with an intraclass coefficient of more than 85%.DiscussionThis work demonstrates the feasibility of developing a robust, high-performance pipeline for an automated, quantitative scoring of pimonidazole-detectable hypoxia in patient tumors.
Collapse
Affiliation(s)
- Iram Siddiqui
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- *Correspondence: Iram Siddiqui,
| | - Jade Bilkey
- Spatio-temporal Targeting and Amplification of Radiation Response (STTARR), University Health Network, Toronto, ON, Canada
| | - Trevor D. McKee
- Spatio-temporal Targeting and Amplification of Radiation Response (STTARR), University Health Network, Toronto, ON, Canada
| | - Stefano Serra
- Department of Pathology, Toronto General Hospital, Toronto, ON, Canada
| | - Melania Pintilie
- Department of Biostatistics, The Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Trevor Do
- Spatio-temporal Targeting and Amplification of Radiation Response (STTARR), University Health Network, Toronto, ON, Canada
| | - Jing Xu
- Department of Medical Oncology, The Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Ming-Sound Tsao
- Department of Pathology, Toronto General Hospital, Toronto, ON, Canada
| | - Steve Gallinger
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Hepato-Pancreatico-Biliary Surgical Oncology Program, University Health Network, Toronto, ON, Canada
| | - Richard P. Hill
- Medicine Program, The Princess Margaret Cancer Centre/Ontario Cancer Institute, Radiation Toronto, ON, Canada
| | - David W. Hedley
- Department of Medical Oncology, The Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Neesha C. Dhani
- Department of Medical Oncology, The Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
3
|
Wadsworth BJ, Lee CM, Bennewith KL. Transiently hypoxic tumour cell turnover and radiation sensitivity in human tumour xenografts. Br J Cancer 2022; 126:1616-1626. [PMID: 35031765 PMCID: PMC9130130 DOI: 10.1038/s41416-021-01691-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/24/2021] [Accepted: 12/23/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Solid tumour perfusion can be unstable, creating transiently hypoxic cells that can contribute to radiation resistance. We investigated the in vivo lifetime of transiently hypoxic tumour cells and chronically hypoxic tumour cells during tumour growth and following irradiation. METHODS Hypoxic cells in SiHa and WiDr human tumour xenografts were labelled using pimonidazole and EF5, and turnover was quantified as the loss of labelled cells over time. The perfusion-modifying drug pentoxifylline was used to reoxygenate transiently hypoxic cells prior to hypoxia marker administration or irradiation. RESULTS Chronically hypoxic cells constantly turnover in SiHa and WiDr tumours, with half-lives ranging from 42-82 h and significant numbers surviving >96 h. Transiently hypoxic cells constitute 26% of the total hypoxic cells in WiDr tumours. These transiently hypoxic cells survive at least 24 h, but then rapidly turnover with a half-life of 34 h and are undetectable 72 h after labelling. Transiently hypoxic cells are radiation-resistant, although vascular dysfunction induced by 10 Gy of ionising radiation preferentially kills transiently hypoxic cells. CONCLUSIONS Transiently hypoxic tumour cells survive up to 72 h in WiDr tumours and are radiation-resistant, although transiently hypoxic cells are sensitive to vascular dysfunction induced by high doses of ionising radiation.
Collapse
Affiliation(s)
- Brennan J. Wadsworth
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Che-Min Lee
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC Canada
| | - Kevin L. Bennewith
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
4
|
Datta A, West C, O'Connor JPB, Choudhury A, Hoskin P. Impact of hypoxia on cervical cancer outcomes. Int J Gynecol Cancer 2021; 31:1459-1470. [PMID: 34593564 DOI: 10.1136/ijgc-2021-002806] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/14/2021] [Indexed: 01/22/2023] Open
Abstract
The annual global incidence of cervical cancer is approximately 604 000 cases/342 000 deaths, making it the fourth most common cancer in women. Cervical cancer is a major healthcare problem in low and middle income countries where 85% of new cases and deaths occur. Secondary prevention measures have reduced incidence and mortality in developed countries over the past 30 years, but cervical cancer remains a major cause of cancer deaths in women. For women who present with Fédération Internationale de Gynécologie et d'Obstétrique (FIGO 2018) stages IB3 or upwards, chemoradiation is the established treatment. Despite high rates of local control, overall survival is less than 50%, largely due to distant relapse. Reducing the health burden of cervical cancer requires greater individualization of treatment, identifying those at risk of relapse and progression for modified or intensified treatment. Hypoxia is a well known feature of solid tumors and an established therapeutic target. Low tumorous oxygenation increases the risk of local invasion, metastasis and treatment failure. While meta-analyses show benefit, many individual trials targeting hypoxia failed in part due to not selecting patients most likely to benefit. This review summarizes the available hypoxia-targeted strategies and identifies further research and new treatment paradigms needed to improve patient outcomes. The applications and limitations of hypoxia biomarkers for treatment selection and response monitoring are discussed. Finally, areas of greatest unmet clinical need are identified to measure and target hypoxia and therefore improve cervical cancer outcomes.
Collapse
Affiliation(s)
- Anubhav Datta
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Radiology, The Christie NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
| | - James P B O'Connor
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Oncology, The Christie Hospital NHS Trust, Manchester, UK
| | - Peter Hoskin
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Oncology, Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| |
Collapse
|
5
|
Piscatelli JA, Ban J, Lucas AT, Zamboni WC. Complex Factors and Challenges that Affect the Pharmacology, Safety and Efficacy of Nanocarrier Drug Delivery Systems. Pharmaceutics 2021; 13:114. [PMID: 33477395 PMCID: PMC7830329 DOI: 10.3390/pharmaceutics13010114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/01/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Major developments in nanomedicines, such as nanoparticles (NPs), nanosomes, and conjugates, have revolutionized drug delivery capabilities over the past four decades. Although nanocarrier agents provide numerous advantages (e.g., greater solubility and duration of systemic exposure) compared to their small-molecule counterparts, there is considerable inter-patient variability seen in the systemic disposition, tumor delivery and overall pharmacological effects (i.e., anti-tumor efficacy and unwanted toxicity) of NP agents. This review aims to provide a summary of fundamental factors that affect the disposition of NPs in the treatment of cancer and why they should be evaluated during preclinical and clinical development. Furthermore, this chapter will highlight some of the translational challenges associated with elements of NPs and how these issues can only be addressed by detailed and novel pharmacology studies.
Collapse
Affiliation(s)
- Joseph A. Piscatelli
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
| | - Jisun Ban
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
| | - Andrew T. Lucas
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Imaging Hypoxia. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00074-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
7
|
Bader SB, Dewhirst MW, Hammond EM. Cyclic Hypoxia: An Update on Its Characteristics, Methods to Measure It and Biological Implications in Cancer. Cancers (Basel) 2020; 13:E23. [PMID: 33374581 PMCID: PMC7793090 DOI: 10.3390/cancers13010023] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
Regions of hypoxia occur in most if not all solid cancers. Although the presence of tumor hypoxia is a common occurrence, the levels of hypoxia and proportion of the tumor that are hypoxic vary significantly. Importantly, even within tumors, oxygen levels fluctuate due to changes in red blood cell flux, vascular remodeling and thermoregulation. Together, this leads to cyclic or intermittent hypoxia. Tumor hypoxia predicts for poor patient outcome, in part due to increased resistance to all standard therapies. However, it is less clear how cyclic hypoxia impacts therapy response. Here, we discuss the causes of cyclic hypoxia and, importantly, which imaging modalities are best suited to detecting cyclic vs. chronic hypoxia. In addition, we provide a comparison of the biological response to chronic and cyclic hypoxia, including how the levels of reactive oxygen species and HIF-1 are likely impacted. Together, we highlight the importance of remembering that tumor hypoxia is not a static condition and that the fluctuations in oxygen levels have significant biological consequences.
Collapse
Affiliation(s)
- Samuel B. Bader
- Department of Oncology, The Oxford Institute for Radiation Oncology, Oxford University, Oxford OX3 7DQ, UK;
| | - Mark W. Dewhirst
- Radiation Oncology Department, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ester M. Hammond
- Department of Oncology, The Oxford Institute for Radiation Oncology, Oxford University, Oxford OX3 7DQ, UK;
| |
Collapse
|
8
|
Simões JCS, Sarpaki S, Papadimitroulas P, Therrien B, Loudos G. Conjugated Photosensitizers for Imaging and PDT in Cancer Research. J Med Chem 2020; 63:14119-14150. [PMID: 32990442 DOI: 10.1021/acs.jmedchem.0c00047] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Early cancer detection and perfect understanding of the disease are imperative toward efficient treatments. It is straightforward that, for choosing a specific cancer treatment methodology, diagnostic agents undertake a critical role. Imaging is an extremely intriguing tool since it assumes a follow up to treatments to survey the accomplishment of the treatment and to recognize any conceivable repeating injuries. It also permits analysis of the disease, as well as to pursue treatment and monitor the possible changes that happen on the tumor. Likewise, it allows screening the adequacy of treatment and visualizing the state of the tumor. Additionally, when the treatment is finished, observing the patient is imperative to evaluate the treatment methodology and adjust the treatment if necessary. The goal of this review is to present an overview of conjugated photosensitizers for imaging and therapy.
Collapse
Affiliation(s)
- João C S Simões
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland.,BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | - Sophia Sarpaki
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | | | - Bruno Therrien
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland
| | - George Loudos
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| |
Collapse
|
9
|
Zhu H, Li Q, Shi B, Ge F, Liu Y, Mao Z, Zhu H, Wang S, Yu G, Huang F, Stang PJ. Dual-Emissive Platinum(II) Metallacage with a Sensitive Oxygen Response for Imaging of Hypoxia and Imaging-Guided Chemotherapy. Angew Chem Int Ed Engl 2020; 59:20208-20214. [PMID: 32710650 DOI: 10.1002/anie.202009442] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Indexed: 12/17/2022]
Abstract
Imaging of hypoxia in vivo helps with accurate cancer diagnosis and evaluation of therapeutic outcomes. A PtII metallacage with oxygen-responsive red phosphorescence and steady fluorescence for in vivo hypoxia imaging and chemotherapy is reported. The therapeutic agent and diagnostic probe were integrated into the metallacage through heteroligation-directed self-assembly. Nanoformulation by encapsulating the metallacage into nanoparticles greatly enhanced its stability the in physiological environment, rendering biomedical applications feasible. Apart from enhanced red phosphorescence upon hypoxia, the ratio between red and blue emissions, which only varies with intracellular oxygen level, provides a more precise standard for hypoxia imaging and detection. Moreover, in vivo explorations demonstrate the promising potential applications of the metallacage-loaded nanoparticles as theranostic agents for tumor hypoxia imaging and chemotherapy.
