51
|
Li J, Yang P, Chen F, Tan Y, Huang C, Shen H, Peng C, Feng Y, Sun Y. Hypoxic colorectal cancer-derived extracellular vesicles deliver microRNA-361-3p to facilitate cell proliferation by targeting TRAF3 via the noncanonical NF-κB pathways. Clin Transl Med 2021; 11:e349. [PMID: 33784010 PMCID: PMC7967919 DOI: 10.1002/ctm2.349] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Hypoxic tumour microenvironment (TME) is a key regulator in cancer progression. However, the communications between hypoxic cells and other components in TME during colorectal cancer (CRC) progression via extracellular vesicles (EVs) remain unclear. METHODS High-throughput sequencing was employed to detect aberrantly expressed microRNAs (miRNAs) in hypoxic EVs. Quantitative real-time PCR was used to confirm and screen preliminarily candidate miRNAs. The effects of EVs derived from hypoxia (<1% O2 ) and miR-361-3p on CRC growth were assessed using CCK-8 assays, colony formation assays, EdU assays, flow cytometric assays and mouse xenograft. Then, the specific mechanisms of miR-361-3p were investigated by RNA immunoprecipitation, luciferase reporter assay, Western blot, chromatin immunoprecipitation, immunohistochemistry and rescue experiments. RESULTS The level of miR-361-3p expression was remarkably elevated in hypoxic EVs and can be transferred to CRC cells. Functional experiments exhibited that hypoxic EVs facilitated cell growth and suppressed cell apoptosis by transferring miR-361-3p of CRC. Hypoxia-inducible factor-1α induced the elevation of miR-361-3p levels in hypoxic EVs. Upregulated miR-361-3p in CRC inhibited cell apoptosis and facilitated cell growth by directly targeting TNF receptor-associated factor 3, which consequently activated the noncanonical NF-κB pathway. Moreover, the high expression of circulating exosomal miR-361-3p was correlated to worse prognosis of CRC patients. CONCLUSIONS Altogether, the abnormality of exosomal miR-361-3p derived from hypoxia acts vital roles in the regulation of CRC growth and apoptosis and can be an emerging prognostic biomarker and a therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Jie Li
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Peng Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Fangyu Chen
- Department of Radiation OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yuqian Tan
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Changzhi Huang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Hengyang Shen
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Chaofan Peng
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yifei Feng
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yueming Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
52
|
Weng M, Feng Y, He Y, Yang W, Li J, Zhu Y, Wang T, Wang C, Zhang X, Qiao Y, Li Q, Zhao L, Gao S, Zhang L, Wu Y, Zhao R, Wang G, Li Z, Jin X, Zheng T, Li X. Hypoxia-Induced LIN28A mRNA Promotes the Metastasis of Colon Cancer in a Protein-Coding-Independent Manner. Front Cell Dev Biol 2021; 9:642930. [PMID: 33665193 PMCID: PMC7921329 DOI: 10.3389/fcell.2021.642930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
The hypoxic microenvironment is beneficial to the metastasis but not to the proliferation of cancer cells. However, the mechanisms regarding to hypoxia differentially regulating cancer metastasis and proliferation are largely unknown. In this study, we revealed that hypoxia induced the expression of LIN28A at mRNA level but segregated LIN28A mRNAs in the P-bodies and thus inhibits the production of LIN28A protein. This unexpected finding suggests that there may be non-coding role for LIN28A mRNA in the progression of colon cancer. We further showed that the non-coding LIN28A mRNA promotes the metastasis but not proliferation of colon cancer cells in vitro and in vivo. Mechanistically, we revealed that methionyl aminopeptidase 2 (METAP2) is one of the up-regulated metastasis regulators upon over-expression of non-coding LIN28A identified by mass spectrum, and confirmed that it is non-coding LIN28A mRNA instead of LIN28A protein promotes the expression of METAP2. Moreover, we demonstrated that knockdown of DICER abolished the promotional effects of non-coding LIN28A on the metastasis and METAP2 expression. Conclusively, we showed that hypoxia induces the production of LIN28A mRNAs but segregated them into the P-bodies together with miRNAs targeting both LIN28A and METAP2, and then promotes the metastasis by positively regulating the expression of METAP2. This study uncovered a distinctive role of hypoxia in manipulating the metastasis and proliferation by differently regulating the expression of LIN28A at mRNA and protein level.
Collapse
Affiliation(s)
- Mingjiao Weng
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yukuan Feng
- Key Laboratory of Heilongjiang Province for Cancer Prevention and Control, School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Yan He
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Weiwei Yang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Jing Li
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yuanyuan Zhu
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Tianzhen Wang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Chuhan Wang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Xiao Zhang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yu Qiao
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Qi Li
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Lingyu Zhao
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Shuangshu Gao
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Lei Zhang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yiqi Wu
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Ran Zhao
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Guangyu Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhiwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaoming Jin
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, China
| |
Collapse
|
53
|
Farag MM, Abd El Malak NS, Yehia SA, Ahmed MA. Hyaluronic Acid Conjugated Metformin-Phospholipid Sonocomplex: A Biphasic Complexation Approach to Correct Hypoxic Tumour Microenvironment. Int J Nanomedicine 2021; 16:1005-1019. [PMID: 33603365 PMCID: PMC7885809 DOI: 10.2147/ijn.s297634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/14/2021] [Indexed: 01/09/2023] Open
Abstract
Purpose Development of hyaluronic acid conjugated metformin-phospholipid sonocomplexes (HA-MPS), a biphasic complexation product compiled for enhancing both the lipophilicity and targeting potential of Metformin (MET) to CD44 receptors on pancreatic cancer. Methods MET was chemically conjugated to hyaluronic acid (HA) via amide coupling reaction. Then, the HA conjugated MET was physically conjugated to Lipoid™S100 via ultrasound irradiation. A combined D-optimal design was implemented to statistically optimize formulation variables. The HA-MPS were characterized through solubility studies, partition coefficient, drug content uniformity, particle size and zeta potential. The optimized HA-MPS was tested via proton nuclear magnetic resonance, infrared spectroscopy to elucidate the nature of physicochemical interactions in the complex which was further scrutinized on molecular level via molecular docking and dynamic simulation. Results The solubility and partition studies showed a lipophilicity enhancement up to 67 folds as they adopted inverted micelles configuration based on the packing parameter hypothesis. The optimized HA-MPS showed 11.5 folds lower IC50, extra 25% reduction in oxygen consumption rate, better reduction in hypoxia-inducible factor and reactive oxygen species in MiaPaCa-2 cells. Conclusion These results proved better internalization of MET which was reflected by abolishing hypoxic tumour microenvironment, a mainstay toward a normoxic and less resistant pancreatic cancer.
Collapse
Affiliation(s)
- Michael M Farag
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nevine S Abd El Malak
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt.,School of Pharmacy, New Giza University, Giza, Egypt
| | - Soad A Yehia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohammed A Ahmed
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt.,School of Pharmacy, New Giza University, Giza, Egypt
| |
Collapse
|
54
|
Popa LG, Lutuc RS, Mihai MM, Ahmed Salem I, Negoiţă SI, Giurcăneanu C, Fica SV. Hereditary leiomyomatosis and renal cell cancer syndrome - case report and review of the literature. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 61:569-575. [PMID: 33544811 PMCID: PMC7864305 DOI: 10.47162/rjme.61.2.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Hereditary leiomyomatosis and renal cell cancer syndrome (HLRCC) is an exceptionally rare autosomal dominant condition caused by a germline heterozygous mutation of the fumarate hydratase gene. It manifests as multiple piloleiomyomas, associated with numerous, early-onset uterine leiomyomas in female patients, as well as a highly increased risk of renal cell carcinoma (RCC), most often type 2 papillary RCC. HLRCC has been described in association with adrenal cortical hyperplasia, pheochromocytoma, adrenal cortical carcinoma, and other solid tumors, but the exact relationship between these disorders has not yet been clarified. We present a case of HLRCC associated with bilateral adrenal cortical hyperplasia and discuss the pathogenesis, clinical and paraclinical features of HLRCC, as well as the adequate management of these patients.
Collapse
Affiliation(s)
- Liliana Gabriela Popa
- Department of Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania;
| | | | | | | | | | | | | |
Collapse
|
55
|
Bernauer C, Man YKS, Chisholm JC, Lepicard EY, Robinson SP, Shipley JM. Hypoxia and its therapeutic possibilities in paediatric cancers. Br J Cancer 2021; 124:539-551. [PMID: 33106581 PMCID: PMC7851391 DOI: 10.1038/s41416-020-01107-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/20/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
In tumours, hypoxia-a condition in which the demand for oxygen is higher than its availability-is well known to be associated with reduced sensitivity to radiotherapy and chemotherapy, and with immunosuppression. The consequences of hypoxia on tumour biology and patient outcomes have therefore led to the investigation of strategies that can alleviate hypoxia in cancer cells, with the aim of sensitising cells to treatments. An alternative therapeutic approach involves the design of prodrugs that are activated by hypoxic cells. Increasing evidence indicates that hypoxia is not just clinically significant in adult cancers but also in paediatric cancers. We evaluate relevant methods to assess the levels and extent of hypoxia in childhood cancers, including novel imaging strategies such as oxygen-enhanced magnetic resonance imaging (MRI). Preclinical and clinical evidence largely supports the use of hypoxia-targeting drugs in children, and we describe the critical need to identify robust predictive biomarkers for the use of such drugs in future paediatric clinical trials. Ultimately, a more personalised approach to treatment that includes targeting hypoxic tumour cells might improve outcomes in subgroups of paediatric cancer patients.
