1
|
Cimmino A, Fasciglione GF, Gioia M, Marini S, Ciaccio C. Multi-Anticancer Activities of Phytoestrogens in Human Osteosarcoma. Int J Mol Sci 2023; 24:13344. [PMID: 37686148 PMCID: PMC10487502 DOI: 10.3390/ijms241713344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Phytoestrogens are plant-derived bioactive compounds with estrogen-like properties. Their potential health benefits, especially in cancer prevention and treatment, have been a subject of considerable research in the past decade. Phytoestrogens exert their effects, at least in part, through interactions with estrogen receptors (ERs), mimicking or inhibiting the actions of natural estrogens. Recently, there has been growing interest in exploring the impact of phytoestrogens on osteosarcoma (OS), a type of bone malignancy that primarily affects children and young adults and is currently presenting limited treatment options. Considering the critical role of the estrogen/ERs axis in bone development and growth, the modulation of ERs has emerged as a highly promising approach in the treatment of OS. This review provides an extensive overview of current literature on the effects of phytoestrogens on human OS models. It delves into the multiple mechanisms through which these molecules regulate the cell cycle, apoptosis, and key pathways implicated in the growth and progression of OS, including ER signaling. Moreover, potential interactions between phytoestrogens and conventional chemotherapy agents commonly used in OS treatment will be examined. Understanding the impact of these compounds in OS holds great promise for developing novel therapeutic approaches that can augment current OS treatment modalities.
Collapse
Affiliation(s)
| | | | | | | | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, I-00133 Rome, Italy; (A.C.); (G.F.F.); (M.G.); (S.M.)
| |
Collapse
|
2
|
Tobeiha M, Rajabi A, Raisi A, Mohajeri M, Yazdi SM, Davoodvandi A, Aslanbeigi F, Vaziri M, Hamblin MR, Mirzaei H. Potential of natural products in osteosarcoma treatment: Focus on molecular mechanisms. Biomed Pharmacother 2021; 144:112257. [PMID: 34688081 DOI: 10.1016/j.biopha.2021.112257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most frequent type of bone cancer found in children and adolescents, and commonly arises in the metaphyseal region of tubular long bones. Standard therapeutic approaches, such as surgery, chemotherapy, and radiation therapy, are used in the management of osteosarcoma. In recent years, the mortality rate of osteosarcoma has decreased due to advances in treatment methods. Today, the scientific community is investigating the use of different naturally derived active principles against various types of cancer. Natural bioactive compounds can function against cancer cells in two ways. Firstly they can act as classical cytotoxic compounds by non-specifically affecting macromolecules, such as DNA, enzymes, and microtubules, which are also expressed in normal proliferating cells, but to a greater extent by cancer cells. Secondly, they can act against oncogenic signal transduction pathways, many of which are activated in cancer cells. Some bioactive plant-derived agents are gaining increasing attention because of their anti-cancer properties. Moreover, some naturally-derived compounds can significantly promote the effectiveness of standard chemotherapy drugs, and in certain cases are able to ameliorate drug-induced adverse effects caused by chemotherapy. In the present review we summarize the effects of various naturally-occurring bioactive compounds against osteosarcoma.
Collapse
Affiliation(s)
- Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahshad Mohajeri
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Amirhossein Davoodvandi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Aslanbeigi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - MohamadSadegh Vaziri
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
3
|
Kim I, Kim M, Park MK, Naik R, Park JH, Kim BK, Choi Y, Chang KY, Won M, Ban HS, Lee K. The disubstituted adamantyl derivative LW1564 inhibits the growth of cancer cells by targeting mitochondrial respiration and reducing hypoxia-inducible factor (HIF)-1α accumulation. Exp Mol Med 2020; 52:1845-1856. [PMID: 33235318 PMCID: PMC8080809 DOI: 10.1038/s12276-020-00523-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Targeting cancer metabolism has emerged as an important cancer therapeutic strategy. Here, we describe the synthesis and biological evaluation of a novel class of hypoxia-inducible factor (HIF)-1α inhibitors, disubstituted adamantyl derivatives. One such compound, LW1564, significantly suppressed HIF-1α accumulation and inhibited the growth of various cancer cell lines, including HepG2, A549, and HCT116. Measurements of the oxygen consumption rate (OCR) and ATP production rate revealed that LW1564 suppressed mitochondrial respiration, thereby increasing the intracellular oxygen concentration to stimulate HIF-1α degradation. LW1564 also significantly decreased overall ATP levels by inhibiting mitochondrial electron transport chain (ETC) complex I and downregulated mammalian target of rapamycin (mTOR) signaling by increasing the AMP/ATP ratio, which increased AMP-activated protein kinase (AMPK) phosphorylation. Consequently, LW1564 promoted the phosphorylation of acetyl-CoA carboxylase, which inhibited lipid synthesis. In addition, LW1564 significantly inhibited tumor growth in a HepG2 mouse xenograft model. Taken together, the results indicate that LW1564 inhibits the growth of cancer cells by targeting mitochondrial ETC complex I and impairing cancer cell metabolism. We, therefore, suggest that LW1564 may be a potent therapeutic agent for a subset of cancers that rely on oxidative phosphorylation for ATP generation. A drug that curbs the accumulation of a critical protein involved in the oxygen-sensing machinery of cells could offer a potent new therapeutic for treating cancer. Inhyub Kim, University of Science and Technology, Daejeon, South Korea, and colleagues describe a compound called LW1564 that suppresses metabolism within mitochondria, the energy factories of the cell. Less energy production means less oxygen consumption and therefore oxygen molecules build up inside the cell, which in turn stimulates the degradation of HIF-1α, a master regulator of oxygen balance. Many tumors rely on HIF-1α for their aberrant biological characteristics, and without this protein they tend to show reduced growth. The authors demonstrated that LW1564 could limit HIF-1α accumulation and inhibit the proliferation of various cancer cell lines. The drug also inhibited tumor growth in a mouse model of liver cancer.
Collapse
Affiliation(s)
- Inhyub Kim
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, 34141, Korea
| | - Minkyoung Kim
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Korea
| | - Min Kyung Park
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Korea.,College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Korea
| | - Ravi Naik
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Korea
| | - Jae Hyung Park
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Korea
| | - Bo-Kyung Kim
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Korea
| | - Yongseok Choi
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Korea
| | | | - Misun Won
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Korea. .,Department of Functional Genomics, University of Science and Technology, Daejeon, 34141, Korea.
| | - Hyun Seung Ban
- Biotherapeutics Translational Research Center, KRIBB, Daejeon, 34141, Korea.
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Korea.
| |
Collapse
|
4
|
Li T, Mao C, Wang X, Shi Y, Tao Y. Epigenetic crosstalk between hypoxia and tumor driven by HIF regulation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:224. [PMID: 33109235 PMCID: PMC7592369 DOI: 10.1186/s13046-020-01733-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Hypoxia is the major influence factor in physiological and pathological courses which are mainly mediated by hypoxia-inducible factors (HIFs) in response to low oxygen tensions within solid tumors. Under normoxia, HIF signaling pathway is inhibited due to HIF-α subunits degradation. However, in hypoxic conditions, HIF-α is activated and stabilized, and HIF target genes are successively activated, resulting in a series of tumour-specific activities. The activation of HIFs, including HIF-1α, HIF-2α and HIF-3α, subsequently induce downstream target genes which leads to series of responses, the resulting abnormal processes or metabolites in turn affect HIFs stability. Given its functions in tumors progression, HIFs have been regarded as therapeutic targets for improved treatment efficacy. Epigenetics refers to alterations in gene expression that are stable between cell divisions, and sometimes between generations, but do not involve changes in the underlying DNA sequence of the organism. And with the development of research, epigenetic regulation has been found to play an important role in the development of tumors, which providing accumulating basic or clinical evidences for tumor treatments. Here, given how little has been reported about the overall association between hypoxic tumors and epigenetics, we made a more systematic review from epigenetic perspective in hope of helping others better understand hypoxia or HIF pathway, and providing more established and potential therapeutic strategies in tumors to facilitate epigenetic studies of tumors.
Collapse
Affiliation(s)
- Tiansheng Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chao Mao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ying Shi
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
5
|
Abstract
Hypoxia-inducible factors (HIFs) control transcriptional responses to reduced O2 availability. HIFs are heterodimeric proteins composed of an O2-regulated HIF-α subunit and a constitutively expressed HIF-1β subunit. HIF-α subunits are subject to prolyl hydroxylation, which targets the proteins for degradation under normoxic conditions. Small molecule prolyl hydroxylase inhibitors, which stabilize the HIF-α subunits and increase HIF-dependent expression of erythropoietin, are in phase III clinical trials for the treatment of anemia in patients with chronic kidney disease. HIFs contribute to the pathogenesis of many cancers, particularly the clear cell type of renal cell carcinoma in which loss of function of the von Hippel-Lindau tumor suppressor blocks HIF-2α degradation. A small molecule inhibitor that binds to HIF-2α and blocks dimerization with HIF-1β is in clinical trials for the treatment of renal cell carcinoma. Targeting HIFs for stabilization or inhibition may improve outcomes in diseases that are common causes of mortality in the US population.