Collapse
Affiliation(s)
- Huangtianzhi Zhu
- State Key Laboratory of Chemical Engineering, Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China.,Department of Chemistry, University of Utah, Salt Lake City, UT, 84112, USA
| | - Qi Li
- State Key Laboratory of Chemical Engineering, Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China.,Department of Chemistry, University of Utah, Salt Lake City, UT, 84112, USA
| | - Bingbing Shi
- Department of Chemistry, University of Utah, Salt Lake City, UT, 84112, USA
| | - Fujing Ge
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Yuezhou Liu
- State Key Laboratory of Chemical Engineering, Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Hong Zhu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Sheng Wang
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin, 300072, P. R. China
| | - Guocan Yu
- State Key Laboratory of Chemical Engineering, Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Feihe Huang
- State Key Laboratory of Chemical Engineering, Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou, 310027, P. R. China.,Green Catalysis Center and College of Chemistry, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Peter J Stang
- Department of Chemistry, University of Utah, Salt Lake City, UT, 84112, USA
| |
Collapse
|
10
|
Zhu H, Li Q, Shi B, Ge F, Liu Y, Mao Z, Zhu H, Wang S, Yu G, Huang F, Stang PJ. Dual‐Emissive Platinum(II) Metallacage with a Sensitive Oxygen Response for Imaging of Hypoxia and Imaging‐Guided Chemotherapy. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202009442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Huangtianzhi Zhu
- State Key Laboratory of Chemical Engineering Center for Chemistry of High-Performance & Novel Materials Department of Chemistry Zhejiang University Hangzhou 310027 P. R. China
- Department of Chemistry University of Utah Salt Lake City UT 84112 USA
| | - Qi Li
- State Key Laboratory of Chemical Engineering Center for Chemistry of High-Performance & Novel Materials Department of Chemistry Zhejiang University Hangzhou 310027 P. R. China
- Department of Chemistry University of Utah Salt Lake City UT 84112 USA
| | - Bingbing Shi
- Department of Chemistry University of Utah Salt Lake City UT 84112 USA
| | - Fujing Ge
- College of Pharmaceutical Science Zhejiang University Hangzhou 310058 P. R. China
| | - Yuezhou Liu
- State Key Laboratory of Chemical Engineering Center for Chemistry of High-Performance & Novel Materials Department of Chemistry Zhejiang University Hangzhou 310027 P. R. China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 P. R. China
| | - Hong Zhu
- College of Pharmaceutical Science Zhejiang University Hangzhou 310058 P. R. China
| | - Sheng Wang
- School of Life Sciences Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology Tianjin 300072 P. R. China
| | - Guocan Yu
- State Key Laboratory of Chemical Engineering Center for Chemistry of High-Performance & Novel Materials Department of Chemistry Zhejiang University Hangzhou 310027 P. R. China
| | - Feihe Huang
- State Key Laboratory of Chemical Engineering Center for Chemistry of High-Performance & Novel Materials Department of Chemistry Zhejiang University Hangzhou 310027 P. R. China
- Green Catalysis Center and College of Chemistry Zhengzhou University Zhengzhou 450001 P. R. China
| | - Peter J. Stang
- Department of Chemistry University of Utah Salt Lake City UT 84112 USA
| |
Collapse
|
11
|
Virani NA, Kelada OJ, Kunjachan S, Detappe A, Kwon J, Hayashi J, Vazquez-Pagan A, Biancur DE, Ireland T, Kumar R, Sridhar S, Makrigiorgos GM, Berbeco RI. Noninvasive imaging of tumor hypoxia after nanoparticle-mediated tumor vascular disruption. PLoS One 2020; 15:e0236245. [PMID: 32706818 PMCID: PMC7380644 DOI: 10.1371/journal.pone.0236245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/01/2020] [Indexed: 01/09/2023] Open
Abstract
We have previously demonstrated that endothelial targeting of gold nanoparticles followed by external beam irradiation can cause specific tumor vascular disruption in mouse models of cancer. The induced vascular damage may lead to changes in tumor physiology, including tumor hypoxia, thereby compromising future therapeutic interventions. In this study, we investigate the dynamic changes in tumor hypoxia mediated by targeted gold nanoparticles and clinical radiation therapy (RT). By using noninvasive whole-body fluorescence imaging, tumor hypoxia was measured at baseline, on day 2 and day 13, post-tumor vascular disruption. A 2.5-fold increase (P<0.05) in tumor hypoxia was measured two days after combined therapy, resolving by day 13. In addition, the combination of vascular-targeted gold nanoparticles and radiation therapy resulted in a significant (P<0.05) suppression of tumor growth. This is the first study to demonstrate the tumor hypoxic physiological response and recovery after delivery of vascular-targeted gold nanoparticles followed by clinical radiation therapy in a human non-small cell lung cancer athymic Foxn1nu mouse model.
Collapse
Affiliation(s)
- Needa A. Virani
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Olivia J. Kelada
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sijumon Kunjachan
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alexandre Detappe
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston Massachusetts, United States of America
| | - Jihun Kwon
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiation Oncology, Hokkaido University, Sapporo, Japan
| | - Jennifer Hayashi
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Nanomedicine Innovation Center and Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Ana Vazquez-Pagan
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Nanomedicine Innovation Center and Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Douglas E. Biancur
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston Massachusetts, United States of America
| | - Thomas Ireland
- LA-ICP-MS and ICP-ES Laboratories, Boston University, Boston, Massachusetts, United States of America
| | - Rajiv Kumar
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Nanomedicine Innovation Center and Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - Srinivas Sridhar
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Nanomedicine Innovation Center and Department of Physics, Northeastern University, Boston, Massachusetts, United States of America
| | - G. Mike Makrigiorgos
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ross I. Berbeco
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
12
|
Wright WS, Eshaq RS, Lee M, Kaur G, Harris NR. Retinal Physiology and Circulation: Effect of Diabetes. Compr Physiol 2020; 10:933-974. [PMID: 32941691 PMCID: PMC10088460 DOI: 10.1002/cphy.c190021] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In this article, we present a discussion of diabetes and its complications, including the macrovascular and microvascular effects, with the latter of consequence to the retina. We will discuss the anatomy and physiology of the retina, including aspects of metabolism and mechanisms of oxygenation, with the latter accomplished via a combination of the retinal and choroidal blood circulations. Both of these vasculatures are altered in diabetes, with the retinal circulation intimately involved in the pathology of diabetic retinopathy. The later stages of diabetic retinopathy involve poorly controlled angiogenesis that is of great concern, but in our discussion, we will focus more on several alterations in the retinal circulation occurring earlier in the progression of disease, including reductions in blood flow and a possible redistribution of perfusion that may leave some areas of the retina ischemic and hypoxic. Finally, we include in this article a more recent area of investigation regarding the diabetic retinal vasculature, that is, the alterations to the endothelial surface layer that normally plays a vital role in maintaining physiological functions. © 2020 American Physiological Society. Compr Physiol 10:933-974, 2020.
Collapse
Affiliation(s)
- William S Wright
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, South Carolina, USA
| | - Randa S Eshaq
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Minsup Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Gaganpreet Kaur
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
13
|
Jiang MS, Yin XY, Qin B, Xuan SY, Yuan XL, Yin H, Zhu C, Li X, Yang J, Du YZ, Luo LH, You J. Inhibiting Hypoxia and Chemotherapy-Induced Cancer Cell Metastasis under a Valid Therapeutic Effect by an Assistance of Biomimetic Oxygen Delivery. Mol Pharm 2019; 16:4530-4541. [DOI: 10.1021/acs.molpharmaceut.9b00663] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Meng-Shi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Xiao-Yi Yin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Bing Qin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Shao-Yan Xuan
- Department of Pharmacy, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang 312000, P. R. China
| | - Xiao-Ling Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Hang Yin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Chunqi Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Xiang Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Jie Yang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Yong-Zhong Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Li-Hua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| |
Collapse
|
14
|
Dünker N, Jendrossek V. Implementation of the Chick Chorioallantoic Membrane (CAM) Model in Radiation Biology and Experimental Radiation Oncology Research. Cancers (Basel) 2019; 11:cancers11101499. [PMID: 31591362 PMCID: PMC6826367 DOI: 10.3390/cancers11101499] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy (RT) is part of standard cancer treatment. Innovations in treatment planning and increased precision in dose delivery have significantly improved the therapeutic gain of radiotherapy but are reaching their limits due to biologic constraints. Thus, a better understanding of the complex local and systemic responses to RT and of the biological mechanisms causing treatment success or failure is required if we aim to define novel targets for biological therapy optimization. Moreover, optimal treatment schedules and prognostic biomarkers have to be defined for assigning patients to the best treatment option. The complexity of the tumor environment and of the radiation response requires extensive in vivo experiments for the validation of such treatments. So far in vivo investigations have mostly been performed in time- and cost-intensive murine models. Here we propose the implementation of the chick chorioallantoic membrane (CAM) model as a fast, cost-efficient model for semi high-throughput preclinical in vivo screening of the modulation of the radiation effects by molecularly targeted drugs. This review provides a comprehensive overview on the application spectrum, advantages and limitations of the CAM assay and summarizes current knowledge of its applicability for cancer research with special focus on research in radiation biology and experimental radiation oncology.
Collapse
Affiliation(s)
- Nicole Dünker
- Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, University Medicine Essen, 45122 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Medicine Essen, 45122 Essen, Germany.
| |
Collapse
|
15
|
Yamane T, Aikawa M, Yasuda M, Fukushima K, Seto A, Okamoto K, Koyama I, Kuji I. [ 18F]FMISO PET/CT as a preoperative prognostic factor in patients with pancreatic cancer. EJNMMI Res 2019; 9:39. [PMID: 31073705 PMCID: PMC6509312 DOI: 10.1186/s13550-019-0507-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/15/2019] [Indexed: 12/31/2022] Open
Abstract
Background While [18F]fluoromisonidazole (FMISO), a representative PET tracer to detect hypoxia, is reported to be able to prospect the prognosis after treatment for various types of cancers, the relation is unclear for pancreatic cancer. The aim of this study is to assess the feasibility of [18F]FMISO PET/CT as a preoperative prognostic factor in patients with pancreatic cancer. Methods Patients with pancreatic cancer who had been initially planned for surgery received [18F]FMISO PET/CT. Peak standardized uptake value (SUV) of the pancreatic tumor was divided by SUVpeak of the aorta, and tumor blood ratio using SUVpeak (TBRpeak) was calculated. After preoperative examination, surgeons finally decided the operability of the patients. TBRpeak was compared with hypoxia-inducible factor (HIF)-1α immunohistochemistry when the tissues were available. Furthermore, correlation of TBRpeak with the recurrence-free survival and the overall survival were evaluated by Kaplan-Meyer methods. Results We analyzed 25 patients with pancreatic adenocarcinoma (11 women and 14 men, median age, 73 years; range, 58–81 years), and observed for 39–1101 days (median, 369 days). Nine cases (36.0%) were identified as visually positive of pancreatic cancer on [18F]FMISO PET/CT images. TBRpeak of the negative cases was significantly lower than that of the positive cases (median 1.08, interquartile range (IQR) 1.02–1.15 vs median 1.50, IQR 1.25–1.73, p < 0.001), and the cutoff TBRpeak was calculated as 1.24. Five patients were finally considered inoperable. There was no significant difference in TBRpeak of inoperable and operable patients (median 1.48, IQR 1.06–1.98 vs median 1.12, IQR 1.05–1.21, p = 0.10). There was no significant difference between TBRpeak and HIF-1α expression (p = 0.22). The patients were dichotomized by the TBRpeak cutoff, and the higher group showed significantly shorter recurrence-free survival than the other (median 218 vs 441 days, p = 0.002). As for overall survival of 20 cases of operated patients, the higher TBRpeak group showed significantly shorter overall survival than the other (median survival, 415 vs > 1000 days, p = 0.04). Conclusions [18F]FMISO PET/CT has the possibility to be a preoperative prognostic factor in patients with pancreatic cancer. Electronic supplementary material The online version of this article (10.1186/s13550-019-0507-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tomohiko Yamane
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1108, Japan.