Collapse
Affiliation(s)
- Carolina Bernauer
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK
| | - Y K Stella Man
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK
| | - Julia C Chisholm
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Surrey, UK
- Sarcoma Clinical Trials in Children and Young People Team, The Institute of Cancer Research, London, UK
| | - Elise Y Lepicard
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Janet M Shipley
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK.
| |
Collapse
|
56
|
Nishihara K, Hori K, Saito T, Omori T, Sunakawa H, Minamide T, Suyama M, Yamamoto Y, Yoda Y, Shinmura K, Ikematsu H, Yano T. A study of evaluating specific tissue oxygen saturation values of gastrointestinal tumors by removing adherent substances in oxygen saturation imaging. PLoS One 2021; 16:e0243165. [PMID: 33411775 PMCID: PMC7790263 DOI: 10.1371/journal.pone.0243165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/16/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Oxygen saturation (OS) imaging is a new method of endoscopic imaging that has clinical applications in oncology which can directly measure tissue oxygen saturation (Sto2) of the surface of gastrointestinal tract without any additional drugs or devices. This imaging technology is expected to contribute to research into cancer biology which leads to clinical benefit such as prediction to efficacy of chemotherapy or radiotherapy. However, adherent substances on tumors such as blood and white coating, pose a challenge for accurate measurements of the StO2 values in tumors. The aim of this study was to develop algorithms for discriminating between the tumors and their adherent substances, and to investigate whether it is possible to evaluate the tumor specific StO2 values excluding adherent substances during OS imaging. METHODS We plotted areas of tumors and their adherent substances using white-light images of 50 upper digestive tumors: blood (68 plots); reddish tumor (83 plots); white coating (89 plots); and whitish tumor (79 plots). Scatter diagrams and discriminating algorithms using spectrum signal intensity values were constructed and verified using validation datasets. StO2 values were compared between the tumors and tumor adherent substances using OS images of gastrointestinal tumors. RESULTS The discriminating algorithms and their accuracy rates (AR) were as follows: blood vs. reddish tumor: Y> - 4.90X+7.13 (AR: 95.9%) and white coating vs. whitish tumor: Y< -0.52X+0.17 (AR: 96.0%). The StO2 values (median, [range]) were as follows: blood, 79.3% [37.8%-100.0%]; reddish tumor, 74.5% [62.0%-86.9%]; white coating, 73.8% [42.1%-100.0%]; and whitish tumor, 65.7% [53.0%-76.3%]. CONCLUSIONS OS imaging is strongly influenced by adherent substances for evaluating the specific StO2 value of tumors; therefore, it is important to eliminate the information of adherent substances for clinical application of OS imaging.
Collapse
Affiliation(s)
- Keiichiro Nishihara
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Keisuke Hori
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Takaaki Saito
- Imaging Technology Center, FUJIFILM Corporation, Tokyo, Japan
| | - Toshihiko Omori
- Medical Systems Research & Development Center, Research & Development, Management Headquarters, FUJIFILM Corporation, Tokyo Japan
| | - Hironori Sunakawa
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Tatsunori Minamide
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Masayuki Suyama
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Yoichi Yamamoto
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Yusuke Yoda
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Kensuke Shinmura
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Hiroaki Ikematsu
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| | - Tomonori Yano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital, Kashiwanoha, Kashiwa, Japan
| |
Collapse
|
57
|
Abstract
Prostate cancer (PCa) is a clinically heterogeneous disease and has poor patient outcome when tumours progress to castration-resistant and metastatic states. Understanding the mechanistic basis for transition to late stage aggressive disease is vital for both assigning patient risk status in the localised setting and also identifying novel treatment strategies to prevent progression. Subregions of intratumoral hypoxia are found in all solid tumours and are associated with many biologic drivers of tumour progression. Crucially, more recent findings show the co-presence of hypoxia and genomic instability can confer a uniquely adverse prognosis in localised PCa patients. In-depth informatic and functional studies suggests a role for hypoxia in co-operating with oncogenic drivers (e.g. loss of PTEN) and suppressing DNA repair capacity to alter clonal evolution due to an aggressive mutator phenotype. More specifically, hypoxic suppression of homologous recombination represents a “contextual lethal“ vulnerability in hypoxic prostate tumours which could extend the application of existing DNA repair targeting agents such as poly-ADP ribose polymerase inhibitors. Further investigation is now required to assess this relationship on the background of existing genomic alterations relevant to PCa, and also characterise the role of hypoxia in driving early metastatic spread. On this basis, PCa patients with hypoxic tumours can be better stratified into risk categories and treated with appropriate therapies to prevent progression.
Collapse
Affiliation(s)
- Jack Ashton
- Translational Oncogenomics, CRUK Manchester Institute and CRUK Manchester Centre, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert Bristow
- Translational Oncogenomics, CRUK Manchester Institute and CRUK Manchester Centre, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
58
|
Hypoxia and Oxygen-Sensing Signaling in Gene Regulation and Cancer Progression. Int J Mol Sci 2020; 21:ijms21218162. [PMID: 33142830 PMCID: PMC7663541 DOI: 10.3390/ijms21218162] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022] Open
Abstract
Oxygen homeostasis regulation is the most fundamental cellular process for adjusting physiological oxygen variations, and its irregularity leads to various human diseases, including cancer. Hypoxia is closely associated with cancer development, and hypoxia/oxygen-sensing signaling plays critical roles in the modulation of cancer progression. The key molecules of the hypoxia/oxygen-sensing signaling include the transcriptional regulator hypoxia-inducible factor (HIF) which widely controls oxygen responsive genes, the central members of the 2-oxoglutarate (2-OG)-dependent dioxygenases, such as prolyl hydroxylase (PHD or EglN), and an E3 ubiquitin ligase component for HIF degeneration called von Hippel–Lindau (encoding protein pVHL). In this review, we summarize the current knowledge about the canonical hypoxia signaling, HIF transcription factors, and pVHL. In addition, the role of 2-OG-dependent enzymes, such as DNA/RNA-modifying enzymes, JmjC domain-containing enzymes, and prolyl hydroxylases, in gene regulation of cancer progression, is specifically reviewed. We also discuss the therapeutic advancement of targeting hypoxia and oxygen sensing pathways in cancer.
Collapse
|
59
|
Subcellular Location of Tirapazamine Reduction Dramatically Affects Aerobic but Not Anoxic Cytotoxicity. Molecules 2020; 25:molecules25214888. [PMID: 33105798 PMCID: PMC7660101 DOI: 10.3390/molecules25214888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 11/16/2022] Open
Abstract
Hypoxia is an adverse prognostic feature of solid cancers that may be overcome with hypoxia-activated prodrugs (HAPs). Tirapazamine (TPZ) is a HAP which has undergone extensive clinical evaluation in this context and stimulated development of optimized analogues. However the subcellular localization of the oxidoreductases responsible for mediating TPZ-dependent DNA damage remains unclear. Some studies conclude only nuclear-localized oxidoreductases can give rise to radical-mediated DNA damage and thus cytotoxicity, whereas others identify a broader role for endoplasmic reticulum and cytosolic oxidoreductases, indicating the subcellular location of TPZ radical formation is not a critical requirement for DNA damage. To explore this question in intact cells we engineered MDA-231 breast cancer cells to express the TPZ reductase human NADPH: cytochrome P450 oxidoreductase (POR) harboring various subcellular localization sequences to guide this flavoenzyme to the nucleus, endoplasmic reticulum, cytosol or inner surface of the plasma membrane. We show that all POR variants are functional, with differences in rates of metabolism reflecting enzyme expression levels rather than intracellular TPZ concentration gradients. Under anoxic conditions, POR expression in all subcellular compartments increased the sensitivity of the cells to TPZ, but with a fall in cytotoxicity per unit of metabolism (termed ‘metabolic efficiency’) when POR is expressed further from the nucleus. However, under aerobic conditions a much larger increase in cytotoxicity was observed when POR was directed to the nucleus, indicating very high metabolic efficiency. Consequently, nuclear metabolism results in collapse of hypoxic selectivity of TPZ, which was further magnified to the point of reversing O2 dependence (oxic > hypoxic sensitivity) by employing a DNA-affinic TPZ analogue. This aerobic hypersensitivity phenotype was partially rescued by cellular copper depletion, suggesting the possible involvement of Fenton-like chemistry in generating short-range effects mediated by the hydroxyl radical. In addition, the data suggest that under aerobic conditions reoxidation strictly limits the TPZ radical diffusion range resulting in site-specific cytotoxicity. Collectively these novel findings challenge the purported role of intra-nuclear reductases in orchestrating the hypoxia selectivity of TPZ.
Collapse
|
60
|
Aggarwal V, Miranda O, Johnston PA, Sant S. Three dimensional engineered models to study hypoxia biology in breast cancer. Cancer Lett 2020; 490:124-142. [PMID: 32569616 PMCID: PMC7442747 DOI: 10.1016/j.canlet.2020.05.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022]
Abstract
Breast cancer is the second leading cause of mortality among women worldwide. Despite the available therapeutic regimes, variable treatment response is reported among different breast cancer subtypes. Recently, the effects of the tumor microenvironment on tumor progression as well as treatment responses have been widely recognized. Hypoxia and hypoxia inducible factors in the tumor microenvironment have long been known as major players in tumor progression and survival. However, the majority of our understanding of hypoxia biology has been derived from two dimensional (2D) models. Although many hypoxia-targeted therapies have elicited promising results in vitro and in vivo, these results have not been successfully translated into clinical trials. These limitations of 2D models underscore the need to develop and integrate three dimensional (3D) models that recapitulate the complex tumor-stroma interactions in vivo. This review summarizes role of hypoxia in various hallmarks of cancer progression. We then compare traditional 2D experimental systems with novel 3D tissue-engineered models giving accounts of different bioengineering platforms available to develop 3D models and how these 3D models are being exploited to understand the role of hypoxia in breast cancer progression.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Oshin Miranda
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Paul A Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
61
|
Catalytic nanographene oxide with hemin for enhanced photodynamic therapy. J Control Release 2020; 326:442-454. [DOI: 10.1016/j.jconrel.2020.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/26/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
|
62
|
Sonocomplexation as an effective tool to enhance the antitumorigenic effect of metformin: Preparation, in vitro characterization, molecular dynamic simulation & MiaPaCa-2 cell line hypoxia evaluation. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
63
|
Shobaki N, Sato Y, Suzuki Y, Okabe N, Harashima H. Manipulating the function of tumor-associated macrophages by siRNA-loaded lipid nanoparticles for cancer immunotherapy. J Control Release 2020; 325:235-248. [DOI: 10.1016/j.jconrel.2020.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/29/2020] [Accepted: 07/03/2020] [Indexed: 01/04/2023]
|
64
|
Rickard AG, Yoshikawa H, Palmer GM, Liu HQ, Dewhirst MW, Nolan MW, Zhang X. Cherenkov emissions for studying tumor changes during radiation therapy: An exploratory study in domesticated dogs with naturally-occurring cancer. PLoS One 2020; 15:e0238106. [PMID: 32845905 PMCID: PMC7449466 DOI: 10.1371/journal.pone.0238106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/10/2020] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Real-time monitoring of physiological changes of tumor tissue during radiation therapy (RT) could improve therapeutic efficacy and predict therapeutic outcomes. Cherenkov radiation is a normal byproduct of radiation deposited in tissue. Previous studies in rat tumors have confirmed a correlation between Cherenkov emission spectra and optical measurements of blood-oxygen saturation based on the tissue absorption coefficients. The purpose of this study is to determine if it is feasible to image Cherenkov emissions during radiation therapy in larger human-sized tumors of pet dogs with cancer. We also wished to validate the prior work in rats, to determine if Cherenkov emissions have the potential to act an indicator of blood-oxygen saturation or water-content changes in the tumor tissue-both of which have been correlated with patient prognosis. METHODS A DoseOptics camera, built to image the low-intensity emission of Cherenkov radiation, was used to measure Cherenkov intensities in a cohort of cancer-bearing pet dogs during clinical irradiation. Tumor type and location varied, as did the radiation fractionation scheme and beam arrangement, each planned according to institutional standard-of-care. Unmodulated radiation was delivered using multiple 6 MV X-ray beams from a clinical linear accelerator. Each dog was treated with a minimum of 16 Gy total, in ≥3 fractions. Each fraction was split into at least three subfractions per gantry angle. During each subfraction, Cherenkov emissions were imaged. RESULTS We documented significant intra-subfraction differences between the Cherenkov intensities for normal tissue, whole-tumor tissue, tissue at the edge of the tumor and tissue at the center of the tumor (p<0.05). Additionally, intra-subfraction changes suggest that Cherenkov emissions may have captured fluctuating absorption properties within the tumor. CONCLUSION Here we demonstrate that it is possible to obtain Cherenkov emissions from canine cancers within a fraction of radiotherapy. The entire optical spectrum was obtained which includes the window for imaging changes in water and hemoglobin saturation. This lends credence to the goal of using this method during radiotherapy in human patients and client-owned pets.