Collapse
Affiliation(s)
- Gregg L Semenza
- Institute for Cell Engineering, McKusick-Nathans Institute of Genetic Medicine, and Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| |
Collapse
|
6
|
Shen H, Cook K, Gee HE, Hau E. Hypoxia, metabolism, and the circadian clock: new links to overcome radiation resistance in high-grade gliomas. J Exp Clin Cancer Res 2020; 39:129. [PMID: 32631383 PMCID: PMC7339573 DOI: 10.1186/s13046-020-01639-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is the cornerstone of treatment of high-grade gliomas (HGGs). It eradicates tumor cells by inducing oxidative stress and subsequent DNA damage. Unfortunately, almost all HGGs recur locally within several months secondary to radioresistance with intricate molecular mechanisms. Therefore, unravelling specific underlying mechanisms of radioresistance is critical to elucidating novel strategies to improve the radiosensitivity of tumor cells, and enhance the efficacy of radiotherapy. This review addresses our current understanding of how hypoxia and the hypoxia-inducible factor 1 (HIF-1) signaling pathway have a profound impact on the response of HGGs to radiotherapy. In addition, intriguing links between hypoxic signaling, circadian rhythms and cell metabolism have been recently discovered, which may provide insights into our fundamental understanding of radioresistance. Cellular pathways involved in the hypoxic response, DNA repair and metabolism can fluctuate over 24-h periods due to circadian regulation. These oscillatory patterns may have consequences for tumor radioresistance. Timing radiotherapy for specific times of the day (chronoradiotherapy) could be beneficial in patients with HGGs and will be discussed.
Collapse
Affiliation(s)
- Han Shen
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, Westmead, New South Wales, 2145, Australia.
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia.
| | - Kristina Cook
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health & Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Harriet E Gee
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, Westmead, New South Wales, 2145, Australia
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
- Department of Radiation Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, New South Wales, Australia
| | - Eric Hau
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, Westmead, New South Wales, 2145, Australia
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
- Department of Radiation Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, New South Wales, Australia
- Blacktown Hematology and Cancer Centre, Blacktown Hospital, Blacktown, New South Wales, Australia
| |
Collapse
|
7
|
Micheliolide Enhances Radiosensitivities of p53-Deficient Non-Small-Cell Lung Cancer via Promoting HIF-1α Degradation. Int J Mol Sci 2020; 21:ijms21093392. [PMID: 32403326 PMCID: PMC7247679 DOI: 10.3390/ijms21093392] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 02/08/2023] Open
Abstract
Micheliolide (MCL) has shown promising anti-inflammatory and anti-tumor efficacy. However, whether and how MCL enhances the sensitivity of non-small-cell lung cancer (NSCLC) to radiotherapy are still unknown. In the present paper, we found that MCL exerted a tumor cell killing effect on NSCLC cells in a dose-dependent manner, and MCL strongly sensitized p53-deficient NSCLC cells, but not the cells with wild-type p53 to irradiation (IR). Meanwhile, MCL markedly inhibited the expression of hypoxia-inducible factor-1α (HIF-1α) after IR and hypoxic exposure in H1299 and Calu-1 cells rather than in H460 cells. Consistently, radiation- or hypoxia-induced expression of vascular endothelial growth factor (VEGF) was also significantly inhibited by MCL in H1299 and Calu-1 cells, but not in H460 cells. Therefore, inhibition of the HIF-1α pathway might, at least in part, contribute to the radiosensitizing effect of MCL. Further study showed that MCL could accelerate the degradation of HIF-1α through the ubiquitin-proteosome system. In addition, the transfection of wild-type p53 into p53-null cells (H1299) attenuated the effect of MCL on inhibiting HIF-1α expression. These results suggest MCL effectively sensitizes p53-deficient NSCLC cells to IR in a manner of inhibiting the HIF-1α pathway via promoting HIF-1α degradation, and p53 played a negative role in MCL-induced HIF-1α degradation.
Collapse
|
8
|
Yao Y, Feng L, Wang Z, Chen H, Tan N. Programmed delivery of cyclopeptide RA-V and antisense oligonucleotides for combination therapy on hypoxic tumors and for therapeutic self-monitoring. Biomater Sci 2020; 8:256-265. [PMID: 31687671 DOI: 10.1039/c9bm00905a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chemotherapy is a dominant treatment modality for different types and stages of cancer. However, hypoxia is one of the undesirable limitations of chemotherapy, which reduces the therapeutic efficiency in cancer treatment, ultimately leading to failure of the treatment. Herein, an ideal chemosensitization system capable of attenuating the tumor hypoxia microenvironment and enhancing chemotherapy effects in tumors was designed. This system (designated as the RA/RX Liposome) uses for the first time a pH-sensitive liposome to co-deliver cyclopeptide RA-V as chemotherapeutic drugs and antisense oligonucleotides as HIF-1α inhibitors (RX-0047) for attenuating tumor hypoxia, as well as a caspase-8 activation probe for therapeutic self-monitoring. After modification with death receptor 5-specific antibodies (anti-DR5) on the surface of the liposome, the RA/RX Liposome can successfully deliver components targeting colon tumors in vivo. This work should synergistically enhance the therapeutic effects of the treatment by successfully down-regulating HIF-1α expression against tumor hypoxia during the RA-V-induced apoptotic process. More importantly, the RA/RX Liposome can be precisely applied for therapeutic self-monitoring with the light-up fluorescence of the caspase-8 probe.
Collapse
Affiliation(s)
- Yongrong Yao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | | | | | | | | |
Collapse
|
9
|
Thomas LW, Stephen JM, Esposito C, Hoer S, Antrobus R, Ahmed A, Al-Habib H, Ashcroft M. CHCHD4 confers metabolic vulnerabilities to tumour cells through its control of the mitochondrial respiratory chain. Cancer Metab 2019; 7:2. [PMID: 30886710 PMCID: PMC6404347 DOI: 10.1186/s40170-019-0194-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/05/2019] [Indexed: 12/15/2022] Open
Abstract
Background Tumour cells rely on glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) to survive. Thus, mitochondrial OXPHOS has become an increasingly attractive area for therapeutic exploitation in cancer. However, mitochondria are required for intracellular oxygenation and normal physiological processes, and it remains unclear which mitochondrial molecular mechanisms might provide therapeutic benefit. Previously, we discovered that coiled-coil-helix-coiled-coil-helix domain-containing protein 4 (CHCHD4) is critical for regulating intracellular oxygenation and required for the cellular response to hypoxia (low oxygenation) in tumour cells through molecular mechanisms that we do not yet fully understand. Overexpression of CHCHD4 in human cancers correlates with increased tumour progression and poor patient survival. Results Here, we show that elevated CHCHD4 expression provides a proliferative and metabolic advantage to tumour cells in normoxia and hypoxia. Using stable isotope labelling with amino acids in cell culture (SILAC) and analysis of the whole mitochondrial proteome, we show that CHCHD4 dynamically affects the expression of a broad range of mitochondrial respiratory chain subunits from complex I-V, including multiple subunits of complex I (CI) required for complex assembly that are essential for cell survival. We found that loss of CHCHD4 protects tumour cells from respiratory chain inhibition at CI, while elevated CHCHD4 expression in tumour cells leads to significantly increased sensitivity to CI inhibition, in part through the production of mitochondrial reactive oxygen species (ROS). Conclusions Our study highlights an important role for CHCHD4 in regulating tumour cell metabolism and reveals that CHCHD4 confers metabolic vulnerabilities to tumour cells through its control of the mitochondrial respiratory chain and CI biology.