| | - Masayasu Aikawa
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1108, Japan
| | - Masanori Yasuda
- Department of Diagnostic Pathology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1108, Japan
| | - Kenji Fukushima
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1108, Japan
| | - Akira Seto
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1108, Japan
| | - Koujun Okamoto
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1108, Japan
| | - Isamu Koyama
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1108, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1108, Japan
| |
Collapse
|
16
|
Lin CM, Yu CF, Huang HY, Chen FH, Hong JH, Chiang CS. Distinct Tumor Microenvironment at Tumor Edge as a Result of Astrocyte Activation Is Associated With Therapeutic Resistance for Brain Tumor. Front Oncol 2019; 9:307. [PMID: 31106146 PMCID: PMC6498880 DOI: 10.3389/fonc.2019.00307] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/03/2019] [Indexed: 12/01/2022] Open
Abstract
Tumor vasculatures and hypoxia are critical tumor micro-environmental factors associated with tumor response to the therapy and heterogeneous in both time- and location-dependent manner. Using a murine orthotopic anaplastic astrocytoma model, ALTS1C1, this study showed that brain tumor edge had a very unique microenvironment, having higher microvascular density (MVD) and better vessel function than the tumor core, but on the other hand was also positive for hypoxia markers, such as pimonidazole (PIMO), hypoxia inducible factor-1α (HIF-1α), and carbonic anhydrase IV (CAIX). The hypoxia at tumor edge was transient, named as peripheral hypoxia, and caused by different mechanisms from the chronic hypoxia in tumor core. The correlation of CAIX staining with astrocyte activation marker, glial fibrillary acid protein (GFAP), at the tumor edge indicated the involvement of astrocyte activation on the development of peripheral hypoxia. Peripheral hypoxia was a specific trait of orthotopic brain tumors at tumor edge, regardless of tumor origin. The hypoxic cells were resistant to the therapy, regardless of their location. Surviving cells, particularly those at the hypoxic region of tumor edge, are likely the cause of tumor recurrence after the therapy. New therapeutic platform that targets cells in tumor edge is likely to achieve better treatment outcomes.
Collapse
Affiliation(s)
- Chiu-Min Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Fang Yu
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan, Taiwan.,Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Hsueh-Ya Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.,Education & Medical Research National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Fang-Hsin Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan, Taiwan.,Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan.,Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Ji-Hong Hong
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan, Taiwan.,Radiation Biology Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan.,Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.,Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan.,Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
17
|
Repeat FMISO-PET imaging weakly correlates with hypoxia-associated gene expressions for locally advanced HNSCC treated by primary radiochemotherapy. Radiother Oncol 2019; 135:43-50. [PMID: 31015169 DOI: 10.1016/j.radonc.2019.02.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/07/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hypoxia is an important factor of tumour resistance to radiotherapy, chemotherapy and potentially immunotherapy. It can be measured e.g. by positron emission tomography (PET) imaging or hypoxia-associated gene expressions from tumour biopsies. Here we correlate [18F]fluoromisonidazole (FMISO)-PET/CT imaging with hypoxia-associated gene expressions on a cohort of 50 head and neck squamous cell carcinoma (HNSCC) patients and compare their prognostic value for response to radiochemotherapy (RCTx). METHODS FMISO-PET/CT images of 50 HNSCC patients were acquired at four time-points before and during RCTx. For 42 of these patients, hypoxia-associated gene expressions were evaluated by nanoString technology based on a biopsy obtained before any treatment. The FMISO-PET parameters tumour-to-background ratio and hypoxic volume were correlated to the expressions of 58 hypoxia-associated genes using the Spearman correlation coefficient ρ. Three hypoxia-associated gene signatures were compared regarding their correlation with the FMISO-PET parameters using their median expression. In addition, the correlation with tumour volume was analysed. The impact of both hypoxia measurement methods on loco-regional tumour control (LRC) and overall survival (OS) was assessed by Cox regression. RESULTS The median expression of hypoxia-associated genes was weakly correlated to hypoxia measured by FMISO-PET imaging (ρ ≤ 0.43), with higher correlations to imaging after weeks 1 and 2 of treatment (p < 0.001). Moderate correlations were obtained between FMISO-PET imaging and tumour volume (ρ ≤ 0.69). Prognostic models for LRC and OS based on the FMISO-PET parameters could not be improved by including hypoxia classifiers. CONCLUSION We observed low correlations between hypoxia FMISO-PET parameters and expressions of hypoxia-associated genes. Since FMISO-PET showed a superior patient stratification, it may be the preferred biomarker over hypoxia-associated genes for stratifying patients with locally advanced HNSCC treated by primary RCTx.
Collapse
|
18
|
Yoon DW, Kim YS, Hwang S, Khalmuratova R, Lee M, Kim JH, Lee GY, Koh SJ, Park JW, Shin HW. Intermittent hypoxia promotes carcinogenesis in azoxymethane and dextran sodium sulfate-induced colon cancer model. Mol Carcinog 2019; 58:654-665. [PMID: 30575123 DOI: 10.1002/mc.22957] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 10/30/2018] [Accepted: 12/15/2018] [Indexed: 02/06/2023]
Abstract
Intermittent hypoxia (IH), a characteristic of obstructive sleep apnea, is known to promote cancer progression and aggressiveness in mouse models. However, little is known regarding the effect of IH on cancer initiation. Here, the effect of IH on carcinogenesis was explored in azoxymethane (AOM) and dextran sodium sulfate (DSS)-induced colon cancer models with three different protocols. In the first protocol, two other application time points (early or late initiation of IH) were applied. In the second protocol, mice were divided into only two groups, and then exposed to either N or IH conditions for 14 days. In the third protocol, a pharmacological inhibition study for anti-inflammation (5-aminosalicylate) or anti-oxidative stress (N-acetylcysteine [NAC]) was performed. The number of tumors was significantly higher in the IH-1 than in the N or IH-2 groups. 8-oxo-2'-deoxyguanosine (8-OHdG) levels were higher in tumors of the IH-1 group than in that of the N and IH-2 groups. Gene expression related to reactive oxygen species production was higher in the IH-1 group than in the N and IH-2 groups, and it showed a positive correlation with 8-OHdG levels. Prior to cancer development 8-OHdG levels were already elevated in colonic epithelial regions in the IH group, possibly due to an imbalance between oxidative stress and antioxidant systems. NAC treatment resulted in a significant reduction in the number of tumors in mice exposed to IH. In conclusion, IH promotes carcinogenesis in a chemically-induced colon cancer model where elevated 8-OHdG may contribute to the increased tumor induction.
Collapse
Affiliation(s)
- Dae Wui Yoon
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea
| | - Yi-Sook Kim
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul, Korea
| | - Soyoung Hwang
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul, Korea
| | - Roza Khalmuratova
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea
| | - Mingyu Lee
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul, Korea
| | - Jee Hyun Kim
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Gah Young Lee
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Joon Koh
- Division of Gastroenterology, Department of Internal Medicine, Seoul National University Boramae Hospital, Seoul, Korea
| | - Jong-Wan Park
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Woo Shin
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
19
|
Clinical and Pre-clinical Methods for Quantifying Tumor Hypoxia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:19-41. [PMID: 31201714 DOI: 10.1007/978-3-030-12734-3_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, a prevalent characteristic of most solid malignant tumors, contributes to diminished therapeutic responses and more aggressive phenotypes. The term hypoxia has two definitions. One definition would be a physiologic state where the oxygen partial pressure is below the normal physiologic range. For most normal tissues, the normal physiologic range is between 10 and 20 mmHg. Hypoxic regions develop when there is an imbalance between oxygen supply and demand. The impact of hypoxia on cancer therapeutics is significant: hypoxic tissue is 3× less radiosensitive than normoxic tissue, the impaired blood flow found in hypoxic tumor regions influences chemotherapy delivery, and the immune system is dependent on oxygen for functionality. Despite the clinical implications of hypoxia, there is not a universal, ideal method for quantifying hypoxia, particularly cycling hypoxia because of its complexity and heterogeneity across tumor types and individuals. Most standard imaging techniques can be modified and applied to measuring hypoxia and quantifying its effects; however, the benefits and challenges of each imaging modality makes imaging hypoxia case-dependent. In this chapter, a comprehensive overview of the preclinical and clinical methods for quantifying hypoxia is presented along with the advantages and disadvantages of each.
Collapse
|
20
|
Abstract
Purpose In this paper, we provide an overview of a life history theory and how it applies to cancer evolution. Recent Findings We review the literature on trade-offs in tumors, focusing on the trade-offs among cellular proliferation, survival, and motility. Trade-offs are critical natural constraints for almost all evolutionary processes. Many ecological studies show that trade-offs among these cellular functions maintain a genetic diversity. In addition, these trade-offs are not fixed, but rather can shift depending on the ecological circumstances in the microenvironment. This can lead to selection for the cellular capacity to respond to these differing microenvironments in ways that promote the fitness of the cancer cell. We relate these life history trade-offs to the recently developed Evo-Eco indexes and discuss how life history theory can help refine our measures of tumor evolution and ecology. Summary Life history theory provides a framework for understanding how the spatial and temporal variability in the tumor microenvironment—in particular resources and threats—affect trade-offs among cell survival, cell proliferation, and cell migration. We discuss how these trade-offs can potentially be leveraged in cancer therapy to increase the effectiveness of treatment.
Collapse
|
21
|
Hamming-Vrieze O, Navran A, Al-Mamgani A, Vogel WV. Biological PET-guided adaptive radiotherapy for dose escalation in head and neck cancer: a systematic review. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:349-368. [DOI: 10.23736/s1824-4785.18.03087-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
22
|
Lopez T, Ramirez A, Benitez C, Mustafa Z, Pham H, Sanchez R, Ge X. Selectivity Conversion of Protease Inhibitory Antibodies. Antib Ther 2018. [PMID: 30406213 PMCID: PMC7990135 DOI: 10.1093/abt/tby010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Solid tumors are inherently difficult to treat because of large regions of hypoxia and are often chemotherapy- or radiotherapy-resistant. It seems that cancer stem cells reside in hypoxic and adjacent necrotic tumor areas. Therefore, new treatments that are highly selective for tumors and can eradicate cells in both hypoxic and necrotic tumor regions are desirable. Antibody α-radioconjugates couple an α-emitting radionuclide with the specificity of a tumor-targeting monoclonal antibody. The large mass and energy of α-particles result in radiation dose delivery within a smaller area independent of oxygen concentration, thus matching key criteria for killing hypoxic tumor cells. With advances in radionuclide production and chelation chemistry, α-radioconjugate therapy is regaining interest as a cancer therapy. Here, we will review current literature examining radioconjugate therapy specifically targeting necrotic and hypoxic tumor cells and outline how α-radioconjugate therapy could be used to treat tumor regions harboring more resistant cancer cell types. Statement of Significance Tumor-targeting antibodies are excellent vehicles for the delivery of toxic payloads directly to the tumor site. Tumor hypoxia and necrosis promote treatment recurrence, resistance, and metastasis. Targeting these areas with antibody α-radioconjugates would aid in overcoming treatment resistance.