Collapse
Affiliation(s)
- Ashlyn G. Rickard
- Department of Radiation Oncology, Program of Medical Physics, Duke University School of Medicine, Durham, NC, United States of America
| | - Hiroto Yoshikawa
- Department of Clinical Sciences, College of Veterinary Medicine, NC State University, Raleigh, NC, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States of America
| | - Gregory M. Palmer
- Department of Radiation Oncology, Program of Medical Physics, Duke University School of Medicine, Durham, NC, United States of America
- Department of Clinical Sciences, College of Veterinary Medicine, NC State University, Raleigh, NC, United States of America
| | - Harrison Q. Liu
- Program of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Program of Medical Physics, Duke University School of Medicine, Durham, NC, United States of America
- Department of Clinical Sciences, College of Veterinary Medicine, NC State University, Raleigh, NC, United States of America
| | - Michael W. Nolan
- Department of Clinical Sciences, College of Veterinary Medicine, NC State University, Raleigh, NC, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States of America
- Duke Cancer Institute, Duke University, Durham, NC, United States of America
| | - Xiaofeng Zhang
- Artificial Intelligence, Marchex Inc., Seattle, WA, United States of America
| |
Collapse
|
65
|
Wei J, Huang K, Chen Z, Hu M, Bai Y, Lin S, Du H. Characterization of Glycolysis-Associated Molecules in the Tumor Microenvironment Revealed by Pan-Cancer Tissues and Lung Cancer Single Cell Data. Cancers (Basel) 2020; 12:cancers12071788. [PMID: 32635458 PMCID: PMC7408567 DOI: 10.3390/cancers12071788] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022] Open
Abstract
Altered metabolism is a hallmark of cancer and glycolysis is one of the important factors promoting tumor development. There is however still a lack of molecular characterization glycolysis and comprehensive studies related to tumor glycolysis in the pan-cancer landscape. Here, we applied a gene expression signature to quantify glycolysis in 9229 tumors across 25 cancer types and 7875 human lung cancer single cells and verified the robustness of signature using defined glycolysis samples from previous studies. We classified tumors and cells into glycolysis score-high and -low groups, demonstrated their prognostic associations, and identified genome and transcriptome molecular features associated with glycolysis activity. We observed that glycolysis score-high tumors were associated with worse prognosis across cancer types. High glycolysis tumors exhibited specific driver genes altered by copy number aberrations (CNAs) in most cancer types. Tricarboxylic acid (TCA) cycle, DNA replication, tumor proliferation and other cancer hallmarks were more active in glycolysis-high tumors. Glycolysis signature was strongly correlated with hypoxia signature in all 25 cancer tissues (r > 0.7) and cancer single cells (r > 0.8). In addition, HSPA8 and P4HA1 were screened out as the potential modulating factors to glycolysis as their expression were highly correlated with glycolysis score and glycolysis genes, which enables future efforts for therapeutic options to block the glycolysis and control tumor progression. Our study provides a comprehensive molecular-level understanding of glycolysis with a large sample data and demonstrates the hypoxia pressure, growth signals, oncogene mutation and other potential signals could activate glycolysis, thereby to regulate cell cycle, energy material synthesis, cell proliferation and cancer progression.
Collapse
|
66
|
Kozłowska J, Kozioł K, Stasiak M, Obacz J, Guglas K, Poter P, Mackiewicz A, Kolenda T. The role of NEAT1 lncRNA in squamous cell carcinoma of the head and neck is still difficult to define. Contemp Oncol (Pozn) 2020; 24:96-105. [PMID: 32774134 PMCID: PMC7403767 DOI: 10.5114/wo.2020.97635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Nuclear paraspeckle assembly transcript 1 (NEAT1) is considered an oncogene in various cancers, but the role in head and neck squamous cell carcinomas (HNSCC) is not clear. MATERIAL AND METHODS Expression of NEAT1 in HNSCC patients' samples and cell lines was analysed using qRT-PCR. The TCGA expression data of NEAT1 were analysed depending on the clinicopathological parameters and tumour localisation. Correlation and gene set enrichment analysis (GSEA) were conducted, and the results were analysed using the REACTOME and GeneMANIA tools. All statistical analyses were carried out using GraphPad Prism 5 and Statistica 13. RESULTS The NEAT1 was up-regulated in some patients' samples and HNSCC cell lines. Moreover, TCGA data analysis indicated that the expression of NEAT1 was up-regulated in tumour tissue in most of the analysed TCGA cancers, including HNSCC. There were no significant differences in levels of NEAT1 between various tumour localisations. Overall survival of individuals with high expression of NEAT1 was slightly longer than in the low-expression group (p = 0.0553). Analysis of genes that positively and negatively correlated with NEAT1 indicated that they are involved in mRNA metabolism and cellular transport. Moreover, the GSEA revealed that in patients with low NEAT1, the most up-regulated genes were in clusters associated with the cAMP-dependent pathway, the MYC pathway, unfolded protein response, the MTORC1 signalling pathway, oxidative phosphorylation, and DNA repair. CONCLUSIONS Patients with low expression of NEAT1 display worse overall survival, presumably due to up-regulation of certain oncogenic signalling pathways that are important for cancerogenesis.
Collapse
Affiliation(s)
- Joanna Kozłowska
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kinga Kozioł
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Stasiak
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Justyna Obacz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kacper Guglas
- Laboratory of Cancer Genetics, Greater Poland Cancer Centre, Poznan, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Paulina Poter
- Department of Oncologic Pathology and Prophylaxis, Poznan University of Medical Sciences, Greater Poland Cancer Centre, Poznan, Poland
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| | - Tomasz Kolenda
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
67
|
Waller J, Onderdonk B, Flood A, Swartz H, Shah J, Shah A, Aydogan B, Halpern H, Hasan Y. The clinical utility of imaging methods used to measure hypoxia in cervical cancer. Br J Radiol 2020; 93:20190640. [PMID: 32286849 PMCID: PMC7336054 DOI: 10.1259/bjr.20190640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 03/18/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
While it is well-established that hypoxia is a major factor that affects clinical outcomes in cervical cancer, widespread usage of clinically available methods to detect and evaluate hypoxia during the course of treatment have not been established. This review compares these methods, summarizes their strengths and weaknesses, and assesses the pathways for their useful employment to alter clinical practice. We conducted a search on PubMed for literature pertaining to imaging hypoxic cervical cancer, and implemented keywords related to oxygen measurement tools to improve the relevance of the search results.Oxygenation level-dependent applications of MRI have demonstrated hypoxia-induced radioresistance, and changes in cervix tumor oxygenation from hyperoxic therapy.The hypoxic areas within tumors can be indirectly identified in dynamic contrast-enhanced images, where they generally display low signal enhancement, and diffusion-weighted images, which demonstrates areas of restricted diffusion (which correlates with hypoxia). Positron emmision tomography, used independently and with other imaging modalities, has demonstrated utility in imaging hypoxia through tracers specific for low oxygen levels, like Cu-ATSM tracers and nitroimidazoles. Detecting hypoxia in the tumors of patients diagnosed with cervical cancer via medical imaging and non-imaging tools like electron paramagnetic resonance oximetry can be utilized clinically, such as for guiding radiation and post-treatment surveillance, for a more personalized approach to treatment. The merits of these methods warrant further investigation via comparative effectiveness research and large clinical trials into their clinical applications.
Collapse
Affiliation(s)
- Joseph Waller
- Drexel College of Medicine, 2900 W Queen Ln, PA 19129, United States
| | - Benjamin Onderdonk
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Ann Flood
- Department of Radiology, Dartmouth Geisel School of Medicine, 1 Rope Ferry Rd, NH 03755, United States
| | - Harold Swartz
- Department of Radiology, Dartmouth Geisel School of Medicine, 1 Rope Ferry Rd, NH 03755, United States
| | - Jaffer Shah
- Drexel College of Medicine, 2900 W Queen Ln, PA 19129, United States
| | - Asghar Shah
- Brown University, Providence, RI 02912, United States
| | - Bulent Aydogan
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Howard Halpern
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Yasmin Hasan
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| |
Collapse
|
68
|
Gonzalez-Avila G, Sommer B, García-Hernández AA, Ramos C. Matrix Metalloproteinases' Role in Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1245:97-131. [PMID: 32266655 DOI: 10.1007/978-3-030-40146-7_5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer cells evolve in the tumor microenvironment (TME) by the acquisition of characteristics that allow them to initiate their passage through a series of events that constitute the metastatic cascade. For this purpose, tumor cells maintain a crosstalk with TME non-neoplastic cells transforming them into their allies. "Corrupted" cells such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and tumor-associated neutrophils (TANs) as well as neoplastic cells express and secrete matrix metalloproteinases (MMPs). Moreover, TME metabolic conditions such as hypoxia and acidification induce MMPs' synthesis in both cancer and stromal cells. MMPs' participation in TME consists in promoting events, for example, epithelial-mesenchymal transition (EMT), apoptosis resistance, angiogenesis, and lymphangiogenesis. MMPs also facilitate tumor cell migration through the basement membrane (BM) and extracellular matrix (ECM). The aim of the present chapter is to discuss MMPs' contribution to the evolution of cancer cells, their cellular origin, and their influence in the main processes that take place in the TME.