Collapse
Affiliation(s)
- Luke W. Thomas
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
| | - Jenna M. Stephen
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
| | - Cinzia Esposito
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
- Present address: Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Simon Hoer
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY UK
| | - Afshan Ahmed
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
- Present address: AstraZeneca Ltd., Cambridge, UK
| | - Hasan Al-Habib
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
| | - Margaret Ashcroft
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH UK
| |
Collapse
|
10
|
Baker LCJ, Sikka A, Price JM, Boult JKR, Lepicard EY, Box G, Jamin Y, Spinks TJ, Kramer-Marek G, Leach MO, Eccles SA, Box C, Robinson SP. Evaluating Imaging Biomarkers of Acquired Resistance to Targeted EGFR Therapy in Xenograft Models of Human Head and Neck Squamous Cell Carcinoma. Front Oncol 2018; 8:271. [PMID: 30083516 PMCID: PMC6064942 DOI: 10.3389/fonc.2018.00271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/02/2018] [Indexed: 01/18/2023] Open
Abstract
Background: Overexpression of EGFR is a negative prognostic factor in head and neck squamous cell carcinoma (HNSCC). Patients with HNSCC who respond to EGFR-targeted tyrosine kinase inhibitors (TKIs) eventually develop acquired resistance. Strategies to identify HNSCC patients likely to benefit from EGFR-targeted therapies, together with biomarkers of treatment response, would have clinical value. Methods: Functional MRI and 18F-FDG PET were used to visualize and quantify imaging biomarkers associated with drug response within size-matched EGFR TKI-resistant CAL 27 (CALR) and sensitive (CALS) HNSCC xenografts in vivo, and pathological correlates sought. Results: Intrinsic susceptibility, oxygen-enhanced and dynamic contrast-enhanced MRI revealed significantly slower baseline R 2 ∗ , lower hyperoxia-induced Δ R 2 ∗ and volume transfer constant Ktrans in the CALR tumors which were associated with significantly lower Hoechst 33342 uptake and greater pimonidazole-adduct formation. There was no difference in oxygen-induced ΔR1 or water diffusivity between the CALR and CALS xenografts. PET revealed significantly higher relative uptake of 18F-FDG in the CALR cohort, which was associated with significantly greater Glut-1 expression. Conclusions: CALR xenografts established from HNSCC cells resistant to EGFR TKIs are more hypoxic, poorly perfused and glycolytic than sensitive CALS tumors. MRI combined with PET can be used to non-invasively assess HNSCC response/resistance to EGFR inhibition.
Collapse
Affiliation(s)
- Lauren C. J. Baker
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Arti Sikka
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Jonathan M. Price
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Jessica K. R. Boult
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Elise Y. Lepicard
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Gary Box
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Yann Jamin
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Terry J. Spinks
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Gabriela Kramer-Marek
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Martin O. Leach
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Suzanne A. Eccles
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Carol Box
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Simon P. Robinson
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
11
|
Frontiñán-Rubio J, Santiago-Mora RM, Nieva-Velasco CM, Ferrín G, Martínez-González A, Gómez MV, Moreno M, Ariza J, Lozano E, Arjona-Gutiérrez J, Gil-Agudo A, De la Mata M, Pesic M, Peinado JR, Villalba JM, Pérez-Romasanta L, Pérez-García VM, Alcaín FJ, Durán-Prado M. Regulation of the oxidative balance with coenzyme Q10 sensitizes human glioblastoma cells to radiation and temozolomide. Radiother Oncol 2018; 128:236-244. [PMID: 29784452 DOI: 10.1016/j.radonc.2018.04.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 04/05/2018] [Accepted: 04/29/2018] [Indexed: 10/16/2022]
Abstract
OBJECTIVES To investigate how the modulation of the oxidative balance affects cytotoxic therapies in glioblastoma, in vitro. MATERIAL AND METHODS Human glioblastoma U251 and T98 cells and normal astrocytes C8D1A were loaded with coenzyme Q10 (CoQ). Mitochondrial superoxide ion (O2-) and H2O2 were measured by fluorescence microscopy. OXPHOS performance was assessed in U251 cells with an oxytherm Clark-type electrode. Radio- and chemotherapy cytotoxicity was assessed by immunostaining of γH2AX (24 h), annexin V and nuclei morphology, at short (72 h) and long (15 d) time. Hif-1α, SOD1, SOD2 and NQO1 were determined by immunolabeling. Catalase activity was measured by classic enzymatic assay. Glutathione levels and total antioxidant capacity were quantified using commercial kits. RESULTS CoQ did not affect oxygen consumption but reduced the level of O2- and H2O2 while shifted to a pro-oxidant cell status mainly due to a decrease in catalase activity and SOD2 level. Hif-1α was dampened, echoed by a decrease lactate and several key metabolites involved in glutathione synthesis. CoQ-treated cells were twofold more sensitive than control to radiation-induced DNA damage and apoptosis in short and long-term clonogenic assays, potentiating TMZ-induced cytotoxicity, without affecting non-transformed astrocytes. CONCLUSIONS CoQ acts as sensitizer for cytotoxic therapies, disarming GBM cells, but not normal astrocytes, against further pro-oxidant injuries, being potentially useful in clinical practice for this fatal pathology.
Collapse
Affiliation(s)
- Javier Frontiñán-Rubio
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - Raquel María Santiago-Mora
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - Consuelo María Nieva-Velasco
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - Gustavo Ferrín
- Instituto Maimónides de Investigación Biomédica en Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Córdoba, Spain
| | | | - María Victoria Gómez
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies and Instituto Regional de Investigación Científica Aplicada, University of Castilla-La Mancha, Ciudad Real, Spain
| | - María Moreno
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies and Instituto Regional de Investigación Científica Aplicada, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Julia Ariza
- Department of Cell Biology, Physiology and Immunology, Faculty of Sciences, University of Córdoba, Agrifood Campus of International Excellence ceiA3, Córdoba, Spain
| | - Eva Lozano
- Radiotherapy Unit, University Hospital of Ciudad Real, Spain
| | | | | | - Manuel De la Mata
- Instituto Maimónides de Investigación Biomédica en Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Córdoba, Spain
| | - Milica Pesic
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Serbia
| | - Juan Ramón Peinado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - José M Villalba
- Department of Cell Biology, Physiology and Immunology, Faculty of Sciences, University of Córdoba, Agrifood Campus of International Excellence ceiA3, Córdoba, Spain
| | | | - Víctor M Pérez-García
- Laboratory of Mathematical Oncology, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Francisco J Alcaín
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain
| | - Mario Durán-Prado
- Department of Medical Sciences, Faculty of Medicine, University of Castilla-la Mancha, Ciudad Real, Spain; Oxidative Stress and Neurodegeneration Group, Regional Centre for Biomedical Research, Ciudad Real, Spain.
| |
Collapse
|
12
|
The novel hypoxia-inducible factor-1α inhibitor IDF-11774 regulates cancer metabolism, thereby suppressing tumor growth. Cell Death Dis 2017; 8:e2843. [PMID: 28569777 PMCID: PMC5520894 DOI: 10.1038/cddis.2017.235] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/15/2017] [Accepted: 04/20/2017] [Indexed: 12/13/2022]
Abstract
HIF-1 is associated with poor prognoses and therapeutic resistance in cancer patients. We previously developed a novel hypoxia-inducible factor (HIF)-1 inhibitor, IDF-11774, a clinical candidate for cancer therapy. We also reported that IDF-1174 inhibited HSP70 chaperone activity and suppressed accumulation of HIF-1α. In this study, IDF-11774 inhibited the accumulation of HIF-1α in vitro and in vivo in colorectal carcinoma HCT116 cells under hypoxic conditions. Moreover, IDF-11774 treatment suppressed angiogenesis of cancer cells by reducing the expression of HIF-1 target genes, reduced glucose uptake, thereby sensitizing cells to growth under low glucose conditions, and decreased the extracellular acidification rate (ECAR) and oxygen consumption rate of cancer cells. Metabolic profiling of IDF-11774-treated cells revealed low levels of NAD+, NADP+, and lactate, as well as of intermediates in glycolysis and the tricarboxylic acid cycle. In addition, we observed elevated AMP and diminished ATP levels, resulting in a high AMP/ATP ratio. The level of AMP-activated protein kinase phosphorylation also increased, leading to inhibition of mTOR signaling in treated cells. In vivo xenograft assays demonstrated that IDF-11774 exhibited substantial anticancer efficacy in mouse models containing KRAS, PTEN, or VHL mutations, which often occur in malignant cancers. Collectively, our data indicate that IDF-11774 suppressed hypoxia-induced HIF-1α accumulation and repressed tumor growth by targeting energy production-related cancer metabolism.