Collapse
Affiliation(s)
- Tyler Lopez
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Aaron Ramirez
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Chris Benitez
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Zahid Mustafa
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Henry Pham
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Ramon Sanchez
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| | - Xin Ge
- Department of Chemical and Environmental Engineering, Bourns College of Engineering, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
23
|
Staudacher AH, Liapis V, Brown MP. Selectivity Conversion of Protease Inhibitory Antibodies. Antib Ther 2018; 1:55-63. [PMID: 30406213 PMCID: PMC7990135 DOI: 10.1093/abt/tby008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 11/14/2022] Open
Abstract
Background: Proteases are one of the largest pharmaceutical targets for drug developments. Their dysregulations result in a wide variety of diseases. Because proteolytic networks usually consist of protease family members that share high structural and catalytic homology, distinguishing them using small molecule inhibitors is often challenging. To achieve specific inhibition, this study described a novel approach for the generation of protease inhibitory antibodies. As a proof of concept, we aimed to convert a matrix metalloproteinase (MMP)-14 specific inhibitor to MMP-9 specific inhibitory antibodies with high selectivity. Methods: An error-prone single-chain Fv (scFv) library of an MMP-14 inhibitor 3A2 was generated for yeast surface display. A dual-color competitive FACS was developed for selection on MMP-9 catalytic domain (cdMMP-9) and counter-selection on cdMMP-14 simultaneously, which were fused/conjugated with different fluorophores. Isolated MMP-9 inhibitory scFvs were biochemically characterized by inhibition assays on MMP-2/-9/-12/-14, proteolytic stability tests, inhibition mode determination, competitive ELISA with TIMP-2 (a native inhibitor of MMPs), and paratope mutagenesis assays. Results: We converted an MMP-14 specific inhibitor 3A2 into a panel of MMP-9 specific inhibitory antibodies with dramatic selectivity shifts of 690-4,500 folds. Isolated scFvs inhibited cdMMP-9 at nM potency with high selectivity over MMP-2/-12/-14 and exhibited decent proteolytic stability. Biochemical characterizations revealed that these scFvs were competitive inhibitors binding to cdMMP-9 near its reaction cleft via their CDR-H3s. Conclusions: This study developed a novel approach able to convert the selectivity of inhibitory antibodies among closely related protease family members. This methodology can be directly applied for mAbs inhibiting many proteases of biomedical importance.
Collapse
Affiliation(s)
- Alexander H Staudacher
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
- School of Medicine, University of Adelaide, Adelaide, Australia
| | - Vasilios Liapis
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
- School of Medicine, University of Adelaide, Adelaide, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, Australia
| |
Collapse
|
24
|
Marcu LG, Reid P, Bezak E. The Promise of Novel Biomarkers for Head and Neck Cancer from an Imaging Perspective. Int J Mol Sci 2018; 19:E2511. [PMID: 30149561 PMCID: PMC6165113 DOI: 10.3390/ijms19092511] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/18/2018] [Accepted: 08/23/2018] [Indexed: 01/25/2023] Open
Abstract
It is an agreed fact that overall survival among head and neck cancer patients has increased over the last decade. Several factors however, are still held responsible for treatment failure requiring more in-depth evaluation. Among these, hypoxia and proliferation-specific parameters are the main culprits, along with the more recently researched cancer stem cells. This paper aims to present the latest developments in the field of biomarkers for hypoxia, stemness and tumour proliferation, from an imaging perspective that includes both Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) as well as functional magnetic resonance imaging (MRI). Quantitative imaging of biomarkers is a prerequisite for accurate treatment response assessment, bringing us closer to the highly needed personalised therapy.
Collapse
Affiliation(s)
- Loredana G Marcu
- Faculty of Science, University of Oradea, 410087 Oradea, Romania.
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Paul Reid
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Eva Bezak
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA 5001, Australia.
- Department of Physics, University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
25
|
Leger S, Zwanenburg A, Pilz K, Zschaeck S, Zöphel K, Kotzerke J, Schreiber A, Zips D, Krause M, Baumann M, Troost EGC, Richter C, Löck S. CT imaging during treatment improves radiomic models for patients with locally advanced head and neck cancer. Radiother Oncol 2018; 130:10-17. [PMID: 30087056 DOI: 10.1016/j.radonc.2018.07.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 06/27/2018] [Accepted: 07/24/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE The development of radiomic risk models to predict clinical outcome is usually based on pre-treatment imaging, such as computed tomography (CT) scans used for radiation treatment planning. Imaging data acquired during the course of treatment may improve their prognostic performance. We compared the performance of radiomic risk models based on the pre-treatment CT and CT scans acquired in the second week of therapy. MATERIAL AND METHODS Treatment planning and second week CT scans of 78 head and neck squamous cell carcinoma patients treated with primary radiochemotherapy were collected. 1538 image features were extracted from each image. Prognostic models for loco-regional tumour control (LRC) and overall survival (OS) were built using 6 feature selection methods and 6 machine learning algorithms. Prognostic performance was assessed using the concordance index (C-Index). Furthermore, patients were stratified into risk groups and differences in LRC and OS were evaluated by log-rank tests. RESULTS The performance of radiomic risk model in predicting LRC was improved using the second week CT scans (C-Index: 0.79), in comparison to the pre-treatment CT scans (C-Index: 0.65). This was confirmed by Kaplan-Meier analyses, in which risk stratification based on the second week CT could be improved for LRC (p = 0.002) compared to pre-treatment CT (p = 0.063). CONCLUSION Incorporation of imaging during treatment may be a promising way to improve radiomic risk models for clinical treatment adaption, i.e., to select patients that may benefit from dose modification.
Collapse
Affiliation(s)
- Stefan Leger
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.
| | - Alex Zwanenburg
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Karoline Pilz
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Department of Radiotherapy, Hospital Dresden-Friedrichstadt, Germany
| | - Sebastian Zschaeck
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Charité Universitätsmedizin Berlin, Department of Radiation Oncology, Germany
| | - Klaus Zöphel
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, PET Center, Institute of Radiopharmaceutical Cancer Research, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, PET Center, Institute of Radiopharmaceutical Cancer Research, Germany
| | - Andreas Schreiber
- Department of Radiotherapy, Hospital Dresden-Friedrichstadt, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls Universität Tübingen, Germany
| | - Mechthild Krause
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology OncoRay, Germany
| | - Michael Baumann
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology OncoRay, Germany
| | - Esther G C Troost
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology OncoRay, Germany
| | - Christian Richter
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology OncoRay, Germany
| | - Steffen Löck
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| |
Collapse
|
26
|
Baird IR, Patrick BO, Skov KA, James BR. Nitroimidazoles with a halogen-containing side-chain. CAN J CHEM 2018. [DOI: 10.1139/cjc-2017-0604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Syntheses are reported for: nine 2-nitroimidazoles, the abbreviated names all beginning with E, based on derivation from Etanidazole); five 2-methyl-5-nitroimidazoles (M compounds, derived from Metronidazole); and five 2-methyl-4-nitroimidazoles (labelled 2M4N compounds). The nitroimidazoles all have an amide side-chain at the N1 atom of the imidazole, with 17 of them containing one to five halogen atoms. The aim is to study compounds for comparison with EF5 (the number showing the presence of five F-atoms), a previously reported, pentafluoropropylacetamide derivative of 2-nitroimidazole that is currently used as a hypoxia marker drug to detect cancerous tumours. The new compounds are characterized by standard methods, including X-ray structural data for the fluorinated MF5, 2M4NF5, and 2M4NF1(−1) species, with the “–1” indicating two C-atoms in an alkylamide chain rather than the three C-atoms in the propylacetamide of EF5. Intra- and inter-molecular H-bonding is seen in the solid state structures, likely an important property in biological use; another key property of the nitroimidazoles is their reduction potentials, and the measured CV data confirm that 2NO2Im compounds with longer side-chains and more F-atoms (like EF5) are worth investigating for possible activity as hypoxia-selective, bioreductive agents.
Collapse
Affiliation(s)
- Ian R. Baird
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| | - Brian O. Patrick
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| | - Kirsten A. Skov
- Advanced Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Brian R. James
- Department of Chemistry, The University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| |
Collapse
|
27
|
Lamy S, Muhire É, Annabi B. Antiproliferative efficacy of elderberries and elderflowers (Sambucus canadensis) on glioma and brain endothelial cells under normoxic and hypoxic conditions. J Funct Foods 2018; 40:164-179. [DOI: 10.1016/j.jff.2017.10.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
28
|
Zhou H, Arias-Ramos N, López-Larrubia P, Mason RP, Cerdán S, Pacheco-Torres J. Oxygenation Imaging by Nuclear Magnetic Resonance Methods. Methods Mol Biol 2018; 1718:297-313. [PMID: 29341016 DOI: 10.1007/978-1-4939-7531-0_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxygen monitoring is a topic of exhaustive research due to its central role in many biological processes, from energy metabolism to gene regulation. The ability to monitor in vivo the physiological distribution and the dynamics of oxygen from subcellular to macroscopic levels is a prerequisite to better understand the mechanisms associated with both normal and disease states (cancer, neurodegeneration, stroke, etc.). This chapter focuses on magnetic resonance imaging (MRI) based techniques to assess oxygenation in vivo. The first methodology uses injected fluorinated agents to provide quantitative pO2 measurements with high precision and suitable spatial and temporal resolution for many applications. The second method exploits changes in endogenous contrasts, i.e., deoxyhemoglobin and oxygen molecules through measurements of T 2* and T 1, in response to an intervention to qualitatively evaluate hypoxia and its potential modulation.
Collapse
Affiliation(s)
- Heling Zhou
- Prognostic Imaging Research Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Nuria Arias-Ramos
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas 'Alberto Sols' C.S.I.C./U.A.M., Madrid, Spain
| | - Ralph P Mason
- Prognostic Imaging Research Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas 'Alberto Sols' C.S.I.C./U.A.M., Madrid, Spain
| | - Jesús Pacheco-Torres
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, San Juan de Alicante, Alicante, Spain.
| |
Collapse
|
29
|
Does Hypoxia Cause Carcinogenic Iron Accumulation in Alcoholic Liver Disease (ALD)? Cancers (Basel) 2017; 9:cancers9110145. [PMID: 29068390 PMCID: PMC5704163 DOI: 10.3390/cancers9110145] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) is a leading health risk worldwide. Hepatic iron overload is frequently observed in ALD patients and it is an important and independent factor for disease progression, survival, and the development of primary liver cancer (HCC). At a systemic level, iron homeostasis is controlled by the liver-secreted hormone hepcidin. Hepcidin regulation is complex and still not completely understood. It is modulated by many pathophysiological conditions associated with ALD, such as inflammation, anemia, oxidative stress/H2O2, or hypoxia. Namely, the data on hypoxia-signaling of hepcidin are conflicting, which seems to be mainly due to interpretational limitations of in vivo data and methodological challenges. Hence, it is often overlooked that hepcidin-secreting hepatocytes are physiologically exposed to 2–7% oxygen, and that key oxygen species such as H2O2 act as signaling messengers in such a hypoxic environment. Indeed, with the recently introduced glucose oxidase/catalase (GOX/CAT) system it has been possible to independently study hypoxia and H2O2 signaling. First preliminary data indicate that hypoxia enhances H2O2-mediated induction of hepcidin, pointing towards oxidases such as NADPH oxidase 4 (NOX4). We here review and discuss novel concepts of hypoxia signaling that could help to better understand hepcidin-associated iron overload in ALD.
Collapse
|
30
|
Xu Z, Li XF, Zou H, Sun X, Shen B. 18F-Fluoromisonidazole in tumor hypoxia imaging. Oncotarget 2017; 8:94969-94979. [PMID: 29212283 PMCID: PMC5706929 DOI: 10.18632/oncotarget.21662] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/21/2017] [Indexed: 12/19/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that is closely associated with radiotherapy and chemotherapy resistance, metastasis and tumors prognosis. Thus, it is important to assess hypoxia in tumors for estimating prognosis and selecting appropriate treatment procedures. 18F-Fluoromisonidazole positron emission tomography (18F-FMISO PET) has been widely used to visualize tumor hypoxia in a comprehensive and noninvasive way, both in the clinical and preclinical settings. Here we review the concept, mechanisms and detection methods of tumor hypoxia. Furthermore, we discuss the correlation between 18F-FMISO PET and other detection methods, current applications of 18F-FMISO PET and the development prospects of this imaging technology.