Collapse
Affiliation(s)
- Georgina Gonzalez-Avila
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico.
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - A Armando García-Hernández
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - Carlos Ramos
- Laboratorio de Biología Celular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| |
Collapse
|
69
|
Bhandari V, Li CH, Bristow RG, Boutros PC. Divergent mutational processes distinguish hypoxic and normoxic tumours. Nat Commun 2020; 11:737. [PMID: 32024819 PMCID: PMC7002770 DOI: 10.1038/s41467-019-14052-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 11/29/2019] [Indexed: 12/14/2022] Open
Abstract
Many primary tumours have low levels of molecular oxygen (hypoxia), and hypoxic tumours respond poorly to therapy. Pan-cancer molecular hallmarks of tumour hypoxia remain poorly understood, with limited comprehension of its associations with specific mutational processes, non-coding driver genes and evolutionary features. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole genome sequencing data from 2658 cancers across 38 tumour types, we quantify hypoxia in 1188 tumours spanning 27 cancer types. Elevated hypoxia associates with increased mutational load across cancer types, irrespective of underlying mutational class. The proportion of mutations attributed to several mutational signatures of unknown aetiology directly associates with the level of hypoxia, suggesting underlying mutational processes for these signatures. At the gene level, driver mutations in TP53, MYC and PTEN are enriched in hypoxic tumours, and mutations in PTEN interact with hypoxia to direct tumour evolutionary trajectories. Overall, hypoxia plays a critical role in shaping the genomic and evolutionary landscapes of cancer.
Collapse
Affiliation(s)
- Vinayak Bhandari
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Constance H Li
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, USA
| | - Robert G Bristow
- Division of Cancer Sciences, Faculty of Biology, Health and Medicine, University of Manchester, Manchester, UK.
- The Christie NHS Foundation Trust, Manchester, UK.
- CRUK Manchester Institute and Centre, Manchester, UK.
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Department of Human Genetics, University of California, Los Angeles, USA.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada.
- Vector Institute for Artificial Intelligence, Toronto, Canada.
- Department of Urology, University of California, Los Angeles, USA.
- Jonsson Comprehensive Cancer Centre, University of California Los Angeles, Los Angeles, USA.
- Institute for Precision Health, University of California Los Angeles, Los Angeles, USA.
| |
Collapse
|
70
|
Taylor E, Zhou J, Lindsay P, Foltz W, Cheung M, Siddiqui I, Hosni A, Amir AE, Kim J, Hill RP, Jaffray DA, Hedley DW. Quantifying Reoxygenation in Pancreatic Cancer During Stereotactic Body Radiotherapy. Sci Rep 2020; 10:1638. [PMID: 32005829 PMCID: PMC6994660 DOI: 10.1038/s41598-019-57364-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/18/2019] [Indexed: 02/05/2023] Open
Abstract
Hypoxia, the state of low oxygenation that often arises in solid tumours due to their high metabolism and irregular vasculature, is a major contributor to the resistance of tumours to radiation therapy (RT) and other treatments. Conventional RT extends treatment over several weeks or more, and nominally allows time for oxygen levels to increase ("reoxygenation") as cancer cells are killed by RT, mitigating the impact of hypoxia. Recent advances in RT have led to an increase in the use stereotactic body radiotherapy (SBRT), which delivers high doses in five or fewer fractions. For cancers such as pancreatic adenocarcinoma for which hypoxia varies significantly between patients, SBRT might not be optimal, depending on the extent to which reoxygenation occurs during its short duration. We used fluoro-5-deoxy-α-D-arabinofuranosyl)-2-nitroimidazole positron-emission tomography (FAZA-PET) imaging to quantify hypoxia before and after 5-fraction SBRT delivered to patient-derived pancreatic cancer xenografts orthotopically implanted in mice. An imaging technique using only the pre-treatment FAZA-PET scan and repeat dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) scans throughout treatment was able to predict the change in hypoxia. Our results support the further testing of this technique for imaging of reoxygenation in the clinic.
Collapse
Affiliation(s)
- Edward Taylor
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Jitao Zhou
- Department of Abdominal Oncology, Cancer Center and Laboratory of Signal Transduction and Molecular Targeting Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Patricia Lindsay
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Warren Foltz
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - May Cheung
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Iram Siddiqui
- Department of Pathology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Ali Hosni
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Ahmed El Amir
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - John Kim
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - Richard P Hill
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - David A Jaffray
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | - David W Hedley
- Ontario Cancer Institute, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada.
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada.
| |
Collapse
|
71
|
Dan Q, Hu D, Ge Y, Zhang S, Li S, Gao D, Luo W, Ma T, Liu X, Zheng H, Li Y, Sheng Z. Ultrasmall theranostic nanozymes to modulate tumor hypoxia for augmenting photodynamic therapy and radiotherapy. Biomater Sci 2020; 8:973-987. [DOI: 10.1039/c9bm01742a] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A theranostic nanozyme (Au NCs-ICG) decomposes intratumoral H2O2 to O2, subsequently enhancing photodynamic therapy and radiotherapy with the guidance of multimodal imaging.
Collapse
|
72
|
Kumari R, Sunil D, Ningthoujam RS. Hypoxia-responsive nanoparticle based drug delivery systems in cancer therapy: An up-to-date review. J Control Release 2019; 319:135-156. [PMID: 31881315 DOI: 10.1016/j.jconrel.2019.12.041] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 02/08/2023]
Abstract
Hypoxia is a salient feature observed in most solid malignancies that holds a pivotal role in angiogenesis, metastasis and resistance to conventional cancer therapeutic approaches, and thus enables cancer progression. However, the typical characteristics of hypoxic cells such as low oxygen levels and highly bio-reductive environment can offer stimuli-responsive drug release to aid in tumor-specific chemo, radio, photodyanamic and sonodynamic therapies. This approach based on targeting the poorly oxygenated tumor habitats offers the prospective to overcome the difficulties that arises due to heterogenic nature of tumor and could be possibly used in the design of diagnostic as well as therapeutic nanocarriers for targeting various types of solid cancers. Consequently, hypoxia triggered nanoparticle based drug delivery systems is a rapidly progressing research area in developing effective strategies to combat drug-resistance in solid tumors. The present review presents the recent advances in the development of hypoxia-responsive nanovehicles for drug delivery to heterogeneous tumors. The initial sections of the article provides insights into the development of hypoxia in growing cancer and its role in disease progression. The current limitations and the future prospective of hypoxia-stimulated nanomachines for cancer treatment are also discussed.
Collapse
Affiliation(s)
- Rashmi Kumari
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576 104, Karnataka, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576 104, Karnataka, India.
| | | |
Collapse
|
73
|
Li R, Zhang J, Guo J, Xu Y, Duan K, Zheng J, Wan H, Yuan Z, Chen H. Application of Nitroimidazole-Carbobane-Modified Phenylalanine Derivatives as Dual-Target Boron Carriers in Boron Neutron Capture Therapy. Mol Pharm 2019; 17:202-211. [PMID: 31763850 DOI: 10.1021/acs.molpharmaceut.9b00898] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Boron neutron capture therapy (BNCT) has received extensive attention as noninvasive cell-level oncotherapy for treating solid cancer tumors. However, boron-containing drugs such as l-boronophenylalanine (BPA) and sodium borocaptate have low boron content and/or poor tumor-targeting ability, limiting their application. In this study, we designed and synthesized a series of nontoxic, dual-target boron carriers (B139, B142, and B151) with the ability to accumulate specifically in tumor cells. We found that the B139 uptake into hypoxic tumor regions was high, with a 70-fold boron content compared to BPA. In addition, in vivo observation showed that B139 can be trapped in tumor cells for a prolonged period and maintains an effective therapeutic concentration, with a peak boron concentration of 50.7 μg/g and a high tumor: blood boron ratio of >3, achieving ideal BNCT conditions. Cytotoxicity evaluation in mice further proved that B139 is safe and reliable. Therefore, B139 has great potential for BNCT application as a dual-target, safe, and efficient boron carrier.
Collapse
Affiliation(s)
- Ruixi Li
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Juanjuan Zhang
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Jingxuan Guo
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Yue Xu
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Kunyuan Duan
- Department of Pharmacy , Qujing Medical College , Qujing 655000 , China
| | - Jinrong Zheng
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Hao Wan
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| |
Collapse
|
74
|
Matolay O, Méhes G. Sustain, Adapt, and Overcome-Hypoxia Associated Changes in the Progression of Lymphatic Neoplasia. Front Oncol 2019; 9:1277. [PMID: 31824854 PMCID: PMC6881299 DOI: 10.3389/fonc.2019.01277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/04/2019] [Indexed: 12/15/2022] Open
Abstract
Irregular perfusion and related tissue hypoxia is a common feature of solid tumors the role of which in the survival and progression cancer has been gradually recognized. Adaptation and selection mechanisms in hypoxic areas in solid tumors are regulated by Hypoxia Inducible transcriptional factor 1 (HIF1) and other hypoxia mediators and are associated with aggressive clinical behavior in a large spectrum of malignancies. Aggressive forms of lymphatic neoplasias present with solid tumor-like features, also including rapid cell growth, necrosis and angiogenesis, the clinical potential of which is still underestimated. While the role of regional hypoxia in normal B-cell maturation and malignant transformation is becoming evident, the impact of tissue hypoxia on their behavior is not well-understood. Compared to some of the common solid cancer types data for some of the key regulators, such as HIF1 and HIF2, and for their downstream effectors are available in a limited fashion. In the current review we aim to overview the physiological aspects of major hypoxia pathways during B-cell maturation and adaptation-related changes reported in lymphatic neoplasia covering important targets, such as carbonic anhydrases IX and XII (CAIX, CAXII), glucose transporter 1 (GLUT-1) and vascular endothelial growth factor (VEGF). In conclusion, experimental and clinical results direct to important but currently unexploited role of hypoxia-driven resistance mechanisms especially in aggressive forms of B-cell neoplasia.
Collapse
Affiliation(s)
- Orsolya Matolay
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
75
|
Lama-Sherpa TD, Shevde LA. An Emerging Regulatory Role for the Tumor Microenvironment in the DNA Damage Response to Double-Strand Breaks. Mol Cancer Res 2019; 18:185-193. [PMID: 31676722 DOI: 10.1158/1541-7786.mcr-19-0665] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/23/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022]
Abstract
Radiation, alkylating agents, and platinum-based chemotherapy treatments eliminate cancer cells through the induction of excessive DNA damage. The resultant DNA damage challenges the cancer cell's DNA repair capacity. Among the different types of DNA damage induced in cells, double-strand breaks (DSB) are the most lethal if left unrepaired. Unrepaired DSBs in tumor cells exacerbate existing gene deletions, chromosome losses and rearrangements, and aberrant features that characteristically enable tumor progression, metastasis, and drug resistance. Tumor microenvironmental factors like hypoxia, inflammation, cellular metabolism, and the immune system profoundly influence DSB repair mechanisms. Here, we put into context the role of the microenvironment in governing DSB repair mechanisms.