Collapse
|
13
|
Farquhar MJ, Humphreys IS, Rudge SA, Wilson GK, Bhattacharya B, Ciaccia M, Hu K, Zhang Q, Mailly L, Reynolds GM, Ashcroft M, Balfe P, Baumert TF, Roessler S, Wakelam MJO, McKeating JA. Autotaxin-lysophosphatidic acid receptor signalling regulates hepatitis C virus replication. J Hepatol 2017; 66:919-929. [PMID: 28126468 DOI: 10.1016/j.jhep.2017.01.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/09/2016] [Accepted: 01/08/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Chronic hepatitis C is a global health problem with an estimated 170 million hepatitis C virus (HCV) infected individuals at risk of progressive liver disease and hepatocellular carcinoma (HCC). Autotaxin (ATX, gene name: ENPP2) is a phospholipase with diverse roles in the physiological and pathological processes including inflammation and oncogenesis. Clinical studies have reported increased ATX expression in chronic hepatitis C, however, the pathways regulating ATX and its role in the viral life cycle are not well understood. METHODS In vitro hepatocyte and ex vivo liver culture systems along with chimeric humanized liver mice and HCC tissue enabled us to assess the interplay between ATX and the HCV life cycle. RESULTS HCV infection increased hepatocellular ATX RNA and protein expression. HCV infection stabilizes hypoxia inducible factors (HIFs) and we investigated a role for these transcription factors to regulate ATX. In vitro studies show that low oxygen increases hepatocellular ATX expression and transcriptome analysis showed a positive correlation between ATX mRNA levels and hypoxia gene score in HCC tumour tissue associated with HCV and other aetiologies. Importantly, inhibiting ATX-lysophosphatidic acid (LPA) signalling reduced HCV replication, demonstrating a positive role for this phospholipase in the viral life cycle. LPA activates phosphoinositide-3-kinase that stabilizes HIF-1α and inhibiting the HIF signalling pathway abrogates the pro-viral activity of LPA. CONCLUSIONS Our data support a model where HCV infection increases ATX expression which supports viral replication and HCC progression. LAY SUMMARY Chronic hepatitis C is a global health problem with infected individuals at risk of developing liver disease that can progress to hepatocellular carcinoma. Autotaxin generates the biologically active lipid lysophosphatidic acid that has been reported to play a tumorigenic role in a wide number of cancers. In this study we show that hepatitis C virus infection increases autotaxin expression via hypoxia inducible transcription factor and provides an environment in the liver that promotes fibrosis and liver injury. Importantly, we show a new role for lysophosphatidic acid in positively regulating hepatitis C virus replication.
Collapse
Affiliation(s)
- Michelle J Farquhar
- Viral Hepatitis Laboratory, Centre for Human Virology, University of Birmingham, UK
| | - Isla S Humphreys
- Viral Hepatitis Laboratory, Centre for Human Virology, University of Birmingham, UK
| | | | - Garrick K Wilson
- Viral Hepatitis Laboratory, Centre for Human Virology, University of Birmingham, UK
| | | | | | - Ke Hu
- Viral Hepatitis Laboratory, Centre for Human Virology, University of Birmingham, UK
| | | | - Laurent Mailly
- INSERM U1110, University of Strasbourg, 3 Rue Koeberlé, F-67000 Strasbourg, France
| | - Gary M Reynolds
- NIHR Liver Biomedical Research Unit, University of Birmingham, Birmingham, UK
| | | | - Peter Balfe
- Viral Hepatitis Laboratory, Centre for Human Virology, University of Birmingham, UK
| | - Thomas F Baumert
- INSERM U1110, University of Strasbourg, 3 Rue Koeberlé, F-67000 Strasbourg, France
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Jane A McKeating
- Viral Hepatitis Laboratory, Centre for Human Virology, University of Birmingham, UK.
| |
Collapse
|
14
|
Burrell JS, Walker-Samuel S, Boult JK, Baker LC, Jamin Y, Halliday J, Waterton JC, Robinson SP. Investigating the Vascular Phenotype of Subcutaneously and Orthotopically Propagated PC3 Prostate Cancer Xenografts Using Combined Carbogen Ultrasmall Superparamagnetic Iron Oxide MRI. Top Magn Reson Imaging 2016; 25:237-243. [PMID: 27748709 PMCID: PMC5068556 DOI: 10.1097/rmr.0000000000000102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The aim of this study was to use the combined carbogen-ultrasmall superparamagnetic iron oxide (CUSPIO) magnetic resonance imaging (MRI) method, which uses spatial correlations in independent susceptibility imaging biomarkers, to investigate and compare the impact of tumor size and anatomical site on vascular structure and function in vivo. Mice bearing either subcutaneous or orthotopic PC3 LN3 prostate tumors were imaged at 7 T, using a multi-gradient echo sequence to quantify R2, before and during carbogen (95% O2/5% CO2) breathing, and subsequently following intravenous administration of USPIO particles. Carbogen and USPIO-induced changes in R2 were used to inform on hemodynamic vasculature and fractional blood volume (%), respectively. The CUSPIO imaging data were also segmented to identify and assess five categories of R2 response. Small and large subcutaneous and orthotopic tumor cohorts all exhibited significantly (P < 0.05) different median baseline R2, ΔR2carbogen, and fractional blood volume. CUSPIO imaging showed that small subcutaneous tumors predominantly exhibited a negative ΔR2carbogen followed by a positive ΔR2USPIO, consistent with a well perfused tumor vasculature. Large subcutaneous tumors exhibited a small positive ΔR2carbogen and relatively low fractional blood volume, suggesting less functional vasculature. Orthotopic tumors revealed a large, positive ΔR2carbogen, consistent with vascular steal, and which may indicate that vascular function is more dependent on site of implantation than tumor size. Regions exhibiting significant ΔR2carbogen, but no significant ΔR2USPIO, suggesting transient vascular shutdown over the experimental timecourse, were apparent in all 3 cohorts. CUSPIO imaging can inform on efficient drug delivery via functional vasculature in vivo, and on appropriate tumor model selection for pre-clinical therapy trials.
Collapse
Affiliation(s)
- Jake S. Burrell
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Surrey
| | - Simon Walker-Samuel
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Surrey
- Centre for Advanced Biomedical Imaging, Department of Medicine and Institute of Child Health, University College London, London
| | - Jessica K.R. Boult
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Surrey
| | - Lauren C.J. Baker
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Surrey
| | - Yann Jamin
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Surrey
| | - Jane Halliday
- R&D Personalised Healthcare & Biomarkers, AstraZeneca, Alderley Park, Macclesfield, UK
| | - John C. Waterton
- R&D Personalised Healthcare & Biomarkers, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Simon P. Robinson
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, Surrey
| |
Collapse
|
15
|
Baker LCJ, Boult JKR, Thomas M, Koehler A, Nayak T, Tessier J, Ooi CH, Birzele F, Belousov A, Zajac M, Horn C, LeFave C, Robinson SP. Acute tumour response to a bispecific Ang-2-VEGF-A antibody: insights from multiparametric MRI and gene expression profiling. Br J Cancer 2016; 115:691-702. [PMID: 27529514 PMCID: PMC5023775 DOI: 10.1038/bjc.2016.236] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/03/2016] [Accepted: 07/06/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND To assess antivascular effects, and evaluate clinically translatable magnetic resonance imaging (MRI) biomarkers of tumour response in vivo, following treatment with vanucizumab, a bispecific human antibody against angiopoietin-2 (Ang-2) and vascular endothelial growth factor-A (VEGF-A). METHODS Colo205 colon cancer xenografts were imaged before and 5 days after treatment with a single 10 mg kg(-1) dose of either vanucizumab, bevacizumab (anti-human VEGF-A), LC06 (anti-murine/human Ang-2) or omalizumab (anti-human IgE control). Volumetric response was assessed using T2-weighted MRI, and diffusion-weighted, dynamic contrast-enhanced (DCE) and susceptibility contrast MRI used to quantify tumour water diffusivity (apparent diffusion coefficient (ADC), × 10(6) mm(2) s(-1)), vascular perfusion/permeability (K(trans), min(-1)) and fractional blood volume (fBV, %) respectively. Pathological correlates were sought, and preliminary gene expression profiling performed. RESULTS Treatment with vanucizumab, bevacizumab or LC06 induced a significant (P<0.01) cytolentic response compared with control. There was no significant change in tumour ADC in any treatment group. Uptake of Gd-DTPA was restricted to the tumour periphery in all post-treatment groups. A significant reduction in tumour K(trans) (P<0.05) and fBV (P<0.01) was determined 5 days after treatment with vanucizumab only. This was associated with a significant (P<0.05) reduction in Hoechst 33342 uptake compared with control. Gene expression profiling identified 20 human genes exclusively regulated by vanucizumab, 6 of which are known to be involved in vasculogenesis and angiogenesis. CONCLUSIONS Vanucizumab is a promising antitumour and antiangiogenic treatment, whose antivascular activity can be monitored using DCE and susceptibility contrast MRI. Differential gene expression in vanucizumab-treated tumours is regulated by the combined effect of Ang-2 and VEGF-A inhibition.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/diagnostic imaging
- Adenocarcinoma/drug therapy
- Adenocarcinoma/pathology
- Angiogenesis Inhibitors/immunology
- Angiogenesis Inhibitors/therapeutic use
- Angiopoietin-2/antagonists & inhibitors
- Angiopoietin-2/immunology
- Animals
- Antibodies, Bispecific/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Bevacizumab/therapeutic use
- Cell Line, Tumor
- Colonic Neoplasms/blood supply
- Colonic Neoplasms/diagnostic imaging
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/pathology
- DNA Replication/drug effects
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immunoglobulin E/immunology
- Magnetic Resonance Imaging/methods
- Mice
- Molecular Targeted Therapy
- Neovascularization, Pathologic/diagnostic imaging
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Omalizumab/therapeutic use
- Tumor Burden
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lauren CJ Baker
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Jessica KR Boult
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Markus Thomas
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg DE-82377, Germany
| | - Astrid Koehler
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg DE-82377, Germany
| | - Tapan Nayak
- Roche pRED, Roche Innovation Center, Basel CH-4070, Switzerland
| | - Jean Tessier
- Roche pRED, Roche Innovation Center, Basel CH-4070, Switzerland
| | - Chia-Huey Ooi
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg DE-82377, Germany
| | - Fabian Birzele
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg DE-82377, Germany
| | - Anton Belousov
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Penzberg DE-82377, Germany
| | | | - Carsten Horn
- Roche pRED, Roche Innovation Center, Basel CH-4070, Switzerland
| | - Clare LeFave
- Roche pRED, Roche Innovation Center, New York, NY 10016, USA
| | - Simon P Robinson
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|
16
|
Zhao CB, Shi L, Pu HH, Zhang QY. The Promoting Effect of Radiation on Glucose Metabolism in Breast Cancer Cells under the Treatment of Cobalt Chloride. Pathol Oncol Res 2016; 23:47-53. [DOI: 10.1007/s12253-016-0076-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 05/16/2016] [Indexed: 01/18/2023]
|
17
|
Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat Rev Drug Discov 2016; 15:639-660. [PMID: 27256476 DOI: 10.1038/nrd.2016.75] [Citation(s) in RCA: 500] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adipose tissue regulates numerous physiological processes, and its dysfunction in obese humans is associated with disrupted metabolic homeostasis, insulin resistance and type 2 diabetes mellitus (T2DM). Although several US-approved treatments for obesity and T2DM exist, these are limited by adverse effects and a lack of effective long-term glucose control. In this Review, we provide an overview of the role of adipose tissue in metabolic homeostasis and assess emerging novel therapeutic strategies targeting adipose tissue, including adipokine-based strategies, promotion of white adipose tissue beiging as well as reduction of inflammation and fibrosis.