Collapse
Affiliation(s)
- Zuoyu Xu
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiao-Feng Li
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyan Zou
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang, China
| | - Xilin Sun
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Baozhong Shen
- Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, Heilongjiang, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
31
|
Maley CC, Aktipis A, Graham TA, Sottoriva A, Boddy AM, Janiszewska M, Silva AS, Gerlinger M, Yuan Y, Pienta KJ, Anderson KS, Gatenby R, Swanton C, Posada D, Wu CI, Schiffman JD, Hwang ES, Polyak K, Anderson ARA, Brown JS, Greaves M, Shibata D. Classifying the evolutionary and ecological features of neoplasms. Nat Rev Cancer 2017; 17:605-619. [PMID: 28912577 PMCID: PMC5811185 DOI: 10.1038/nrc.2017.69] [Citation(s) in RCA: 259] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neoplasms change over time through a process of cell-level evolution, driven by genetic and epigenetic alterations. However, the ecology of the microenvironment of a neoplastic cell determines which changes provide adaptive benefits. There is widespread recognition of the importance of these evolutionary and ecological processes in cancer, but to date, no system has been proposed for drawing clinically relevant distinctions between how different tumours are evolving. On the basis of a consensus conference of experts in the fields of cancer evolution and cancer ecology, we propose a framework for classifying tumours that is based on four relevant components. These are the diversity of neoplastic cells (intratumoural heterogeneity) and changes over time in that diversity, which make up an evolutionary index (Evo-index), as well as the hazards to neoplastic cell survival and the resources available to neoplastic cells, which make up an ecological index (Eco-index). We review evidence demonstrating the importance of each of these factors and describe multiple methods that can be used to measure them. Development of this classification system holds promise for enabling clinicians to personalize optimal interventions based on the evolvability of the patient's tumour. The Evo- and Eco-indices provide a common lexicon for communicating about how neoplasms change in response to interventions, with potential implications for clinical trials, personalized medicine and basic cancer research.
Collapse
Affiliation(s)
- Carlo C Maley
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 1001 S. McAllister Ave, Tempe, Arizona 85287, USA
| | - Athena Aktipis
- Department of Psychology, Center for Evolution and Medicine, Arizona State University, 651 E. University Drive, Tempe, Arizona 85287, USA
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Amy M Boddy
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | - Michalina Janiszewska
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue D740C, Boston, Massachusetts 02215, USA
| | - Ariosto S Silva
- Department of Cancer Imaging and Metabolism, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Marco Gerlinger
- Centre for Evolution and Cancer, Division of Molecular Pathology, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Kenneth J Pienta
- Brady Urological Institute, The Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, Maryland 21287, USA
| | - Karen S Anderson
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 1001 S. McAllister Ave, Tempe, Arizona 85287, USA
| | - Robert Gatenby
- Cancer Biology and Evolution Program, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - David Posada
- Department of Biochemistry, Genetics and Immunology and Biomedical Research Center (CINBIO), University of Vigo, Spain; Galicia Sur Health Research Institute, Vigo, 36310, Spain
| | - Chung-I Wu
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois 60637, USA
| | - Joshua D Schiffman
- Departments of Pediatrics and Oncological Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, Utah 84108, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University and Duke Cancer Institute, 465 Seeley Mudd Building, Durham, North Carolina 27710, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue D740C, Boston, Massachusetts 02215, USA
| | - Alexander R A Anderson
- Integrated Mathematical Oncology Department, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Joel S Brown
- Integrated Mathematical Oncology Department, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK
| | - Darryl Shibata
- Department of Pathology, Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, NOR2424, Los Angeles, California 90033, USA
| |
Collapse
|
32
|
Yang R, Chen LH, Hansen LJ, Carpenter AB, Moure CJ, Liu H, Pirozzi CJ, Diplas BH, Waitkus MS, Greer PK, Zhu H, McLendon RE, Bigner DD, He Y, Yan H. Cic Loss Promotes Gliomagenesis via Aberrant Neural Stem Cell Proliferation and Differentiation. Cancer Res 2017; 77:6097-6108. [PMID: 28939681 DOI: 10.1158/0008-5472.can-17-1018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/21/2017] [Accepted: 09/08/2017] [Indexed: 12/26/2022]
Abstract
Inactivating mutations in the transcriptional repression factor Capicua (CIC) occur in approximately 50% of human oligodendrogliomas, but mechanistic links to pathogenesis are unclear. To address this question, we generated Cic-deficient mice and human oligodendroglioma cell models. Genetic deficiency in mice resulted in a partially penetrant embryonic or perinatal lethal phenotype, with the production of an aberrant proliferative neural population in surviving animals. In vitro cultured neural stem cells derived from Cic conditional knockout mice bypassed an EGF requirement for proliferation and displayed a defect in their potential for oligodendrocyte differentiation. Cic is known to participate in gene suppression that can be relieved by EGFR signal, but we found that cic also activated expression of a broad range of EGFR-independent genes. In an orthotopic mouse model of glioma, we found that Cic loss potentiated the formation and reduced the latency in tumor development. Collectively, our results define an important role for Cic in regulating neural cell proliferation and lineage specification, and suggest mechanistic explanations for how CIC mutations may impact the pathogenesis and therapeutic targeting of oligodendroglioma. Cancer Res; 77(22); 6097-108. ©2017 AACR.
Collapse
Affiliation(s)
- Rui Yang
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Lee H Chen
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Landon J Hansen
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Austin B Carpenter
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Casey J Moure
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Heng Liu
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Christopher J Pirozzi
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Bill H Diplas
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Matthew S Waitkus
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Paula K Greer
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Huishan Zhu
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Roger E McLendon
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Darell D Bigner
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina
| | - Yiping He
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina.
| | - Hai Yan
- Department of Pathology and the Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
33
|
Chang YCC, Ackerstaff E, Tschudi Y, Jimenez B, Foltz W, Fisher C, Lilge L, Cho H, Carlin S, Gillies RJ, Balagurunathan Y, Yechieli RL, Subhawong T, Turkbey B, Pollack A, Stoyanova R. Delineation of Tumor Habitats based on Dynamic Contrast Enhanced MRI. Sci Rep 2017; 7:9746. [PMID: 28851989 PMCID: PMC5575347 DOI: 10.1038/s41598-017-09932-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 08/01/2017] [Indexed: 02/03/2023] Open
Abstract
Tumor heterogeneity can be elucidated by mapping subregions of the lesion with differential imaging characteristics, called habitats. Dynamic Contrast Enhanced (DCE-)MRI can depict the tumor microenvironments by identifying areas with variable perfusion and vascular permeability, since individual tumor habitats vary in the rate and magnitude of the contrast uptake and washout. Of particular interest is identifying areas of hypoxia, characterized by inadequate perfusion and hyper-permeable vasculature. An automatic procedure for delineation of tumor habitats from DCE-MRI was developed as a two-part process involving: (1) statistical testing in order to determine the number of the underlying habitats; and (2) an unsupervised pattern recognition technique to recover the temporal contrast patterns and locations of the associated habitats. The technique is examined on simulated data and DCE-MRI, obtained from prostate and brain pre-clinical cancer models, as well as clinical data from sarcoma and prostate cancer patients. The procedure successfully identified habitats previously associated with well-perfused, hypoxic and/or necrotic tumor compartments. Given the association of tumor hypoxia with more aggressive tumor phenotypes, the obtained in vivo information could impact management of cancer patients considerably.
Collapse
Affiliation(s)
| | - Ellen Ackerstaff
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yohann Tschudi
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Bryan Jimenez
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Warren Foltz
- STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Carl Fisher
- Department of Medical Biophysics, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Lothar Lilge
- Department of Medical Biophysics, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - HyungJoon Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Sean Carlin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Robert J Gillies
- Cancer Imaging and Metabolism, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | | | - Raphael L Yechieli
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ty Subhawong
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
34
|
Redondo PA, Pavlou M, Loizidou M, Cheema U. Elements of the niche for adult stem cell expansion. J Tissue Eng 2017; 8:2041731417725464. [PMID: 28890779 PMCID: PMC5574483 DOI: 10.1177/2041731417725464] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022] Open
Abstract
Adult stem cells are crucial for tissue homeostasis. These cells reside within exclusive locations in tissues, termed niches, which protect adult stem cell fidelity and regulate their many functions through biophysical-, biochemical- and cellular-mediated mechanisms. There is a growing understanding of how these mechanisms and their components contribute towards maintaining stem cell quiescence, self-renewal, expansion and differentiation patterns. In vitro expansion of adult stem cells is a powerful tool for understanding stem cell biology, and for tissue engineering and regenerative medicine applications. However, it is technically challenging, since adult stem cell removal from their native microenvironment has negative repercussions on their sustainability. In this review, we overview specific elements of the biomimetic niche and how recreating such elements can help in vitro propagation of adult stem cells.
Collapse
Affiliation(s)
- Patricia A Redondo
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Marina Pavlou
- Institute of Orthopaedics & Musculoskeletal Science, University College London, London, UK
| | - Marilena Loizidou
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Umber Cheema
- Institute of Orthopaedics & Musculoskeletal Science, University College London, London, UK
| |
Collapse
|
35
|
Löck S, Perrin R, Seidlitz A, Bandurska-Luque A, Zschaeck S, Zöphel K, Krause M, Steinbach J, Kotzerke J, Zips D, Troost EGC, Baumann M. Residual tumour hypoxia in head-and-neck cancer patients undergoing primary radiochemotherapy, final results of a prospective trial on repeat FMISO-PET imaging. Radiother Oncol 2017; 124:533-540. [PMID: 28843726 DOI: 10.1016/j.radonc.2017.08.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Hypoxia is a well recognised parameter of tumour resistance to radiotherapy, a number of anticancer drugs and potentially immunotherapy. In a previously published exploration cohort of 25 head and neck squamous cell carcinoma (HNSCC) patients on [18F]fluoromisonidazole positron emission tomography (FMISO-PET) we identified residual tumour hypoxia during radiochemotherapy, not before start of treatment, as the driving mechanism of hypoxia-mediated therapy resistance. Several quantitative FMISO-PET parameters were identified as potential prognostic biomarkers. Here we present the results of the prospective validation cohort, and the overall results of the study. METHODS FMISO-PET/CT images of further 25 HNSCC patients were acquired at four time-points before and during radiochemotherapy (RCHT). Peak standardised uptake value, tumour-to-background ratio, and hypoxic volume were analysed. The impact of the potential prognostic parameters on loco-regional tumour control (LRC) was validated by the concordance index (ci) using univariable and multivariable Cox models based on the exploration cohort. Log-rank tests were employed to compare the endpoint between risk groups. RESULTS The two cohorts differed significantly in several baseline parameters, e.g., tumour volume, hypoxic volume, HPV status, and intercurrent death. Validation was successful for several FMISO-PET parameters and showed the highest performance (ci=0.77-0.81) after weeks 1 and 2 of treatment. Cut-off values for the FMISO-PET parameters could be validated after week 2 of RCHT. Median values for the residual hypoxic volume, defined as the ratio of the hypoxic volume in week 2 of RCHT and at baseline, stratified patients into groups of significantly different LRC when applied to the respective other cohort. CONCLUSION Our study validates that residual tumour hypoxia during radiochemotherapy is a major driver of therapy resistance of HNSCC, and that hypoxia after the second week of treatment measured by FMISO-PET may serve as biomarker for selection of patients at high risk of loco-regional recurrence after state-of-the art radiochemotherapy.