Collapse
Affiliation(s)
| | - Lalita A Shevde
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama. .,O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
76
|
Sahu A, Kwon I, Tae G. Improving cancer therapy through the nanomaterials-assisted alleviation of hypoxia. Biomaterials 2019; 228:119578. [PMID: 31678843 DOI: 10.1016/j.biomaterials.2019.119578] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 02/08/2023]
Abstract
Hypoxia, resulting from the imbalance between oxygen supply and consumption is a critical component of the tumor microenvironment. It has a paramount impact on cancer growth, metastasis and has long been known as a major obstacle for cancer therapy. However, none of the clinically approved anticancer therapeutics currently available for human use directly tackles this problem. Previous clinical trials of targeting tumor hypoxia with bioreductive prodrugs have failed to demonstrate satisfactory results. Therefore, new ideas are needed to overcome the hypoxia barrier. The method of modulating hypoxia to improve the therapeutic activity is of great interest but remains a considerable challenge. One of the emerging concepts is to supply or generate oxygen at the tumor site to increase the partial oxygen pressure and thereby reverse the hypoxia and its effects. In this review, we present an overview of the recent progress in the development of novel nanomaterials for the alleviation of hypoxic microenvironment. Two main strategies for hypoxia augmentation, i) direct delivery of O2 into the tumor, and ii) in situ O2 generations in the tumor microenvironment through different methods such as catalytic decomposition of endogenous hydrogen peroxide (H2O2) and light-triggered water splitting are discussed in detail. At present, these emerging nanomaterials are in their early phase and expected to grow rapidly in the coming years. Despite the promising start, there are several challenges needed to overcome for successful clinical translation.
Collapse
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Inchan Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
77
|
Vermeulen MA, van Deurzen CH, Schroder CP, Martens JW, van Diest PJ. Expression of hypoxia-induced proteins in ductal carcinoma in situ and invasive cancer of the male breast. J Clin Pathol 2019; 73:204-208. [PMID: 31653758 DOI: 10.1136/jclinpath-2019-206116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 11/04/2022]
Abstract
AIMS The aim of this study was to determine the role of hypoxia in male breast carcinogenesis by evaluating the expression of the hypoxia-related proteins, hypoxia-inducible factor-1α (HIF-1α), carbonic anhydrase IX (CAIX) and glucose transporter-1 (Glut-1), in ductal carcinoma in situ (DCIS) of the male breast in relation to invasive cancer (IC). METHODS Tumour tissue blocks of 18 cases of pure DCIS, 58 DCIS cases adjacent to IC (DCIS-AIC) and the 58 IC cases were stained by immunohistochemistry for HIF-1α, CAIX and Glut-1, and expression frequencies and patterns (diffuse and/or perinecrotic) were noted. RESULTS HIF-1α overexpression was observed in 61.1% (11/18) of pure DCIS, in 37.9% (22/58) of DCIS-AIC and in 36.2% (21/58) of IC cases (not significant (n.s.)). CAIX overexpression was observed in 16.7% (3/18) of pure DCIS, in 37.9% (22/58) of DCIS-AIC and in 24.1% (14/58) of IC cases (n.s.). Glut-1 overexpression was observed in 61.1% (11/18) of pure DCIS, in 75.9% (44/58) of DCIS-AIC and in 62.1% (36/58) of IC cases (n.s.). Expression of hypoxia-related proteins was seen around necrosis in a little over one-third of DCIS cases, and often coincided with expression in adjacent IC when present. All these observations indicate that the hypoxia response is already at its maximum in the preinvasive DCIS stage. CONCLUSIONS In conclusion, male DCIS frequently shows activated hypoxia response, comparable to male IC. This indicates that the activated hypoxia response previously seen in male IC is not a late bystander but likely a genuine carcinogenetic event.
Collapse
Affiliation(s)
- Marijn A Vermeulen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carolien Hm van Deurzen
- Department of Pathology, Erasmus MC Cancer Institute/Erasmus University Medical Center, Rotterdam, The Netherlands.,BOOG Study Center/Dutch Breast Cancer Research Group, Amsterdam, The Netherlands
| | - Carolien P Schroder
- BOOG Study Center/Dutch Breast Cancer Research Group, Amsterdam, The Netherlands.,Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - John Wm Martens
- BOOG Study Center/Dutch Breast Cancer Research Group, Amsterdam, The Netherlands.,Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute/Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
78
|
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
|
79
|
Zhang D, Yang L, Liu X, Gao J, Liu T, Yan Q, Yang X. Hypoxia modulates stem cell properties and induces EMT through N-glycosylation of EpCAM in breast cancer cells. J Cell Physiol 2019; 235:3626-3633. [PMID: 31584203 DOI: 10.1002/jcp.29252] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/26/2019] [Indexed: 12/11/2022]
Abstract
Epithelial cell adhesion molecule (EpCAM), which is a transmembrane glycoprotein, is related to tumor progression. We demonstrated that EpCAM plays important roles in proliferation, apoptosis, and metastasis during breast cancer (BC) progression. But the role of N-glycosylation in EpCAM in tumor aggressiveness is not clear. Here, we evaluated the role of N-glycosylation of EpCAM in stemness and epithelial-mesenchymal transition (EMT) characteristics. EpCAM overexpression increases the expression of stemness markers (NANOG,SOX2, and OCT4) and EMT markers (N-cadherin and vimentin) under the condition of hypoxia in BC. Knockdown of EpCAM and mutation of N-glycosylation of EpCAM maintained in severe hypoxia lead to a significant reduction of stemness/EMT markers. In addition, we found that N-glycosylation of EpCAM is a crucial factor during this process. This demonstrates that EpCAM has a novel regulatory role in stemness/EMT dependence of hypoxia-inducible factor 1-alpha via regulating nuclear factor kappa B in BC cells. Hence, our study reveals EpCAM glycosylation modification as a new regulator of stemness/EMT under hypoxic in BC and points out EpCAM as a potential therapeutic target.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| | - Liu Yang
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| | - Xue Liu
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| | - Jiujiao Gao
- Department of Molecular Medicine, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Tingjiao Liu
- Section of Oral Pathology, College of Stomatology, Dalian Medical University, Dalian, China
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| |
Collapse
|
80
|
Lukovic J, Han K, Pintilie M, Chaudary N, Hill RP, Fyles A, Milosevic M. Intratumoral heterogeneity and hypoxia gene expression signatures: Is a single biopsy adequate? Clin Transl Radiat Oncol 2019; 19:110-115. [PMID: 31650046 PMCID: PMC6804682 DOI: 10.1016/j.ctro.2019.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
A single biopsy may not accurately reflect the global hypoxia status of a tumor due to intratumoral heterogeneity. Compared with individual genes, hypoxia gene expression signatures are generally more consistent across multiple biopsies from different regions of a tumor and may give a more reliable estimate of global hypoxia status. Wherever possible, the use of multiple biopsies provides greater assurance in correctly classifying a tumor as more or less hypoxic.
Background and Purpose Gene expression signatures are often used to identify hypoxic tumors. However, intratumoral heterogeneity raises concern that multiple biopsies may be necessary to assess global hypoxia status. The objective of this study was to compare the impact of heterogeneity on the discriminative capacity of several previously described hypoxia gene signatures and determine if a single biopsy is sufficient to obtain a reliable estimate of hypoxia in cervical cancer. Materials and Methods Multiple biopsies (33) were obtained from 11 locally advanced (FIGO IB to IVB) cervical cancers prior to treatment. Ten hypoxia gene signatures were analyzed. Variance component analysis was used to determine the ratio of within-tumor variability to total-tumor variability when one to five biopsies are available for analysis (W/T1–5). The mean standardized error in the signature scores was estimated by comparing the score using one biopsy randomly selected from each tumor to the ‘global’ score using all available biopsies. Results The ten hypoxia signatures were comprised of 6–99 genes each. The W/T1 ratios for individual genes commonly found in the signatures ranged from 0.17 to 0.73. W/T1 ratios for the signatures were generally lower (0.21–0.45), implying greater capacity to discriminate among tumors. With additional biopsies, the signature W/T ratios (ie W/T2-5) decreased further. The mean error in the signature scores varied from 0.27 to 0.40 of one standard deviation, suggesting high capacity to discriminate among tumors with different global hypoxia scores. Conclusions Compared with individual probes, hypoxia gene expression signatures are generally more consistent across multiple biopsies from different regions of a tumor and more tolerant of intratumoral heterogeneity.
Collapse
Affiliation(s)
- Jelena Lukovic
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, Ontario M5G 2M9, Canada.,University of Toronto, Department of Radiation Oncology, 149 College Street, Toronto, Ontario M5T 1P5, Canada
| | - Kathy Han
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, Ontario M5G 2M9, Canada.,University of Toronto, Department of Radiation Oncology, 149 College Street, Toronto, Ontario M5T 1P5, Canada.,University of Toronto, Institute of Medical Sciences, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Melania Pintilie
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, Ontario M5G 2M9, Canada.,Princess Margaret Cancer Centre, Department of Biostatistics, 610 University Ave, Toronto, Ontario M5G 2M9, Canada
| | - Naz Chaudary
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, Ontario M5G 2M9, Canada
| | - Richard P Hill
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, Ontario M5G 2M9, Canada.,University of Toronto, Department of Radiation Oncology, 149 College Street, Toronto, Ontario M5T 1P5, Canada.,University of Toronto, Department of Medical Biophysics, Toronto, Ontario M5G 2M9, Canada
| | - Anthony Fyles
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, Ontario M5G 2M9, Canada.,University of Toronto, Department of Radiation Oncology, 149 College Street, Toronto, Ontario M5T 1P5, Canada.,University of Toronto, Institute of Medical Sciences, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Michael Milosevic
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, Ontario M5G 2M9, Canada.,University of Toronto, Department of Radiation Oncology, 149 College Street, Toronto, Ontario M5T 1P5, Canada.,University of Toronto, Institute of Medical Sciences, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
81
|
Quantitative Analysis of Carbonic Anhydrase IX Uncovers Hypoxia-Related Functional Differences in Classical Hodgkin Lymphoma Subtypes. Int J Mol Sci 2019; 20:ijms20143463. [PMID: 31311071 PMCID: PMC6678552 DOI: 10.3390/ijms20143463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/05/2019] [Accepted: 07/13/2019] [Indexed: 02/07/2023] Open
Abstract
Upregulation of carbonic anhydrase IX (CAIX) was found to be associated with unfavorable prognosis and resistance to treatment in a broad spectrum of malignancies, recently also in classical Hodgkin’s lymphoma (cHL). As demonstrated, variable CAIX expression in a significant number of cHL cases was associated with poor treatment response. The current study focused on the quantification CAIX immunopositivity and its relative expression compared to the total CD30+ neoplastic pool using digital image analysis. One hundred and one lymph node samples featuring cHL histology were analyzed for both CD30 and CAIX by immunohistochemistry. Whole histological slides were scanned and immunopositivity was determined as the histoscore (H-score) using the DensitoQuant software module (3DHistech Kft., Budapest, Hungary). CAIX positivity was observed in the HRS-cells of 56/101 cases (55.44%) and frequently observed in the proximity of necrotic foci. CAIX H-scores were highly variable (range: 2.16–90.36, mean 18.7 ± 18.8). Individual CAIX values were independent of the much higher CD30 values (range 3.46–151.3, mean 52.37 ± 30.74). The CAIX/CD30 index proved to be the highest in the aggressive lymphocyte-depleted (LD) subtype (CAIX/CD30: 0.876). The CAIX expression and the CAIX/CD30 relative index can be precisely determined by image analysis, and values reflect the extent of a tumor mass undergoing hypoxic-stress-related adaptation in the most aggressive forms of cHL.