Collapse
Affiliation(s)
- Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center
| | - Perry E Bickel
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center
| |
Collapse
|
18
|
Subhani S, Vavilala DT, Mukherji M. HIF inhibitors for ischemic retinopathies and cancers: options beyond anti-VEGF therapies. Angiogenesis 2016; 19:257-73. [DOI: 10.1007/s10456-016-9510-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/16/2016] [Indexed: 12/15/2022]
|
19
|
Abstract
The added value of diffusion-weighted magnetic resonance imaging (DW-MRI) for the detection, localization, and staging of primary prostate cancer has been extensively reported in original studies and meta-analyses. More recently, DW-MRI and related techniques have been used to noninvasively assess prostate cancer aggressiveness and estimate its biological behavior. The present article aims to summarize the potential applications of DW-MRI for noninvasive optimization of pretherapeutic risk assessment, patient management decisions, and evaluation of treatment response.
Collapse
|
20
|
Ban HS, Uto Y, Won M, Nakamura H. Hypoxia-inducible factor (HIF) inhibitors: a patent survey (2011-2015). Expert Opin Ther Pat 2016; 26:309-22. [DOI: 10.1517/13543776.2016.1146252] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
21
|
Li D, Wang X, Wang S, Cheng J. Correlation between BOLD-MRI and HIF expression level in renal carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:13759-13763. [PMID: 26722605 PMCID: PMC4680550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/28/2015] [Indexed: 06/05/2023]
Abstract
Occupying about 2%~3% of all malignant tumors, renal carcinoma is the most common primary cancer in kidney. The oxidative level of tumor cells is of vital role for optimizing treatment plan, evaluating efficacy and predicting prognosis. This study thus investigated the R2(*) value in mouse renal carcinoma model and the correlation between tumor hypoxia and expression level of hypoxia inducible factor-1 (HIF-1). A total of 20 BALB/C nude mice (4~6 weeks old) were inoculated with human ACHN renal carcinoma cells to generate renal cancer model. After the tumor diameter reached 0.5 cm, all animals were examined by BOLD-MRI, both under normal inhalation (R2a(*)) and carbogen treatment (R2b(*)). The alternation of R2(*) values (ΔR2(*)=R2a(*) - R2b(*)) was calculated. Mice were then sacrificed for Immunohistochemical (IHC) staining targeting HIF-1α and HIF-2α. The positive score of HIF was then analyzed for its correlation with R2(*) value. In 18 mice finished both experiments, Pearson correlation analysis revealed significant negative correlation between R2a(*) and ΔR2(*) (r=-0.48, P<0.05) and positive relationship between ΔR2(*) and HIF-2α (r=0.38, P<0.05). HIF-1α level, however, did not correlated with tumor R(*) values. The positive correlation between ΔR2(*) and HIF-2α, but not HIF-1α, suggested potential role of combined BOLD-MRI technique and HIF-1α staining in clinical diagnosis of renal carcinoma. HIF-2α may work as biological marker for renal cancer.
Collapse
Affiliation(s)
- Dong Li
- Department of Radiology, Liyang People’s HospitalLiyang 213300, Jiangsu, China
| | - Xingming Wang
- Department of Radiology, Liyang People’s HospitalLiyang 213300, Jiangsu, China
| | - Shuai Wang
- Department of Radiology, Liyang People’s HospitalLiyang 213300, Jiangsu, China
| | - Jie Cheng
- Department of Radiology, the First People’s Hospital of ChangzhouChangzhou 213000, Jiangsu, China
| |
Collapse
|
22
|
Papaevangelou E, Almeida GS, Jamin Y, Robinson SP, deSouza NM. Diffusion-weighted MRI for imaging cell death after cytotoxic or apoptosis-inducing therapy. Br J Cancer 2015; 112:1471-9. [PMID: 25880014 PMCID: PMC4453679 DOI: 10.1038/bjc.2015.134] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/10/2015] [Accepted: 03/17/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Non-invasive serial imaging is desirable to detect processes such as necrotic and apoptotic cell death in cancer patients undergoing treatment. This study investigated the use of diffusion-weighted (DW-) magnetic resonance imaging (MRI) for imaging cell death induced by either a cytotoxic drug (irinotecan), or the apoptosis-inducing agent birinapant, in human tumour xenografts in vivo. METHODS Nude mice bearing human SW620 colon carcinoma xenografts were treated with vehicle, irinotecan (50 mg kg(-1)) or birinapant (30 mg kg(-1)) for up to 5 days. DW-MRI was performed prior to and on days 1, 3 and 5 during treatment. Assessment of tumour apoptosis and necrosis ex vivo was used to validate the imaging findings. RESULTS Both irinotecan and birinapant induced significant tumour growth delay. Irinotecan induced a small increase in the tumour apparent diffusion coefficient (ADC) after 1 day, with a 20 and 30% increase at days 3 and 5 respectively. ADC was unchanged in the vehicle- and birinapant-treated tumours despite a growth delay in the latter. Histological analysis showed that irinotecan increased necrosis at days 3 and 5, and induced apoptosis after 1 day, compared with vehicle. Birinapant induced apoptosis after day 3, but had no effect on tumour necrosis. CONCLUSIONS Tumour ADC changes after irinotecan treatment were associated with the induction of a mixture of necrotic and apoptotic cell death, whereas induction of apoptosis alone with birinapant was not sufficient to induce changes in tissue microstructure that were detectable with DW-MRI. ADC is a useful non-invasive biomarker for early detection of response to cytotoxic drugs, but false negatives may arise while detecting apoptotic response to birinapant.