Collapse
Affiliation(s)
- Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany
| | - Rosalind Perrin
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Center for Proton Therapy, Paul Scherrer Institute, Switzerland
| | - Annekatrin Seidlitz
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Anna Bandurska-Luque
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Sebastian Zschaeck
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Klaus Zöphel
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Germany; Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Jörg Steinbach
- National Center for Tumor Diseases, partner site Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Eberhard Karls Universität Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen, Germany
| | - Esther G C Troost
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Germany.
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Germany; Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
Yoon DW, So D, Min S, Kim J, Lee M, Khalmuratova R, Cho CH, Park JW, Shin HW. Accelerated tumor growth under intermittent hypoxia is associated with hypoxia-inducible factor-1-dependent adaptive responses to hypoxia. Oncotarget 2017; 8:61592-61603. [PMID: 28977888 PMCID: PMC5617448 DOI: 10.18632/oncotarget.18644] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/29/2017] [Indexed: 12/16/2022] Open
Abstract
Mounting evidence has revealed a causative role of intermittent hypoxia (IH) in cancer progression in mouse models of obstructive sleep apnea (OSA), but most studies have focused on the effects of IH following tumor implantation using an exposure to single IH frequency. Thus, we aimed to investigate 1) the potential effect of IH on the initial tumor growth in patients with OSA without an interaction with other mechanisms induced by IH in mice and 2) the influence of the IH frequency on tumor growth, which were tested using pre-conditioning with IH (Pre-IH) and 2 different IH frequencies, respectively. Pre-IH was achieved by alternatively maintaining melanoma cells between normoxia (10 min, 21% O2) and hypoxia (50 min, 1% O2) for 7 days (12 cycles per day) before administering them to mice. The conditions for IH-1 and IH-2 were 90 s of 12% FiO2 followed by 270s of 21% FiO2 (10 cycles/h), and 90 s of 12% FiO2 and 90 s of 21% FiO2 (20 cycles/h), respectively, for 8 h per day. Tumor growth was significantly higher in the Pre-IH group than in the normoxia group. In addition, the IH-2 group showed more accelerated tumor growth compared to the normoxia and IH-1 groups. Immunohistochemistry and gene-expression results consistently showed the up-regulation of molecules associated with HIF-1α-dependent hypoxic adaptation in tumors of the Pre-IH and IH-2 groups. Our findings reveal that IH increased tumor progression in a frequency-dependent manner, regardless of whether it was introduced before or after in vivo tumor cell implantation.
Collapse
Affiliation(s)
- Dae Wui Yoon
- Obstructive Upper Airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Daeho So
- Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Sra Min
- Obstructive Upper Airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Jiyoung Kim
- Obstructive Upper Airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Mingyu Lee
- Obstructive Upper Airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Roza Khalmuratova
- Obstructive Upper Airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Chung-Hyun Cho
- Obstructive Upper Airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jong-Wan Park
- Obstructive Upper Airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyun-Woo Shin
- Obstructive Upper Airway Research (OUaR) Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
37
|
Guo Y, Zhang P, Zhang H, Zhang P, Xu R. RNAi for contactin 2 inhibits proliferation of U87-glioma stem cells by downregulating AICD, EGFR, and HES1. Onco Targets Ther 2017; 10:791-801. [PMID: 28243115 PMCID: PMC5315346 DOI: 10.2147/ott.s113390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma is the most common form of malignant brain tumors and has a poor prognosis. Glioma stem cells (GSCs) are thought to be responsible for the aberrant proliferation and invasion. Targeting the signaling pathways that promote proliferation in GSCs is one of the strategies for glioma treatment. In this study, we found increased expression of contactin 2 (CNTN2) and amyloid β precursor protein (APP) in U87-derived GSCs (U87-GSCs). RNA interference (RNAi) for CNTN2 downregulated the expression of APP intracellular domain (AICD), which is the proteolytic product of APP. Treatment with CNTN2 RNAi inhibited the proliferation of U87-GSCs. CNTN2 RNAi decreased the expression of epidermal growth factor receptor and HES1, which are potential targets of AICD. In summary, inhibition of the CNTN2/APP signaling pathway may repress the proliferation in U87-GSCs via downregulating the expression of HES1 and epidermal growth factor receptor. CNTN2/APP/AICD signaling pathway plays an important role in U87 glial tumorigenesis. Further studies are warranted to elucidate the role of these signaling pathways in other sources of GSCs. Depending on their role in proliferation in other sources of GSCs, members of the CNTN2/APP/AICD signaling pathway may provide novel targets for the development of therapy for glioblastomas.
Collapse
Affiliation(s)
| | - Peidong Zhang
- Department of Cardiovascular Medicine, Zhujiang Hospital; Second Clinical Medical College, Southern Medical University, Guangzhou
| | - Hongtian Zhang
- Department of Neurosurgery, Affiliated Bayi Brain Hospital, The Military General Hospital of Beijing PLA, The Bayi Clinical Medical Institute of Southern Medical University, Beijing, People's Republic of China
| | - Peng Zhang
- Department of Neurosurgery, Affiliated Bayi Brain Hospital, The Military General Hospital of Beijing PLA, The Bayi Clinical Medical Institute of Southern Medical University, Beijing, People's Republic of China
| | - Ruxiang Xu
- Department of Neurosurgery, Affiliated Bayi Brain Hospital, The Military General Hospital of Beijing PLA, The Bayi Clinical Medical Institute of Southern Medical University, Beijing, People's Republic of China
| |
Collapse
|
38
|
Ramamonjisoa N, Ackerstaff E. Characterization of the Tumor Microenvironment and Tumor-Stroma Interaction by Non-invasive Preclinical Imaging. Front Oncol 2017; 7:3. [PMID: 28197395 PMCID: PMC5281579 DOI: 10.3389/fonc.2017.00003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Tumors are often characterized by hypoxia, vascular abnormalities, low extracellular pH, increased interstitial fluid pressure, altered choline-phospholipid metabolism, and aerobic glycolysis (Warburg effect). The impact of these tumor characteristics has been investigated extensively in the context of tumor development, progression, and treatment response, resulting in a number of non-invasive imaging biomarkers. More recent evidence suggests that cancer cells undergo metabolic reprograming, beyond aerobic glycolysis, in the course of tumor development and progression. The resulting altered metabolic content in tumors has the ability to affect cell signaling and block cellular differentiation. Additional emerging evidence reveals that the interaction between tumor and stroma cells can alter tumor metabolism (leading to metabolic reprograming) as well as tumor growth and vascular features. This review will summarize previous and current preclinical, non-invasive, multimodal imaging efforts to characterize the tumor microenvironment, including its stromal components and understand tumor-stroma interaction in cancer development, progression, and treatment response.
Collapse
Affiliation(s)
- Nirilanto Ramamonjisoa
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ellen Ackerstaff
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
39
|
Li Q, Lin Q, Yun Z. Hypoxia-activated cytotoxicity of benznidazole against clonogenic tumor cells. Cancer Biol Ther 2016; 17:1266-1273. [PMID: 27786593 DOI: 10.1080/15384047.2016.1250988] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Solid tumors contain numerous regions with insufficient oxygen concentrations, a condition termed hypoxia. Tumor hypoxia is significantly associated with metastasis, refractory to conventional cancer therapies, and poor patient survival. Therefore, eradication of hypoxic tumor cells will likely have significant impact on the overall progression-free patient survival. This article reports a new discovery that Benznidazole, a bioreductive drug currently used to treat Chagas disease caused by the parasitic protozoan Trypanosoma cruzi, is activated by hypoxia and can kill clonogenic tumor cells especially those under severe hypoxic conditions (≤0.1 % O2). This type of hypoxia selectivity is important in that severely hypoxic tumor microenvironment is where tumor cells exhibit the strongest resistance to therapy. Mechanistically, activation of Benznidazole coincides with the stabilization of the Hypoxia-Inducible Factor 1α (HIF-1α), suggesting that Benznidazole is activated after tumor cells have entered into a fully hypoxic state. Under such hypoxic conditions, Benznidazole induces the formation of 53BP1 foci, a hallmark of DNA double-stranded breaks that can cause clonogenic inhibition or cell death. These results demonstrate that Benznidazole is a hypoxia-activated cytotoxin with the potential to specifically eliminate hypoxic tumor cells.
Collapse
Affiliation(s)
- Quhuan Li
- a Department of Therapeutic Radiology , Yale School of Medicine , New Haven , CT , USA
| | - Qun Lin
- a Department of Therapeutic Radiology , Yale School of Medicine , New Haven , CT , USA
| | - Zhong Yun
- a Department of Therapeutic Radiology , Yale School of Medicine , New Haven , CT , USA
| |
Collapse
|
40
|
Alite F, Stang K, Balasubramanian N, Adams W, Shaikh MP, Small C, Sethi A, Nagda S, Emami B, Harkenrider MM. Local control dependence on consecutive vs. nonconsecutive fractionation in lung stereotactic body radiation therapy. Radiother Oncol 2016; 121:9-14. [DOI: 10.1016/j.radonc.2016.07.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 07/26/2016] [Accepted: 07/31/2016] [Indexed: 11/26/2022]
|
41
|
Kalavska K, Chovanec M, Zatovicova M, Takacova M, Gronesova P, Svetlovska D, Baratova M, Miskovska V, Obertova J, Palacka P, Rajec J, Sycova-Mila Z, Cierna Z, Kajo K, Spanik S, Babal P, Mardiak J, Pastorekova S, Mego M. Prognostic value of serum carbonic anhydrase IX in testicular germ cell tumor patients. Oncol Lett 2016; 12:2590-2598. [PMID: 27698832 PMCID: PMC5038507 DOI: 10.3892/ol.2016.5010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/27/2016] [Indexed: 11/19/2022] Open
Abstract
Despite the fact that testicular germ cell tumors (TGCTs) are one of the most chemosensitive solid tumors, a small proportion of patients fail to be cured following cisplatin-based first line chemotherapy. Upregulation of carbonic anhydrase IX (CA IX) in various solid tumors is associated with poor outcome. The current prospective study investigated the prognostic value of serum CA IX level in TGCTs. In total, 83 patients (16 non-metastatic following orchiectomy with no evidence of disease, 57 metastatic chemotherapy-naïve and 10 metastatic relapsed chemotherapy-pretreated) starting adjuvant and/or new line of chemotherapy and 35 healthy controls were enrolled in the study. Serum CA IX values were determined using an enzyme-linked immunosorbent assay, and intratumoral CA IX was analyzed by immunohistochemistry. Metastatic chemotherapy-naïve patients had significantly higher mean CA IX serum levels than healthy controls (490.6 vs. 249.6 pg/ml, P=0.005), while there was no difference in serum CA IX levels in non-metastatic or relapsed TGCT patients compared with healthy controls. There was no significant difference in the mean serum CA IX levels between different groups of patients and between the first and second cycle of chemotherapy, nor association with patients/tumor characteristics. Serum CA IX was not prognostic for progression-free survival [hazard ratio (HR)=0.81, P=0.730] or overall survival (HR=0.64, P=0.480). However, there was a significant association between intratumoral CA IX expression and serum CA IX concentration (rho=0.51, P=0.040). These results suggest that serum CA IX level correlates with tumor CA IX expression in TGCT patients, but fails to exhibit either a prognostic value or an association with patients/tumor characteristics.