Collapse
|
82
|
Synthesis and bioevaluation of novel radioiodinated PEG-modified 2-nitroimidazole derivatives for tumor hypoxia imaging. J Radioanal Nucl Chem 2019. [DOI: 10.1007/s10967-019-06649-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
83
|
Hay MP, Shin HN, Wong WW, Sahimi WW, Vaz ATD, Yadav P, Anderson RF, Hicks KO, Wilson WR. Benzotriazine Di-Oxide Prodrugs for Exploiting Hypoxia and Low Extracellular pH in Tumors. Molecules 2019; 24:E2524. [PMID: 31295864 PMCID: PMC6680510 DOI: 10.3390/molecules24142524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 12/31/2022] Open
Abstract
Extracellular acidification is an important feature of tumor microenvironments but has yet to be successfully exploited in cancer therapy. The reversal of the pH gradient across the plasma membrane in cells that regulate intracellular pH (pHi) has potential to drive the selective uptake of weak acids at low extracellular pH (pHe). Here, we investigate the dual targeting of low pHe and hypoxia, another key feature of tumor microenvironments. We prepared eight bioreductive prodrugs based on the benzotriazine di-oxide (BTO) nucleus by appending alkanoic or aminoalkanoic acid sidechains. The BTO acids showed modest selectivity for both low pHe (pH 6.5 versus 7.4, ratios 2 to 5-fold) and anoxia (ratios 2 to 8-fold) in SiHa and FaDu cell cultures. Related neutral BTOs were not selective for acidosis, but had greater cytotoxic potency and hypoxic selectivity than the BTO acids. Investigation of the uptake and metabolism of representative BTO acids confirmed enhanced uptake at low pHe, but lower intracellular concentrations than expected for passive diffusion. Further, the modulation of intracellular reductase activity and competition by the cell-excluded electron acceptor WST-1 suggests that the majority of metabolic reductions of BTO acids occur at the cell surface, compromising the engagement of the resulting free radicals with intracellular targets. Thus, the present study provides support for designing bioreductive prodrugs that exploit pH-dependent partitioning, suggesting, however, that that the approach should be applied to prodrugs with obligate intracellular activation.
Collapse
Affiliation(s)
- Michael P Hay
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Symonds St, Auckland 1142, New Zealand
| | - Hong Nam Shin
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Way Wua Wong
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Wan Wan Sahimi
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Aaron T D Vaz
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Pooja Yadav
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Robert F Anderson
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Symonds St, Auckland 1142, New Zealand
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Kevin O Hicks
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Symonds St, Auckland 1142, New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Symonds St, Auckland 1142, New Zealand.
| |
Collapse
|
84
|
BRCA1 regulates the cancer stem cell fate of breast cancer cells in the context of hypoxia and histone deacetylase inhibitors. Sci Rep 2019; 9:9702. [PMID: 31273285 PMCID: PMC6609720 DOI: 10.1038/s41598-019-46210-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 06/21/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer cell stemness is essential for enabling malignant progression and clonal evolution. Cancer cell fate is likely determined by complex mechanisms involving both cell-intrinsic pathways and stress signals from tumor microenvironment. In this study, we examined the role of the tumor suppressor BRCA1 and hypoxia in the regulation of cancer cell stemness using genetically matched breast cancer cell lines. We have found that BRCA1, a multifunctional protein involved in DNA repair and epigenetic regulation, plays a critical role in the regulation of cancer stem cell (CSC)-like characteristics. Reconstitution of BRCA1 resulted in significant decrease of the CSC-like populations in breast cancer cells whereas down-regulation of BRCA1 resulted in significant increase of the CSC-like populations. Furthermore, the BRCA1-reconstituted tumor cells are more sensitive to the histone deacetylase (HDAC) inhibitor-induced loss of stemness than the BRCA1-deficient cells are. Surprisingly, hypoxia preferentially blocks HDAC inhibitor-induced differentiation of the BRCA1-reconstituted breast cancer cells. In light of the increasing numbers of clinical trials involving HDAC inhibitors in human cancers, our observations strongly suggest that the BRCA1 status and tumor hypoxia should be considered as potentially important clinical parameters that may affect the therapeutic efficacy of HDAC inhibitors.
Collapse
|
85
|
Park S, Safi R, Liu X, Baldi R, Liu W, Liu J, Locasale JW, Chang CY, McDonnell DP. Inhibition of ERRα Prevents Mitochondrial Pyruvate Uptake Exposing NADPH-Generating Pathways as Targetable Vulnerabilities in Breast Cancer. Cell Rep 2019; 27:3587-3601.e4. [PMID: 31216477 PMCID: PMC6604861 DOI: 10.1016/j.celrep.2019.05.066] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 04/03/2019] [Accepted: 05/17/2019] [Indexed: 12/13/2022] Open
Abstract
Most cancer cells exhibit metabolic flexibility, enabling them to withstand fluctuations in intratumoral concentrations of glucose (and other nutrients) and changes in oxygen availability. While these adaptive responses make it difficult to achieve clinically useful anti-tumor responses when targeting a single metabolic pathway, they can also serve as targetable metabolic vulnerabilities that can be therapeutically exploited. Previously, we demonstrated that inhibition of estrogen-related receptor alpha (ERRα) significantly disrupts mitochondrial metabolism and that this results in substantial antitumor activity in animal models of breast cancer. Here we show that ERRα inhibition interferes with pyruvate entry into mitochondria by inhibiting the expression of mitochondrial pyruvate carrier 1 (MPC1). This results in a dramatic increase in the reliance of cells on glutamine oxidation and the pentose phosphate pathway to maintain nicotinamide adenine dinucleotide phosphate (NADPH) homeostasis. In this manner, ERRα inhibition increases the efficacy of glutaminase and glucose-6-phosphate dehydrogenase inhibitors, a finding that has clinical significance.
Collapse
Affiliation(s)
- Sunghee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Rachid Safi
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaojing Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert Baldi
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Wen Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
86
|
Lee HH, Dickson BD, Stevenson RJ, Yang S, Tercel M. Optimised synthesis of a nitroCBI hypoxia-activated prodrug with substantial anticancer activity. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
87
|
Li M, Luo Z, Peng Z, Cai K. Cascade-amplification of therapeutic efficacy: An emerging opportunity in cancer treatment. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1555. [PMID: 31016872 DOI: 10.1002/wnan.1555] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/24/2022]
Abstract
Increasing research evidence reveals that cancer is complex disease involving many biological factors, processes and systems, which may severely limit the actual efficacy of conventional monotonic anticancer approaches. To overcome these obstacles in cancer treatment, a new strategy has been proposed by combining multiple synergistic therapeutic modalities accessing different but inherently related targets and acting sequentially. A major benefit of this strategy is that the multi-target mechanism could result in a cascade-amplification effect leading to enhanced anticancer activity. In this review, we provide a critical discussion on the application of cascade-amplification strategy in the treatment of various cancer indications, focusing on the rational combination of therapeutic agents and their mechanisms of action. A concise yet comprehensive analysis on the potential therapeutic benefit of this strategy was also included. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Menghuan Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.,Department of Biotechnology, School of Life Science, Chongqing University, Chongqing, China
| | - Zhong Luo
- Department of Biotechnology, School of Life Science, Chongqing University, Chongqing, China
| | - Zhihong Peng
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
88
|
Jackson RK, Liew LP, Hay MP. Overcoming Radioresistance: Small Molecule Radiosensitisers and Hypoxia-activated Prodrugs. Clin Oncol (R Coll Radiol) 2019; 31:290-302. [PMID: 30853148 DOI: 10.1016/j.clon.2019.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/12/2019] [Indexed: 12/25/2022]
Abstract
The role of hypoxia in radiation resistance is well established and many approaches to overcome hypoxia in tumours have been explored, with variable success. Two small molecule strategies for targeting hypoxia have dominated preclinical and clinical efforts. One approach has been the use of electron-affinic nitroheterocycles as oxygen-mimetic sensitisers. These agents are best exemplified by the 5-nitroimidazole nimorazole, which has limited use in conjunction with radiotherapy in head and neck squamous cell carcinoma. The second approach seeks to leverage tumour hypoxia as a tumour-specific address for hypoxia-activated prodrugs. These prodrugs are selectively activated by reductases under hypoxia to release cytotoxins, which in some instances may diffuse to kill surrounding oxic tumour tissue. A number of these hypoxia-activated prodrugs have been examined in clinical trial and the merits and shortcomings of recent examples are discussed. There has been an evolution from delivering DNA-interactive cytotoxins to molecularly targeted agents. Efforts to implement these strategies clinically continue today, but success has been elusive. Several issues have been identified that compromised these clinical campaigns. A failure to consider the extravascular transport and the micropharmacokinetic properties of the prodrugs has reduced efficacy. One key element for these 'targeted' approaches is the need to co-develop biomarkers to identify appropriate patients. Hypoxia-activated prodrugs require biomarkers for hypoxia, but also for appropriate activating reductases in tumours, as well as markers of intrinsic sensitivity to the released drug. The field is still evolving and changes in radiation delivery and the impact of immune-oncology will provide fertile ground for future innovation.