Collapse
Affiliation(s)
- E Papaevangelou
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - G S Almeida
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - Y Jamin
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - S P Robinson
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - N M deSouza
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| |
Collapse
|
23
|
Madhu B, Dadulescu M, Griffiths J. Artefacts in 1H NMR-based metabolomic studies on cell cultures. MAGMA (NEW YORK, N.Y.) 2015; 28:161-71. [PMID: 25108704 DOI: 10.1007/s10334-014-0458-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/17/2014] [Accepted: 07/22/2014] [Indexed: 10/24/2022]
Abstract
OBJECT Metabolomic studies on cultured cells involve assays of cell extracts and culture medium, both of which are often performed by (1)H NMR. Cell culture is nowadays performed in plastic dishes or flasks, and the extraction of metabolites from the cells is typically performed with perchloric acid, methanol-chloroform, or acetonitrile, ideally while the cells are still adherent to the culture dish. We conducted this investigation to identify contaminants from cell culture plasticware in metabolomic studies. MATERIALS AND METHODS Human diploid fibroblasts (IMR90) (n = 6), HeLa cells (n = 6), and transformed astrocytes with HIF-1 knockout (Astro-KO) (n = 6) were cultured. Cells were seeded in 100 mm Petri dishes with 10 ml complete growth medium (Dulbecco's minimum essential medium) containing 10 % foetal bovine serum (FBS). Cell cultures were incubated at 37 °C in 5 % CO2 for approximately 3 days. Metabolites were extracted by use of a perchloric acid procedure. (1)H NMR spectroscopy was used for metabolite analysis. "Null sample" (i.e. cell-free) experiments were performed by either rinsing dishes with medium or incubating the medium in Petri dishes from five different manufacturers for 72 h and then by performing a dummy "extraction" of each Petri dish by the perchloric acid, methanol-chloroform, or acetonitrile procedures. Principal components analysis was used for classification of samples and to determine the contaminants arising from plasticware. RESULTS We found that even brief rinsing of cell culture plasticware with culture medium elutes artefactual chemicals, the (1)H NMR signals of which could confound assays of acetate, succinate, and glycolate. Incubation of culture medium in cell-culture dishes for 72 h (as in a typical cell-culture experiment) followed by perchloric extraction in the dishes enhanced elution of the artefacts. These artefacts were present, but somewhat less pronounced, in the (1)H NMR spectra of null samples extracted with methanol and acetonitrile. Ethanol, lactate, alanine, fructose, and fumarate signals that appear in the (1)H NMR spectrum of the unused (pure) medium originate from FBS. CONCLUSIONS Plastic Petri dishes from five different manufacturers gave rise to essentially identical artefactual peaks. Use of a pH indicator to assist neutralisation introduced still more artefactual signals in the aromatic region, as well as methanol and ethanol signals. Methanol and acetonitrile extracts also contained artefacts arising from the plasticware, although the amounts were less than in the perchloric acid extracts. Finally, we provide suggestions for minimizing these artefacts. The best practice would be to run a "null" extraction with every batch of cellular metabolomics experiments to test for contamination and to provide a "background" spectrum.
Collapse
Affiliation(s)
- Basetti Madhu
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK,
| | | | | |
Collapse
|
24
|
Liu MT, Chen MK, Huang CC, Huang CY. Prognostic Value of Molecular Markers and Implication for Molecular Targeted Therapies in Nasopharyngeal Carcinoma: An Update in an Era of New Targeted Molecules Development. World J Oncol 2015; 6:243-261. [PMID: 29147412 PMCID: PMC5649942 DOI: 10.14740/wjon610w] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2012] [Indexed: 12/15/2022] Open
Abstract
The aim of the study was to evaluate the prognostic significance of molecular biomarkers which could provide information for more accurate prognostication and development of novel therapeutic strategies for nasopharyngeal carcinoma (NPC). NPC is a unique malignant epithelial carcinoma of head and neck region, with an intimate association with the Epstein-Barr virus (EBV). Currently, the prediction of NPC prognosis is mainly based on the clinical TNM staging; however, NPC patients with the same clinical stage often present different clinical outcomes, suggesting that the TNM stage is insufficient to precisely predict the prognosis of this disease. In this review, we give an overview of the prognostic value of molecular markers in NPC and discuss potential strategies of targeted therapies for treatment of NPC. Molecular biomarkers, which play roles in abnormal proliferation signaling pathways (such as Wnt/β-catenin pathway), intracellular mitogenic signal aberration (such as hypoxia-inducible factor (HIF)-1α), receptor-mediated aberrations (such as vascular endothelial growth factor (VEGF)), tumor suppressors (such as p16 and p27 activity), cell cycle aberrations (such as cyclin D1 and cyclin E), cell adhesion aberrations (such as E-cadherin), apoptosis dysregualtion (such as survivin) and centromere aberration (centromere protein H), are prognostic markers for NPC. Plasma EBV DNA concentrations and EBV-encoded latent membrane proteins are also prognostic markers for NPC. Implication of molecular targeted therapies in NPC was discussed. Such therapies could have potential in combination with different cytotoxic agents to combat and eradicate tumor cells. In order to further improve overall survival for patients with loco-regionally advanced NPC, the development of innovative strategies, including prognostic molecular markers and molecular targeted agents is needed.
Collapse
Affiliation(s)
- Mu-Tai Liu
- Department of Radiation Oncology, Changhua Christian Hospital, 135 Nan Shiau Street, Changhua, Taiwan 500, ROC.,Department of Oncology, National Taiwan University Hospital, 7 Chung San South Road, Taipei, Taiwan 100, ROC.,Department of Medicine, Chang Shan Medical University, 110 Section 1, Chien- Kuo N. Road, Taichung, Taiwan 402, ROC.,Department of Radiology, Yuanpei University of Science and Technology, 306 Yuanpei Street, Hsinchu, Taiwan 300, ROC
| | - Mu-Kuan Chen
- Department of Radiology, Yuanpei University of Science and Technology, 306 Yuanpei Street, Hsinchu, Taiwan 300, ROC.,Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, 135 Nan Shiau Street, Changhua, Taiwan 500, ROC
| | - Chia-Chun Huang
- Department of Radiation Oncology, Changhua Christian Hospital, 135 Nan Shiau Street, Changhua, Taiwan 500, ROC
| | - Chao-Yuan Huang
- Department of Oncology, National Taiwan University Hospital, 7 Chung San South Road, Taipei, Taiwan 100, ROC
| |
Collapse
|
25
|
Pettersen EO, Ebbesen P, Gieling RG, Williams KJ, Dubois L, Lambin P, Ward C, Meehan J, Kunkler IH, Langdon SP, Ree AH, Flatmark K, Lyng H, Calzada MJ, Peso LD, Landazuri MO, Görlach A, Flamm H, Kieninger J, Urban G, Weltin A, Singleton DC, Haider S, Buffa FM, Harris AL, Scozzafava A, Supuran CT, Moser I, Jobst G, Busk M, Toustrup K, Overgaard J, Alsner J, Pouyssegur J, Chiche J, Mazure N, Marchiq I, Parks S, Ahmed A, Ashcroft M, Pastorekova S, Cao Y, Rouschop KM, Wouters BG, Koritzinsky M, Mujcic H, Cojocari D. Targeting tumour hypoxia to prevent cancer metastasis. From biology, biosensing and technology to drug development: the METOXIA consortium. J Enzyme Inhib Med Chem 2014; 30:689-721. [PMID: 25347767 DOI: 10.3109/14756366.2014.966704] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/15/2014] [Indexed: 01/06/2023] Open
Abstract
The hypoxic areas of solid cancers represent a negative prognostic factor irrespective of which treatment modality is chosen for the patient. Still, after almost 80 years of focus on the problems created by hypoxia in solid tumours, we still largely lack methods to deal efficiently with these treatment-resistant cells. The consequences of this lack may be serious for many patients: Not only is there a negative correlation between the hypoxic fraction in tumours and the outcome of radiotherapy as well as many types of chemotherapy, a correlation has been shown between the hypoxic fraction in tumours and cancer metastasis. Thus, on a fundamental basis the great variety of problems related to hypoxia in cancer treatment has to do with the broad range of functions oxygen (and lack of oxygen) have in cells and tissues. Therefore, activation-deactivation of oxygen-regulated cascades related to metabolism or external signalling are important areas for the identification of mechanisms as potential targets for hypoxia-specific treatment. Also the chemistry related to reactive oxygen radicals (ROS) and the biological handling of ROS are part of the problem complex. The problem is further complicated by the great variety in oxygen concentrations found in tissues. For tumour hypoxia to be used as a marker for individualisation of treatment there is a need for non-invasive methods to measure oxygen routinely in patient tumours. A large-scale collaborative EU-financed project 2009-2014 denoted METOXIA has studied all the mentioned aspects of hypoxia with the aim of selecting potential targets for new hypoxia-specific therapy and develop the first stage of tests for this therapy. A new non-invasive PET-imaging method based on the 2-nitroimidazole [(18)F]-HX4 was found to be promising in a clinical trial on NSCLC patients. New preclinical models for testing of the metastatic potential of cells were developed, both in vitro (2D as well as 3D models) and in mice (orthotopic grafting). Low density quantitative real-time polymerase chain reaction (qPCR)-based assays were developed measuring multiple hypoxia-responsive markers in parallel to identify tumour hypoxia-related patterns of gene expression. As possible targets for new therapy two main regulatory cascades were prioritised: The hypoxia-inducible-factor (HIF)-regulated cascades operating at moderate to weak hypoxia (<1% O(2)), and the unfolded protein response (UPR) activated by endoplasmatic reticulum (ER) stress and operating at more severe hypoxia (<0.2%). The prioritised targets were the HIF-regulated proteins carbonic anhydrase IX (CAIX), the lactate transporter MCT4 and the PERK/eIF2α/ATF4-arm of the UPR. The METOXIA project has developed patented compounds targeting CAIX with a preclinical documented effect. Since hypoxia-specific treatments alone are not curative they will have to be combined with traditional anti-cancer therapy to eradicate the aerobic cancer cell population as well.