Collapse
Affiliation(s)
- Katarina Kalavska
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia; Department of Oncology, National Cancer Institute, 833 10 Bratislava, Slovakia; Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, 811 04 Bratislava, Slovakia
| | - Michal Chovanec
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia; Department of Oncology, National Cancer Institute, 833 10 Bratislava, Slovakia; Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, 833 10 Bratislava, Slovakia
| | - Miriam Zatovicova
- Department of Molecular Medicine, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 811 04 Bratislava, Slovakia
| | - Martina Takacova
- Department of Molecular Medicine, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 811 04 Bratislava, Slovakia
| | - Paulina Gronesova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, 811 04 Bratislava, Slovakia
| | - Daniela Svetlovska
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia; Department of Oncology, National Cancer Institute, 833 10 Bratislava, Slovakia
| | - Magdalena Baratova
- Department of Molecular Medicine, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 811 04 Bratislava, Slovakia
| | - Vera Miskovska
- First Department of Oncology, Faculty of Medicine, Comenius University and St. Elizabeth Cancer Institute, 81 250 Bratislava, Slovakia; Department of Oncology, St. Elizabeth Cancer Institute, 81 250 Bratislava, Slovakia
| | - Jana Obertova
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia
| | - Patrik Palacka
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia
| | - Jan Rajec
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia
| | - Zuzana Sycova-Mila
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia
| | - Zuzana Cierna
- Department of Pathology, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia
| | - Karol Kajo
- Department of Pathology, Slovak Medical University, 833 03 Bratislava, Slovakia
| | - Stanislav Spanik
- First Department of Oncology, Faculty of Medicine, Comenius University and St. Elizabeth Cancer Institute, 81 250 Bratislava, Slovakia; Department of Oncology, St. Elizabeth Cancer Institute, 81 250 Bratislava, Slovakia
| | - Pavel Babal
- Department of Pathology, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia
| | - Jozef Mardiak
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia; Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, 833 10 Bratislava, Slovakia
| | - Silvia Pastorekova
- Department of Molecular Medicine, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 811 04 Bratislava, Slovakia
| | - Michal Mego
- Translational Research Unit, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia; Department of Oncology, National Cancer Institute, 833 10 Bratislava, Slovakia; Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, 833 10 Bratislava, Slovakia
| |
Collapse
|
42
|
Matschke J, Riffkin H, Klein D, Handrick R, Lüdemann L, Metzen E, Shlomi T, Stuschke M, Jendrossek V. Targeted Inhibition of Glutamine-Dependent Glutathione Metabolism Overcomes Death Resistance Induced by Chronic Cycling Hypoxia. Antioxid Redox Signal 2016; 25:89-107. [PMID: 27021152 DOI: 10.1089/ars.2015.6589] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AIMS Tumor hypoxia is a major biological factor causing poor patient outcome. Evidence is increasing that improved protection against reactive oxygen species (ROS) participates in therapy resistance of chronically hypoxic cancer cells. We aimed at characterizing the relevance of improved ROS defense for radiation resistance of cancer cells with tolerance to cycling anoxia/re-oxygenation stress ("anoxia-tolerant") and at designing rational treatment strategies for overcoming the resulting therapy resistance by targeting the underlying mechanisms identified in an in vitro model. RESULTS We demonstrate that chronic exposure of NCH-H460 lung adenocarcinoma, DU145 prostate cancer, and T98G glioblastoma cells to cycling anoxia/re-oxygenation stress induced upregulation of the aspartate-aminotransferase glutamic-oxaloacetic transaminase (GOT1), particularly in RAS-driven anoxia-tolerant NCI-H460 cells. Altered glutamine utilization of the anoxia-tolerant cancer cells contributed to the observed decrease in cellular ROS levels, the increase in cellular glutathione levels, and improved cell survival on ROS-inducing treatments, including exposure to ionizing radiation. Importantly, targeting glutamine-dependent antioxidant capacity or glutathione metabolism allowed us to hit anoxia-tolerant cancer cells and to overcome their increased resistance to radiation-induced cell death. Targeting glutathione metabolism by Piperlongumine also improved the radiation response of anoxia-tolerant NCI-H460 cells in vivo. INNOVATION Improved antioxidant capacity downstream of up-regulated GOT1-expression is a characteristic of anoxia-tolerant cancer cells and is predictive for a specific vulnerability to inhibition of glutamine utilization or glutathione metabolism, respectively. CONCLUSION Unraveling the molecular alterations underlying improved ROS defense of anoxia-tolerant cancer cells allows the design of rational strategies for overcoming radiation resistance caused by tumor cell heterogeneity in hypoxic tumors. Antioxid. Redox Signal. 25, 89-107.
Collapse
Affiliation(s)
- Johann Matschke
- 1 Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen, Germany
| | - Helena Riffkin
- 1 Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen, Germany
| | - Diana Klein
- 1 Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen, Germany
| | - René Handrick
- 2 Institute of Applied Biotechnology (IAB), University of Applied Sciences , Biberach, Germany
| | - Lutz Lüdemann
- 3 Department of Radiotherapy, University Hospital Essen , Essen, Germany
| | - Eric Metzen
- 4 Institute of Physiology, University Hospital Essen , Essen, Germany
| | - Tomer Shlomi
- 5 Department of Computer Science and Biology & Lokey Center for Life Science and Engineering, Technion, Haifa, Israel
| | - Martin Stuschke
- 3 Department of Radiotherapy, University Hospital Essen , Essen, Germany
| | - Verena Jendrossek
- 1 Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen, Germany
| |
Collapse
|
43
|
Michiels C, Tellier C, Feron O. Cycling hypoxia: A key feature of the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2016; 1866:76-86. [PMID: 27343712 DOI: 10.1016/j.bbcan.2016.06.004] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 06/18/2016] [Accepted: 06/21/2016] [Indexed: 01/06/2023]
Abstract
A compelling body of evidence indicates that most human solid tumors contain hypoxic areas. Hypoxia is the consequence not only of the chaotic proliferation of cancer cells that places them at distance from the nearest capillary but also of the abnormal structure of the new vasculature network resulting in transient blood flow. Hence two types of hypoxia are observed in tumors: chronic and cycling (intermittent) hypoxia. Most of the current work aims at understanding the role of chronic hypoxia in tumor growth, response to treatment and metastasis. Only recently, cycling hypoxia, with spatial and temporal fluctuations in oxygen levels, has emerged as another key feature of the tumor environment that triggers different responses in comparison to chronic hypoxia. Either type of hypoxia is associated with distinct effects not only in cancer cells but also in stromal cells. In particular, cycling hypoxia has been demonstrated to favor, to a higher extent than chronic hypoxia, angiogenesis, resistance to anti-cancer treatments, intratumoral inflammation and tumor metastasis. These review details these effects as well as the signaling pathway it triggers to switch on specific transcriptomic programs. Understanding the signaling pathways through which cycling hypoxia induces these processes that support the development of an aggressive cancer could convey to the emergence of promising new cancer treatments.
Collapse
Affiliation(s)
- Carine Michiels
- URBC-NARILIS, University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium.
| | - Céline Tellier
- URBC-NARILIS, University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, 53 Avenue Mounier, B1.53.09, B-1200 Brussels, Belgium.
| |
Collapse
|
44
|
Mrozek S, Luzi A, Gonzalez L, Kerhuel L, Fourcade O, Geeraerts T. Cerebral and extracerebral vulnerability to hypoxic insults after diffuse traumatic brain injury in rats. Brain Res 2016; 1646:334-341. [PMID: 27302136 DOI: 10.1016/j.brainres.2016.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/03/2016] [Accepted: 06/04/2016] [Indexed: 11/30/2022]
Abstract
The post-traumatic brain vulnerability suggests that after traumatic brain injury (TBI), the brain may be more susceptible to posttraumatic hypoxic insults. This concept could be extended to 'peripheral' organs, as non-neurologic organ failure is common after TBI. This study aims to characterize and quantify cerebral and extracerebral tissue hypoxia with pimonidazole resulting from a standardized hypoxia-hypotension (HH) phase occurring after a diffuse experimental TBI in rats. Rats were allocated to Sham (n=5), TBI (n=7), HH (n=7) and TBI+HH (n=7) groups. Then, pimonidazole was injected and brain, liver, heart and kidneys were analysed. In the cerebral cortex, post-treatment hypoxia was higher in TBI+HH group than Sham group (p=0.003), HH group (p=0.003) and TBI group (p=0.002). Large trends in thalamus, hippocampus and striatum for the TBI+HH group compared to the other groups were observed. For the heart and liver, the 4 groups were comparable. For the kidneys, post-treatment hypoxia was higher in the TBI group compared to the Sham and HH groups, but not more than TBI+HH group. This study reveals that a posttraumatic hypoxic insult occurring after a severe TBI has major hypoxic consequences on brain structures. However, TBI by itself appears to induce renal hypoxia that is not enhanced by posttraumatic hypoxic insult.
Collapse
Affiliation(s)
- Ségolène Mrozek
- Equipe d'accueil' Modélisation de l'aggression tissulaire et nociceptive', University Toulouse 3 Paul Sabatier, Toulouse, France; Departement of Anesthesiology and Critical Care, University Hospital of Toulouse, Toulouse, France.
| | - Aymeric Luzi
- Equipe d'accueil' Modélisation de l'aggression tissulaire et nociceptive', University Toulouse 3 Paul Sabatier, Toulouse, France; Departement of Anesthesiology and Critical Care, University Hospital of Toulouse, Toulouse, France.
| | - Leslie Gonzalez
- Equipe d'accueil' Modélisation de l'aggression tissulaire et nociceptive', University Toulouse 3 Paul Sabatier, Toulouse, France; Departement of Anesthesiology and Critical Care, University Hospital of Toulouse, Toulouse, France.
| | - Lionel Kerhuel
- Equipe d'accueil' Modélisation de l'aggression tissulaire et nociceptive', University Toulouse 3 Paul Sabatier, Toulouse, France; Departement of Anesthesiology and Critical Care, University Hospital of Toulouse, Toulouse, France.
| | - Olivier Fourcade
- Equipe d'accueil' Modélisation de l'aggression tissulaire et nociceptive', University Toulouse 3 Paul Sabatier, Toulouse, France; Departement of Anesthesiology and Critical Care, University Hospital of Toulouse, Toulouse, France.
| | - Thomas Geeraerts
- Equipe d'accueil' Modélisation de l'aggression tissulaire et nociceptive', University Toulouse 3 Paul Sabatier, Toulouse, France; Departement of Anesthesiology and Critical Care, University Hospital of Toulouse, Toulouse, France.
| |
Collapse
|
45
|
White DA, Zhang Z, Li L, Gerberich J, Stojadinovic S, Peschke P, Mason RP. Developing oxygen-enhanced magnetic resonance imaging as a prognostic biomarker of radiation response. Cancer Lett 2016; 380:69-77. [PMID: 27267808 DOI: 10.1016/j.canlet.2016.06.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 11/19/2022]
Abstract
Oxygen-Enhanced Magnetic Resonance Imaging (OE-MRI) techniques were evaluated as potential non-invasive predictive biomarkers of radiation response. Semi quantitative blood-oxygen level dependent (BOLD) and tissue oxygen level dependent (TOLD) contrast, and quantitative responses of relaxation rates (ΔR1 and ΔR2*) to an oxygen breathing challenge during hypofractionated radiotherapy were applied. OE-MRI was performed on subcutaneous Dunning R3327-AT1 rat prostate tumors (n=25) at 4.7 T prior to each irradiation (2F × 15 Gy) to the gross tumor volume. Response to radiation, while inhaling air or oxygen, was assessed by tumor growth delay measured up to four times the initial irradiated tumor volume (VQT). Radiation-induced hypoxia changes were confirmed using a double hypoxia marker assay. Inhaling oxygen during hypofractionated radiotherapy significantly improved radiation response. A correlation was observed between the difference in the 2nd and 1st ΔR1 (ΔΔR1) and VQT for air breathing rats. The TOLD response before the 2nd fraction showed a moderate correlation with VQT for oxygen breathing rats. The correlations indicate useful prognostic factors to predict tumor response to hypofractionation and could readily be applied for patient stratification and personalized radiotherapy treatment planning.