Collapse
Affiliation(s)
- R K Jackson
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - L P Liew
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - M P Hay
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
89
|
Gonzalez-Avila G, Sommer B, Mendoza-Posada DA, Ramos C, Garcia-Hernandez AA, Falfan-Valencia R. Matrix metalloproteinases participation in the metastatic process and their diagnostic and therapeutic applications in cancer. Crit Rev Oncol Hematol 2019; 137:57-83. [PMID: 31014516 DOI: 10.1016/j.critrevonc.2019.02.010] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/11/2019] [Accepted: 02/24/2019] [Indexed: 12/13/2022] Open
Abstract
Matrix metalloproteinases (MMPs) participate from the initial phases of cancer onset to the settlement of a metastatic niche in a second organ. Their role in cancer progression is related to their involvement in the extracellular matrix (ECM) degradation and in the regulation and processing of adhesion and cytoskeletal proteins, growth factors, chemokines and cytokines. MMPs participation in cancer progression makes them an attractive target for cancer therapy. MMPs have also been used for theranostic purposes in the detection of primary tumor and metastatic tissue in which a particular MMP is overexpressed, to follow up on therapy responses, and in the activation of cancer cytotoxic pro-drugs as part of nano-delivery-systems that increase drug concentration in a specific tumor target. Herein, we review MMPs molecular characteristics, their synthesis regulation and enzymatic activity, their participation in the metastatic process, and how their functions have been used to improve cancer treatment.
Collapse
Affiliation(s)
- Georgina Gonzalez-Avila
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico.
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | | | - Carlos Ramos
- Laboratorio de Biología Celular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - A Armando Garcia-Hernandez
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - Ramces Falfan-Valencia
- Laboratorio de HLA, Departamento de Inmunogenética y Alergia, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| |
Collapse
|
90
|
Song KX, Liu S, Zhang MZ, Liang WZ, Liu H, Dong XH, Wang YB, Wang XJ. Hyperbaric oxygen therapy improves the effect of keloid surgery and radiotherapy by reducing the recurrence rate. J Zhejiang Univ Sci B 2019; 19:853-862. [PMID: 30387335 DOI: 10.1631/jzus.b1800132] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Keloids are exuberant cutaneous scars that form due to abnormal growth of fibrous tissue following an injury. The primary aim of this study was to assess the efficacy and mechanism of hyperbaric oxygen therapy (HBOT) to reduce the keloid recurrence rate after surgical excision and radiotherapy. METHODS (1) A total of 240 patients were randomly divided into two groups. Patients in the HBOT group (O group) received HBOT after surgical excision and radiotherapy. Patients in the other group were treated with only surgical excision and radiotherapy (K group). (2) Scar tissue from recurrent patients was collected after a second operation. Hematoxylin and eosin (H&E) staining was used to observe keloid morphology. Certain inflammatory factors (interleukin-6 (IL-6), hypoxia-inducible factor-1α (HIF-1α), tumor necrosis factor-α (TNF-α), nuclear factor κB (NF-κB), and vascular endothelial growth factor (VEGF)) were measured using immunohistochemical staining. RESULTS (1) The recurrence rate of the O group (5.97%) was significantly lower than that of the K group (14.15%), P<0.05. Moreover, patients in the O group reported greater satisfaction than those in the K group (P<0.05). (2) Compared with the recurrent scar tissue of the K group, the expression levels of the inflammatory factors were lower in the recurrent scar tissue of the O group. CONCLUSIONS Adjunctive HBOT effectively reduces the keloid recurrence rate after surgical excision and radiotherapy by improving the oxygen level of the tissue and alleviating the inflammatory process.
Collapse
Affiliation(s)
- Ke-Xin Song
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - Shu Liu
- School of Medicine, North China University of Science and Technology, Tangshan 063000, China
| | - Ming-Zi Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - Wei-Zhong Liang
- Department of Plastic Surgery, China Meitan General Hospital, Beijing 100028, China
| | - Hao Liu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - Xin-Hang Dong
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - You-Bin Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| | - Xiao-Jun Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, China
| |
Collapse
|
91
|
Liew LP, Singleton DC, Wong WW, Cheng GJ, Jamieson SMF, Hay MP. Hypoxia‐Activated Prodrugs of PERK Inhibitors. Chem Asian J 2019; 14:1238-1248. [DOI: 10.1002/asia.201801826] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/04/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Lydia P. Liew
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of Auckland Symonds St Auckland 1010 New Zealand
| | - Dean C. Singleton
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of Auckland Symonds St Auckland 1010 New Zealand
| | - Way W. Wong
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
| | - Gary J. Cheng
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
| | - Stephen M. F. Jamieson
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of Auckland Symonds St Auckland 1010 New Zealand
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
| | - Michael P. Hay
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of Auckland Symonds St Auckland 1010 New Zealand
| |
Collapse
|
92
|
Bhandari V, Hoey C, Liu LY, Lalonde E, Ray J, Livingstone J, Lesurf R, Shiah YJ, Vujcic T, Huang X, Espiritu SMG, Heisler LE, Yousif F, Huang V, Yamaguchi TN, Yao CQ, Sabelnykova VY, Fraser M, Chua MLK, van der Kwast T, Liu SK, Boutros PC, Bristow RG. Molecular landmarks of tumor hypoxia across cancer types. Nat Genet 2019; 51:308-318. [PMID: 30643250 DOI: 10.1038/s41588-018-0318-2] [Citation(s) in RCA: 407] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 11/20/2018] [Indexed: 12/27/2022]
Abstract
Many primary-tumor subregions have low levels of molecular oxygen, termed hypoxia. Hypoxic tumors are at elevated risk for local failure and distant metastasis, but the molecular hallmarks of tumor hypoxia remain poorly defined. To fill this gap, we quantified hypoxia in 8,006 tumors across 19 tumor types. In ten tumor types, hypoxia was associated with elevated genomic instability. In all 19 tumor types, hypoxic tumors exhibited characteristic driver-mutation signatures. We observed widespread hypoxia-associated dysregulation of microRNAs (miRNAs) across cancers and functionally validated miR-133a-3p as a hypoxia-modulated miRNA. In localized prostate cancer, hypoxia was associated with elevated rates of chromothripsis, allelic loss of PTEN and shorter telomeres. These associations are particularly enriched in polyclonal tumors, representing a constellation of features resembling tumor nimbosus, an aggressive cellular phenotype. Overall, this work establishes that tumor hypoxia may drive aggressive molecular features across cancers and shape the clinical trajectory of individual tumors.
Collapse
Affiliation(s)
- Vinayak Bhandari
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Christianne Hoey
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Lydia Y Liu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Emilie Lalonde
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jessica Ray
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Julie Livingstone
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Robert Lesurf
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Yu-Jia Shiah
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Tina Vujcic
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Xiaoyong Huang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Shadrielle M G Espiritu
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Lawrence E Heisler
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Fouad Yousif
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Vincent Huang
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Takafumi N Yamaguchi
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Cindy Q Yao
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Veronica Y Sabelnykova
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Michael Fraser
- Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Melvin L K Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | - Stanley K Liu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. .,Informatics and Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada. .,Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA. .,Department of Urology, University of California, Los Angeles, Los Angeles, CA, USA. .,Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, CA, USA. .,Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Robert G Bristow
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada. .,Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada. .,Division of Cancer Sciences, Faculty of Biology, Health and Medicine, University of Manchester, Manchester, UK. .,The Christie NHS Foundation Trust, Manchester, UK. .,CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK.
| |
Collapse
|
93
|
Al-Qassab Y, Grassilli S, Brugnoli F, Vezzali F, Capitani S, Bertagnolo V. Protective role of all-trans retinoic acid (ATRA) against hypoxia-induced malignant potential of non-invasive breast tumor derived cells. BMC Cancer 2018; 18:1194. [PMID: 30497437 PMCID: PMC6267073 DOI: 10.1186/s12885-018-5038-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/04/2018] [Indexed: 12/14/2022] Open
Abstract
Background The presence of hypoxic areas is common in all breast lesions but no data clearly correlate low oxygenation with the acquisition of malignant features by non-invasive cells, particularly by cells from ductal carcinoma in situ (DCIS), the most frequently diagnosed tumor in women. Methods By using a DCIS-derived cell line, we evaluated the effects of low oxygen availability on malignant features of non-invasive breast tumor cells and the possible role of all-trans retinoic acid (ATRA), a well-known anti-leukemic drug, in counteracting the effects of hypoxia. The involvement of the β2 isoform of PI-PLC (PLC-β2), an ATRA target in myeloid leukemia cells, was also investigated by specific modulation of the protein expression. Results We demonstrated that moderate hypoxia is sufficient to induce, in DCIS-derived cells, motility, epithelial-to-mesenchymal transition (EMT) and expression of the stem cell marker CD133, indicative of their increased malignant potential. Administration of ATRA supports the epithelial-like phenotype of DCIS-derived cells cultured under hypoxia and keeps down the number of CD133 positive cells, abrogating almost completely the effects of poor oxygenation. We also found that the mechanisms triggered by ATRA in non-invasive breast tumor cells cultured under hypoxia is in part mediated by PLC-β2, responsible to counteract the effects of low oxygen availability on CD133 levels. Conclusions Overall, we assigned to hypoxia a role in increasing the malignant potential of DCIS-derived cells and we identified in ATRA, currently used in treatment of acute promyelocytic leukemia (APL), an agonist potentially useful in preventing malignant progression of non-invasive breast lesions showing hypoxic areas.
Collapse
Affiliation(s)
- Yasamin Al-Qassab
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy.,College of Medicine, Department of Anatomy, University of Baghdad, Baghdad, Iraq
| | - Silvia Grassilli
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy
| | - Federica Brugnoli
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy
| | - Federica Vezzali
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy
| | - Silvano Capitani
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy.,LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Valeria Bertagnolo
- Signal Transduction Unit, Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44121, Ferrara, Italy.
| |
Collapse
|
94
|
Graham K, Unger E. Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment. Int J Nanomedicine 2018; 13:6049-6058. [PMID: 30323592 PMCID: PMC6177375 DOI: 10.2147/ijn.s140462] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypoxia exists to some degree in most solid tumors due to inadequate oxygen delivery of the abnormal vasculature which cannot meet the demands of the rapidly proliferating cancer cells. The levels of oxygenation within the same tumor are highly variable from one area to another and can change over time. Tumor hypoxia is an important impediment to effective cancer therapy. In radiotherapy, the primary mechanism is the creation of reactive oxygen species; hypoxic tumors are therefore radiation resistant. A number of chemotherapeutic drugs have been shown to be less effective when exposed to a hypoxic environment which can lead to further disease progression. Hypoxia is also a potent barrier to effective immunotherapy in cancer treatment. Because of the recognition of hypoxia as an important barrier to cancer treatment, a variety of approaches have been undertaken to overcome or reverse tumor hypoxia. Such approaches have included breathing hyperbaric oxygen, artificial hemoglobins, allosteric hemoglobin modifiers, hypoxia activated prodrugs and fluorocarbons (FCs). These approaches have largely failed due to limited efficacy and/or adverse side effects. Oxygen therapeutics, based on liquid FCs, can potentially increase the oxygen-carrying capacity of the blood to reverse tumor hypoxia. Currently, at least two drugs are in clinical trials to reverse tumor hypoxia; one of these is designed to improve permeability of oxygen into the tumor tissue and the other is based upon a low boiling point FC that transports higher amounts of oxygen per gram than previously tested FCs.