Collapse
|
26
|
Zhu J, Huang H, Dong S, Ge L, Zhang Y. Progress in aptamer-mediated drug delivery vehicles for cancer targeting and its implications in addressing chemotherapeutic challenges. Theranostics 2014; 4:931-44. [PMID: 25057317 PMCID: PMC4107293 DOI: 10.7150/thno.9663] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/23/2014] [Indexed: 12/28/2022] Open
Abstract
Aptamers are novel oligonucleotides with flexible three-dimensional configurations that recognize and bind to their cognate targets, including tumor surface receptors, in a high-affinity and highly specific manner. Because of their unique intrinsic properties, a variety of aptamer-mediated nanovehicles have been developed to directionally transport anti-cancer drugs to tumor sites to minimize systemic cytotoxicity and to enhance permeation by these tumoricidal agents. Despite advances in the selection and synthesis of aptamers and in the conjugation and self-assembly of nanotechnologies, current chemotherapy and drug delivery systems face great challenges. These challenges are due to the limitations of aptamers and vehicles and because of complicated tumor mechanisms, including heterogeneity, anti-cancer drug resistance, and hypoxia-induced aberrances. In this review, we will summarize current approaches utilizing tumor surface hallmarks and aptamers and their roles and mechanisms in therapeutic nanovehicles targeting tumors. Delivery forms include nanoparticles, nanotubes, nanogels, aptamer-drug conjugates, and novel molecular trains. Moreover, the obstacles posed by the aforementioned issues will be highlighted, and possible solutions will be acknowledged. Furthermore, future perspectives will be presented, including cutting-edge integration with RNA interference nanotechnology and personalized chemotherapy, which will facilitate innovative approaches to aptamer-based therapeutics.
Collapse
|
27
|
Unwith S, Zhao H, Hennah L, Ma D. The potential role of HIF on tumour progression and dissemination. Int J Cancer 2014; 136:2491-503. [PMID: 24729302 DOI: 10.1002/ijc.28889] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 03/07/2014] [Accepted: 04/03/2014] [Indexed: 12/20/2022]
Abstract
Cancer is the second cause of mortality worldwide, primarily owing to failure to cure metastatic disease. The need to target the metastatic process to reduce mortality is clear and research over the past decade has shown hypoxia-inducible factor-1 (HIF-1) to be one of the promising targets. In order for metastatic disease to be established, multiple steps need to be taken whereby the tumour cells escape into the bloodstream and survive, disseminate and then establish at a premetastatic niche. HIF-1 mediates hypoxia-induced proangiogenic factors such as vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF), which promote extravasation and chemotaxis. The migration of tumour cells is mediated by loss of E-cadherin, which results in a more invasive phenotype; dissemination of the tumour cells by increased vascular permeability and survival in the bloodstream through resistance to apoptosis as well as adhesion at the premetastatic niche are all controlled by factors under the influence of HIF-1. The overexpression of HIF in many aggressive cancer types as well as its role in the establishment of metastatic disease and treatment resistance demonstrate its potential target in therapeutics. Taken together, the role of HIF-1 in cancer and metastatic disease is clear and the need for better treatment targeting metastases is paramount; more aggressive phenotypes with less response to treatment are associated with HIF-1 expression. Our research has shown promise but many questions still remain to be answered.
Collapse
Affiliation(s)
- Sandeep Unwith
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and, Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, United Kingdom
| | | | | | | |
Collapse
|
28
|
Li J, Jamin Y, Boult JKR, Cummings C, Waterton JC, Ulloa J, Sinkus R, Bamber JC, Robinson SP. Tumour biomechanical response to the vascular disrupting agent ZD6126 in vivo assessed by magnetic resonance elastography. Br J Cancer 2014; 110:1727-32. [PMID: 24569471 PMCID: PMC3974089 DOI: 10.1038/bjc.2014.76] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/05/2013] [Accepted: 01/21/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Magnetic resonance elastography (MRE) is an emerging imaging technique that affords non-invasive quantitative assessment and visualization of tissue mechanical properties in vivo. METHODS In this study, MRE was used to quantify (kPa) the absolute value of the complex shear modulus |G*|, elasticity Gd and viscosity Gl of SW620 human colorectal cancer xenografts before and 24 h after treatment with either 200 mg kg(-1) of the vascular disrupting agent ZD6126 (N-acetylcolchinol-O-phosphate) or vehicle control, and the data were compared with changes in water diffusivity measured by diffusion-weighted magnetic resonance imaging. RESULTS A heterogeneous distribution of |G*|, Gd and Gl was observed pre-treatment with an intertumoral coefficient of variation of 13% for |G*|. There were no significant changes in the vehicle-treated cohort. In contrast, ZD6126 induced a significant decrease in the tumour-averaged |G*| (P<0.01), Gd (P<0.01) and Gl (P<0.05), and this was associated with histologically confirmed central necrosis. This reduction in tumour viscoelasticity occurred at a time when no significant change in tumour apparent diffusion coefficient (ADC) was observed. CONCLUSIONS These data demonstrate that MRE can provide early imaging biomarkers for treatment-induced tumour necrosis.
Collapse
Affiliation(s)
- J Li
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - Y Jamin
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - J K R Boult
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - C Cummings
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - J C Waterton
- Personalised Healthcare and Biomarkers, AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - J Ulloa
- Personalised Healthcare and Biomarkers, AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - R Sinkus
- BHF Centre of Excellence, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St Thomas' Hospital, London, SE1 7EH, UK
| | - J C Bamber
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - S P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| |
Collapse
|
29
|
Zeng W, Liu P, Pan W, Singh SR, Wei Y. Hypoxia and hypoxia inducible factors in tumor metabolism. Cancer Lett 2014; 356:263-7. [PMID: 24508030 DOI: 10.1016/j.canlet.2014.01.032] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 01/01/2023]
Abstract
Because of the abnormal vasculature, most growing solid tumors contain regions that experience either acute or chronic hypoxia. However, tumor cells can maintain a high glycolytic rate even when there is enough oxygen supply. Hypoxia-inducible factors (HIFs) play crucial role in the response of tumor cells to this distinct microenvironment by shifting energy production from mitochondria towards glycolysis. In this review, we focus on the metabolism of tumor cell survival in hypoxic microenvironments. Furthermore, we also emphasize the mechanisms by which hypoxia and HIFs regulate tumor metabolism.
Collapse
Affiliation(s)
- Wen Zeng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China; Department of Joint and Bone Disease Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Peiyi Liu
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China; Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Weimin Pan
- Department of Human Movement Studies, Xi'an Physical Education University, Xi'an 710068, China
| | - Shree Ram Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, National Institutes of Health, National Cancer Institute, Frederick, MD 21702, USA.
| | - Yiyong Wei
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China; Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China.