Collapse
Affiliation(s)
- Derek A White
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas 75390, USA; Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, USA
| | - Zhang Zhang
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Li Li
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Jeni Gerberich
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Strahinja Stojadinovic
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | - Ralph P Mason
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas 75390, USA.
| |
Collapse
|
46
|
Ikeda Y, Hisano H, Nishikawa Y, Nagasaki Y. Targeting and Treatment of Tumor Hypoxia by Newly Designed Prodrug Possessing High Permeability in Solid Tumors. Mol Pharm 2016; 13:2283-9. [PMID: 27187083 DOI: 10.1021/acs.molpharmaceut.6b00011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumor hypoxia, which is associated with poor prognosis in cancer, is known to lead to resistance to radiotherapy and anticancer chemotherapy. Impaired drug penetration in hypoxic regions has been recognized as an essential barrier to drug development in solid tumors. Here, we propose novel hypoxia-activated prodrugs, which drastically improved the penetration property of commonly used anticancer drugs in the hypoxic region. In this design, conventional anticancer drugs were modified with 2-nitroimidazole derivatives. The most important point of this study was that the prodrug designed formed a 6-membered cyclic structure to allow liberation of the active drug in the hypoxic region. This design markedly increased the selectivity of the hypoxia-targeted prodrug, resulting in significant reduction of adverse effects in the normoxic region. In vitro studies confirmed the selective activation under hypoxic conditions. In vivo studies showed drastic reduction of adverse effects associated with conventional anticancer drugs and improvement of the survival rate of mice. Immunofluorescence analyses confirmed that the designed prodrug had a tendency to localize at the hypoxic region, in contrast to conventional anticancer drugs, which localize only at the normoxic region.
Collapse
Affiliation(s)
- Yutaka Ikeda
- Department of Materials Science, Master's School of Medical Sciences, University of Tsukuba , Tennoudai 1-1-1, Tsukuba 305-8573, Japan
| | - Hikaru Hisano
- Department of Materials Science, Master's School of Medical Sciences, University of Tsukuba , Tennoudai 1-1-1, Tsukuba 305-8573, Japan
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University , Midorigaoka Higashi-2-jyo 1-1-1, Asahikawa, Hokkaido 078-8510, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Master's School of Medical Sciences, University of Tsukuba , Tennoudai 1-1-1, Tsukuba 305-8573, Japan.,Master's School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tennodai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan.,Satellite Laboratory, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), University of Tsukuba , Tennoudai 1-1-1, Tsukuba 305-8573, Japan
| |
Collapse
|
47
|
Boström PJ, Thoms J, Sykes J, Ahmed O, Evans A, van Rhijn BWG, Mirtti T, Stakhovskyi O, Laato M, Margel D, Pintilie M, Kuk C, Milosevic M, Zlotta AR, Bristow RG. Hypoxia Marker GLUT-1 (Glucose Transporter 1) is an Independent Prognostic Factor for Survival in Bladder Cancer Patients Treated with Radical Cystectomy. Bladder Cancer 2016; 2:101-109. [PMID: 27376131 PMCID: PMC4927886 DOI: 10.3233/blc-150033] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Tumour hypoxia, which is frequent in many cancer types, is associated with treatment resistance and poor prognosis. The role of hypoxia in surgically treated bladder cancer (BC) is not well described. We studied the role of hypoxia in two independent series of urothelial bladder cancers treated with radical cystectomy. METHODS 279 patients from the University Hospital Network (UHN), Toronto, Canada, and Turku University, Finland were studied. Hypoxia biomarkers (HIF1-α, CAIX, GLUT-1) and proliferation marker Ki-67 were analyzed with immunohistochemistry using defined tissue microarrays. Kaplan-Meier methods and Cox proportional hazards regression models were used to investigate prognostic role of the factors. RESULTS In univariate analyses, strong GLUT-1 positivity and a high Ki-67 index were associated with poor survival. In multivariate model containing clinical prognostic variables, GLUT-1 was an independent prognostic factor associated with worse disease-specific survival (HR 2.9, 95% CI 0.7-12.6, Wald p = 0.15 in the Toronto cohort and HR 3.2, 95% CI 1.3-7.5, Wald p = 0.0085 in the Turku cohort). CONCLUSION GLUT-1 is frequently upregulated and is an independent prognostic factor in surgically treated bladder cancer. Further studies are needed to evaluate the potential role of hypoxia-based and targeted therapies in hypoxic bladder tumours.
Collapse
Affiliation(s)
- P J Boström
- Department of Surgical Oncology, University of Toronto and Princess Margaret Hospital (University Health Network), Toronto, ON, Canada; Department of Urology, Turku University Hospital, Turku, Finland
| | - J Thoms
- Department of Radiation Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - J Sykes
- Department of Biostatistics, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - O Ahmed
- Department of Radiation Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - A Evans
- Department of Pathology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - B W G van Rhijn
- Department of Surgical Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - T Mirtti
- Department of Pathology, Helsinki University Hospital, Helsinki, Finland; Institute for Molecular Medicine (FIMM), Helsinki, Finland
| | - O Stakhovskyi
- Department of Surgical Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - M Laato
- Department of Urology, Turku University Hospital , Turku, Finland
| | - D Margel
- Department of Surgical Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - M Pintilie
- Department of Biostatistics, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - C Kuk
- Department of Surgical Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - M Milosevic
- Department of Radiation Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - A R Zlotta
- Department of Surgical Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| | - R G Bristow
- Department of Radiation Oncology, University of Toronto and Princess Margaret Hospital (University Health Network) , Toronto, ON, Canada
| |
Collapse
|
48
|
Tumor Hypoxia: Causative Mechanisms, Microregional Heterogeneities, and the Role of Tissue-Based Hypoxia Markers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 923:77-86. [DOI: 10.1007/978-3-319-38810-6_11] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Abstract
There is an important and strong, but complex influence of the tumor microenvironment on tumor cells' phenotype, aggressiveness, and treatment sensitivity. One of the most frequent and best-studied aspects of the tumor microenvironment is hypoxia. Low oxygen tension often occurs in tumor cells by several mechanisms, for example, poor angiogenesis and increased oxygen consumption. Hypoxia is a heterogeneous concept with oxygen tensions ranging from <0.01% (anoxia) to 5%, and can be chronic, acute, or cycling, all with differential effects on tumor cells. Quantification of tumor hypoxia can be performed directly or indirectly, and with exogenous or endogenous markers. Tumor cells launch different intracellular signaling pathways to survive hypoxia, such as hypoxia-inducible factor 1-mediated gene expression, the unfolded protein response, and AKT-mammalian target of rapamycin signaling. These pathways induce aggressive, metastatic, and treatment-insensitive tumors and are considered potential targets for (additive) therapy. Hypoxia leads to important, yet currently not well-understood changes in microRNA expression, epigenetics, and metabolism. Further, treatment-insensitive tumors arise through hypoxia-induced Darwinian selection of apoptosis-deficient, p53-mutated tumor cells. In conclusion, hypoxia has profound and largely still poorly understood effects on tumor cells with a major effect on the tumor's biology.
Collapse
Affiliation(s)
- Paul N Span
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
50
|
Jeong SK, Kim JS, Lee CG, Park YS, Kim SD, Yoon SO, Han DH, Lee KY, Jeong MH, Jo WS. Tumor associated macrophages provide the survival resistance of tumor cells to hypoxic microenvironmental condition through IL-6 receptor-mediated signals. Immunobiology 2015; 222:55-65. [PMID: 26705936 DOI: 10.1016/j.imbio.2015.11.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 10/23/2015] [Accepted: 11/22/2015] [Indexed: 01/05/2023]
Abstract
Hypoxia and infiltration of tumor-associated macrophages (TAM) are intrinsic features of the tumor microenvironment. Tumor cells that remain viable in hypoxic conditions often possess an increased survival potential and tend to grow aggressively. TAM also respond to a variety of signals in the hypoxic tumor microenvironment and express a more M2-like phenotype. In this study, the established mouse tumor tissues showed a dense infiltration of CD206+ macrophages at the junctions between the normoxic and hypoxic regions and an increased IL-6 receptor (IL-6R) expression of tumor cells in the areas of CD206+ TAM accumulation, which indicates a role of M2 phenotype TAM in survival adaptation of tumor cells preparing for an impending hypoxic injury before changes in oxygen availability. Cocultured mouse FM3A or human MCF-7 tumor cells with tumor infiltrating macrophages isolated from mouse tumor tissues and M2-polarized macrophages generated from human THP-1 cells, respectively, showed significantly decreased rate of cell death in cultures exposed to hypoxia. The acquisition of survival resistance was attributed to increased IL-6 production by M2 TAM and increased expression of IL-6R in tumor cells in the coculture system. MCF-7 cells cocultured with M2 TAM showed activated JAK1/STAT3 and Raf/MEK/JNK pathways contributing to tyrosine and serine phophorylation of STAT3, respectively. However, only tyrosine phosphorylated STAT3 was detected in the nucleus, which induced upregulation of Bcl-2 and downregulation of Bax and Bak. Finally, knockdown of IL-6R by small interfering RNA significantly counteracted coculture-induced signals and completely abolished the survival resistance to hypoxic injury. Thus, we present evidence for the role of M2 phenotype TAM in IL-6 receptor-mediated signals, particularly tyrosine phosphorylation of STAT3, responsible for the prosurvival adaptation of tumor cells to hypoxia.
Collapse
Affiliation(s)
- Soo Kyung Jeong
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea; Department of Microbiology, Dong-A University College of Medicine, Daeshingongwon-gil 32, Seo-gu, Busan 602-714, Republic of Korea
| | - Joong Sun Kim
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea
| | - Chang Geun Lee
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea
| | - You-Soo Park
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea
| | - Sung Dae Kim
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea
| | - Sun Ok Yoon
- Transplantation Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 110-799, Republic of Korea
| | - Dong Hoon Han
- Department of Microbiology, Dong-A University College of Medicine, Daeshingongwon-gil 32, Seo-gu, Busan 602-714, Republic of Korea
| | - Kyu Yeol Lee
- Department of Microbiology, Dong-A University College of Medicine, Daeshingongwon-gil 32, Seo-gu, Busan 602-714, Republic of Korea
| | - Min Ho Jeong
- Department of Microbiology, Dong-A University College of Medicine, Daeshingongwon-gil 32, Seo-gu, Busan 602-714, Republic of Korea.
| | - Wol Soon Jo
- Department of Research Center, Dong Nam Institute of Radiological and Medical Sciences, Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan 619-953, Republic of Korea.
| |
Collapse
|