Collapse
|
95
|
DNA Repair Deficient Chinese Hamster Ovary Cells Exhibiting Differential Sensitivity to Charged Particle Radiation under Aerobic and Hypoxic Conditions. Int J Mol Sci 2018; 19:ijms19082228. [PMID: 30061540 PMCID: PMC6121575 DOI: 10.3390/ijms19082228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 12/31/2022] Open
Abstract
It has been well established that hypoxia significantly increases both cellular and tumor resistance to ionizing radiation. Hypoxia associated radiation resistance has been known for some time but there has been limited success in sensitizing cells to radiation under hypoxic conditions. These studies show that, when irradiated with low linear energy transfer (LET) gamma-rays, poly (ADP-ribose), polymerase (PARP), Fanconi Anemia (FANC), and mutant Chinese Hamster Ovary (CHO) cells respond similarly to the non-homologous end joining (NHEJ) and the homologous recombination (HR) repair mutant CHO cells. Comparable results were observed in cells exposed to 13 keV/μm carbon ions. However, when irradiated with higher LET spread out Bragg peak (SOBP) carbon ions, we observed a decrease in the oxygen enhancement ratio (OER) in all the DNA of repair mutant cell lines. Interestingly, PARP mutant cells were observed as having the largest decrease in OER. Finally, these studies show a significant increase in the relative biological effectiveness (RBE) of high LET SOBP carbon and iron ions in HR and PARP mutants. There was also an increase in the RBE of NHEJ mutants when irradiated to SOBP carbon and iron ions. However, this increase was lower than in other mutant cell lines. These findings indicate that high LET radiation produces unique types of DNA damage under hypoxic conditions and PARP and HR repair pathways play a role in repairing this damage.
Collapse
|
96
|
Hauge A, Wegner CS, Gaustad JV, Simonsen TG, Andersen LMK, Rofstad EK. Diffusion-Weighted MRI Is Insensitive to Changes in the Tumor Microenvironment Induced by Antiangiogenic Therapy. Transl Oncol 2018; 11:1128-1136. [PMID: 30036782 PMCID: PMC6072800 DOI: 10.1016/j.tranon.2018.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/03/2018] [Accepted: 07/06/2018] [Indexed: 12/29/2022] Open
Abstract
Antiangiogenic treatment (AAT) used in combination with radiation therapy or chemotherapy is a promising strategy for the treatment of several cancer diseases. The vascularity and oxygenation of tumors may be changed significantly by AAT, and consequently, a noninvasive method for monitoring AAT-induced changes in these microenvironmental parameters is needed. The purpose of this study was to evaluate the potential usefulness of diffusion-weighted magnetic resonance imaging (DW-MRI). DW-MRI was conducted with a Bruker Biospec 7.05-T scanner using four diffusion weightings and diffusion sensitization gradients in three orthogonal directions. Maps of the apparent diffusion coefficient (ADC) were calculated by using a monoexponential diffusion model. Two cervical carcinoma xenograft models (BK-12, HL-16) were treated with bevacizumab, and two pancreatic carcinoma xenograft models (BxPC-3, Panc-1) were treated with sunitinib. Pimonidazole and CD31 were used as markers of hypoxia and blood vessels, respectively, and fraction of hypoxic tissue (HFPim) and microvascular density (MVD) were quantified by analyzing immunohistochemical preparations. MVD decreased significantly after AAT in BK-12, HL-16, and BxPC-3 tumors, and this decrease was sufficiently large to cause a significant increase in HFPim in BK-12 and BxPC-3 tumors. The ADC maps of treated tumors and untreated control tumors were not significantly different in any of these three tumor models, suggesting that the AAT-induced microenvironmental changes were not detectable by DW-MRI. DW-MRI is insensitive to changes in tumor vascularity and oxygenation induced by bevacizumab or sunitinib treatment.
Collapse
Affiliation(s)
- Anette Hauge
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Catherine S Wegner
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Jon-Vidar Gaustad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Trude G Simonsen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Lise Mari K Andersen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Einar K Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
97
|
Generation of multiparametric MRI maps by using Gd-labelled- RBCs reveals phenotypes and stages of murine prostate cancer. Sci Rep 2018; 8:10567. [PMID: 30002426 PMCID: PMC6043588 DOI: 10.1038/s41598-018-28926-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/26/2018] [Indexed: 11/30/2022] Open
Abstract
Prostate Cancer (PCa) is the second most common and fifth cause of cancer-related mortality in males in Western Countries. The development of innovative tools for an early, more precise and noninvasive diagnosis is a medical need. Vascular volume (Vv) and hypoxia are two of the most important tumor hallmarks. Herein, they have been assessed in TRAMP mice by using MRI. Their quantification has been carried out by injecting autologous Red Blood Cells (RBCs), ex vivo labelled with Gd-HPDO3A or Gd-DOTP complexes, respectively. Gd-labelled-RBCs are stably confined in the intravascular space, also in presence of a very leaky tumor endothelium, thus representing efficient probes for vascular space analysis. Vv enhancement and hypoxia onset have been demonstrated to be present at early stages of PCa and their expression largely increases with tumor development. Moreover, also Diffusion weighted MRI and Amide Proton Transfer MRI have been herein applied to characterize PCa. The herein applied multiparametric MRI (mpMRI) analysis allows a detailed in vivo characterization of PCa, in which each histotype and cancer stage displays a specific MRI pattern. This provides an unprecedented opportunity to feature prostate tumor, making possible a non-invasive, precise and early diagnosis, which could direct treatments towards a more personalized medicine.
Collapse
|
98
|
Sahu A, Choi WI, Tae G. Recent Progress in the Design of Hypoxia-Specific Nano Drug Delivery Systems for Cancer Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| | - Won Il Choi
- Center for Convergence Bioceramic Materials; Convergence R&D Division; Korea Institute of Ceramic Engineering and Technology; 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu Cheongju Chungbuk 28160 Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| |
Collapse
|
99
|
Yang L, Luo D, Yi J, Li L, Zhao Y, Lin M, Guo W, Hu L, Zhou C. Therapy Effects of Advanced Hypopharyngeal and Laryngeal Squamous Cell Carcinoma: Evaluated using Dual-Energy CT Quantitative Parameters. Sci Rep 2018; 8:9064. [PMID: 29899458 PMCID: PMC5998143 DOI: 10.1038/s41598-018-27341-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/16/2018] [Indexed: 12/11/2022] Open
Abstract
The accurate evaluation of the therapeutic effects of advanced laryngeal and hypopharyngeal squamous cell carcinoma (LHSCC) remains challenging. In this study, we determined the value of quantitative parameters derived from dual-energy computed tomography (DECT) for predicting the therapeutic effects of advanced LHSCC and to provide valuable evidence for early judgement of the tumour's response to therapy in clinical practice. We prospectively analysed 41 patients with pathologically confirmed LHSCC. All patients received a DECT scan before therapy. Nineteen of 41 patients showed complete remission (CR), and 22 showed non-complete remission (NCR). The mean of the slope of spectral Hounsfield unit curve (λHU), standardized iodine concentration and effective atomic number in the CR group were significantly lower than the NCR group (P < 0.05). There were no significant differences for T stage, treatment modality and standardized water concentration between two groups (P > 0.05). The best predictor of CR effect was λHU. The 2-year cumulative recurrence rate of patients with higher λHU values was significantly higher than that of patients with lower λHU values (P < 0.05), while the 2-year survival rate of those patients was not significantly different (P > 0.05). DECT could easily identify CR patients and potentially help to choose the appropriate treatment regimen for advanced LHSCC.
Collapse
Affiliation(s)
- Liang Yang
- Department of Diagnostic Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Dehong Luo
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Junlin Yi
- Department of Radiotherapy, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Li
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yanfeng Zhao
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Meng Lin
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wei Guo
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Hu
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunwu Zhou
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| |
Collapse
|
100
|
Ashton TM, McKenna WG, Kunz-Schughart LA, Higgins GS. Oxidative Phosphorylation as an Emerging Target in Cancer Therapy. Clin Cancer Res 2018; 24:2482-2490. [PMID: 29420223 DOI: 10.1158/1078-0432.ccr-17-3070] [Citation(s) in RCA: 682] [Impact Index Per Article: 97.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/07/2018] [Accepted: 01/30/2018] [Indexed: 11/16/2022]
Abstract
Cancer cells have upregulated glycolysis compared with normal cells, which has led many to the assumption that oxidative phosphorylation (OXPHOS) is downregulated in all cancers. However, recent studies have shown that OXPHOS can be also upregulated in certain cancers, including leukemias, lymphomas, pancreatic ductal adenocarcinoma, high OXPHOS subtype melanoma, and endometrial carcinoma, and that this can occur even in the face of active glycolysis. OXPHOS inhibitors could therefore be used to target cancer subtypes in which OXPHOS is upregulated and to alleviate therapeutically adverse tumor hypoxia. Several drugs including metformin, atovaquone, and arsenic trioxide are used clinically for non-oncologic indications, but emerging data demonstrate their potential use as OXPHOS inhibitors. We highlight novel applications of OXPHOS inhibitors with a suitable therapeutic index to target cancer cell metabolism. Clin Cancer Res; 24(11); 2482-90. ©2018 AACR.
Collapse
Affiliation(s)
- Thomas M Ashton
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, Oxford, United Kingdom
| | - W Gillies McKenna
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, Oxford, United Kingdom
| | - Leoni A Kunz-Schughart
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, Oxford, United Kingdom.
- OncoRay, National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Germany
- National Center for Tumor Diseases (NCT), partner site Dresden, Germany
| | - Geoff S Higgins
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, Oxford, United Kingdom.
| |
Collapse
|