| |
Collapse
|
30
|
Case Study: Systems Biology of HIF Metabolism in Cancer. CORRELATION-BASED NETWORK ANALYSIS OF CANCER METABOLISM 2014. [DOI: 10.1007/978-1-4939-0615-4_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
31
|
Lu R, Jiang M, Chen Z, Xu X, Hu H, Zhao X, Gao X, Guo L. Lactate dehydrogenase 5 expression in Non-Hodgkin lymphoma is associated with the induced hypoxia regulated protein and poor prognosis. PLoS One 2013; 8:e74853. [PMID: 24086384 PMCID: PMC3781153 DOI: 10.1371/journal.pone.0074853] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 08/06/2013] [Indexed: 12/31/2022] Open
Abstract
Lactate dehydrogenase 5 (LDH-5) is one of the major isoenzymes catalyzing the biochemical process of pyruvate to lactate. The purpose of this study was to investigate the expression of serum LDH-5 and test whether this enzyme is regulated by tumor hypoxia and represents a prognostic marker in patients with Non-Hodgkin’s lymphoma (NHL). In this study, LDH-5 levels were detected using agarose gel electrophoresis in NHL patients (n = 266) and non-NHL controls including benign lymphadenectasis (n = 30) and healthy cohorts (n = 233). We also explored the expression of LDH-5 and hypoxia-inducible factor (HIF) 1α in NHL and benign controls by immunohistochemistry and immunofluorescence staining, respectively. Moreover, the role of LDH-5 in the progression of NHL was assessed by multivariate Cox analyses and Kaplan-Meier survival estimates. Serum concentrations of LDH-5 were significantly higher in NHL patients (9.3%) than in benign patients and healthy controls (7.5% and 7.2%, respectively, P<0.01). Application of LDH-5 detection increased the sensitivity of NHL detection, identifying 53.4% of NHL patients as positive, compared with the measurement of total LDH levels (36.5% sensitivity). LDH-5 concentrations increased with clinical stage, extra-nodal site involvement, and WHO performance status of patients with NHL. Exposure to a hypoxic environment induced the expression of LDH-5 and its overexpression correlated with HIF1α cytoplasmic accumulation in NHL cells. In multivariate analyses, LDH-5 was an independent marker for progression-free survival in patients with NHL (P<0.001). Overall, the expression of LDH-5 was elevated in NHL, showing an association with tumor hypoxia and unfavorable prognosis. Thus, LDH-5 emerges as a promising prognostic predictor for NHL patients.
Collapse
Affiliation(s)
- Renquan Lu
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Minglei Jiang
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Zhujun Chen
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xiaofeng Xu
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Hongfeng Hu
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xinmin Zhao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xiang Gao
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Guo
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
32
|
Beloueche-Babari M, Jamin Y, Arunan V, Walker-Samuel S, Revill M, Smith PD, Halliday J, Waterton JC, Barjat H, Workman P, Leach MO, Robinson SP. Acute tumour response to the MEK1/2 inhibitor selumetinib (AZD6244, ARRY-142886) evaluated by non-invasive diffusion-weighted MRI. Br J Cancer 2013; 109:1562-9. [PMID: 23942066 PMCID: PMC3776979 DOI: 10.1038/bjc.2013.456] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 07/17/2013] [Accepted: 07/22/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Non-invasive imaging biomarkers underpin the development of molecularly targeted anti-cancer drugs. This study evaluates tumour apparent diffusion coefficient (ADC), measured by diffusion-weighted magnetic resonance imaging (DW-MRI), as a biomarker of response to the MEK1/2 inhibitor selumetinib (AZD6244, ARRY-142886) in human tumour xenografts. METHODS Nude mice bearing human BRAF(V600D) WM266.4 melanoma or BRAF(V600E) Colo205 colon carcinoma xenografts were treated for 4 days with vehicle or selumetinib. DW-MRI was performed before and 2 h after the last dose and excised tumours analysed for levels of phospho-ERK1/2, cleaved caspase 3 (CC3) and necrosis. RESULTS Selumetinib treatment induced tumour stasis and reduced ERK1/2 phosphorylation in both WM266.4 and Colo205 tumour xenografts. Relative to day 0, mean tumour ADC was unchanged in the control groups but was significantly increased by up to 1.6-fold in selumetinib-treated WM266.4 and Colo205 tumours. Histological analysis revealed a significant increase in necrosis in selumetinib-treated WM266.4 and Colo205 xenografts and CC3 staining in selumetinib-treated Colo205 tumours relative to controls. CONCLUSION Changes in ADC following treatment with the MEK1/2 inhibitor selumetinib in responsive human tumour xenografts were concomitant with induction of tumour cell death. ADC may provide a useful non-invasive pharmacodynamic biomarker for early clinical assessment of response to selumetinib and other MEK-ERK1/2 signalling-targeted therapies.
Collapse
Affiliation(s)
- M Beloueche-Babari
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - Y Jamin
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - V Arunan
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - S Walker-Samuel
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - M Revill
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - P D Smith
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - J Halliday
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - J C Waterton
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - H Barjat
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - P Workman
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - M O Leach
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - S P Robinson
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| |
Collapse
|
33
|
Meijer TWH, Kaanders JHAM, Span PN, Bussink J. Targeting hypoxia, HIF-1, and tumor glucose metabolism to improve radiotherapy efficacy. Clin Cancer Res 2013; 18:5585-94. [PMID: 23071360 DOI: 10.1158/1078-0432.ccr-12-0858] [Citation(s) in RCA: 334] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Radiotherapy, an important treatment modality in oncology, kills cells through induction of oxidative stress. However, malignant tumors vary in their response to irradiation as a consequence of resistance mechanisms taking place at the molecular level. It is important to understand these mechanisms of radioresistance, as counteracting them may improve the efficacy of radiotherapy. In this review, we describe how the hypoxia-inducible factor 1 (HIF-1) pathway has a profound effect on the response to radiotherapy. The main focus will be on HIF-1-controlled protection of the vasculature postirradiation and on HIF-1 regulation of glycolysis and the pentose phosphate pathway. This aberrant cellular metabolism increases the antioxidant capacity of tumors, thereby countering the oxidative stress caused by irradiation. From the results of translational studies and the first clinical phase I/II trials, it can be concluded that targeting HIF-1 and tumor glucose metabolism at several levels reduces the antioxidant capacity of tumors, affects the tumor microenvironment, and sensitizes various solid tumors to irradiation.
Collapse
Affiliation(s)
- Tineke W H Meijer
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
34
|
Mimeault M, Batra SK. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer- and metastasis-initiating cells. J Cell Mol Med 2013; 17:30-54. [PMID: 23301832 PMCID: PMC3560853 DOI: 10.1111/jcmm.12004] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 11/20/2012] [Indexed: 12/12/2022] Open
Abstract
Accumulating lines of experimental evidence have revealed that hypoxia-inducible factors, HIF-1α and HIF-2α, are key regulators of the adaptation of cancer- and metastasis-initiating cells and their differentiated progenies to oxygen and nutrient deprivation during cancer progression under normoxic and hypoxic conditions. Particularly, the sustained stimulation of epidermal growth factor receptor (EGFR), insulin-like growth factor-1 receptor (IGF-1R), stem cell factor (SCF) receptor KIT, transforming growth factor-β receptors (TGF-βRs) and Notch and their downstream signalling elements such as phosphatidylinositol 3′-kinase (PI3K)/Akt/molecular target of rapamycin (mTOR) may lead to an enhanced activity of HIFs. Moreover, the up-regulation of HIFs in cancer cells may also occur in the hypoxic intratumoral regions formed within primary and secondary neoplasms as well as in leukaemic cells and metastatic prostate and breast cancer cells homing in the hypoxic endosteal niche of bone marrow. The activated HIFs may induce the expression of numerous gene products such as induced pluripotency-associated transcription factors (Oct-3/4, Nanog and Sox-2), glycolysis- and epithelial-mesenchymal transition (EMT) programme-associated molecules, including CXC chemokine receptor 4 (CXCR4), snail and twist, microRNAs and angiogenic factors such as vascular endothelial growth factor (VEGF). These gene products in turn can play critical roles for high self-renewal ability, survival, altered energy metabolism, invasion and metastases of cancer cells, angiogenic switch and treatment resistance. Consequently, the targeting of HIF signalling network and altered metabolic pathways represents new promising strategies to eradicate the total mass of cancer cells and improve the efficacy of current therapies against aggressive and metastatic cancers and prevent disease relapse.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, College of Medicine, Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | | |
Collapse
|
35
|
Liu Z, Li Y, Yang R. Effects of resveratrol on vascular endothelial growth factor expression in osteosarcoma cells and cell proliferation. Oncol Lett 2012. [PMID: 23205110 DOI: 10.3892/ol.2012.824] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The aim of the current study was to investigate the effects of resveratrol (Res) on vascular endothelial growth factor (VEGF) expression and cell proliferation in the human osteosarcoma cell line U20S. U20S cells were treated with Res at various concentrations (0, 10, 20 and 40 μmol/l) for various times (24, 48 and 72 h). The inhibitory effect of Res on U20S proliferation was observed using methyl thiazolyl tetrazolium (MTT) colorimetry. VEGF expression was determined using real-time polymerase chain reaction (RT-PCR) and western blot analysis. The inhibitory effect of Res on U20S proliferation increased as the concentration of Res increased. The inhibitory effect also increased with time. Res had an inhibitory effect on VEGF expression and significantly inhibited U20S cell proliferation. Res may exert an anti-osteosarcoma effect by inhibiting VEGF expression in tumor cells.
Collapse
Affiliation(s)
- Zhonghe Liu
- Department of Orthopedics, First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100
| | | | | |
Collapse
|