1
|
Suvac A, Ashton J, Bristow RG. Tumour hypoxia in driving genomic instability and tumour evolution. Nat Rev Cancer 2025:10.1038/s41568-024-00781-9. [PMID: 39875616 DOI: 10.1038/s41568-024-00781-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/30/2025]
Abstract
Intratumour hypoxia is a feature of all heterogenous solid tumours. Increased levels or subregions of tumour hypoxia are associated with an adverse clinical prognosis, particularly when this co-occurs with genomic instability. Experimental evidence points to the acquisition of DNA and chromosomal alterations in proliferating hypoxic cells secondary to inhibition of DNA repair pathways such as homologous recombination, base excision repair and mismatch repair. Cell adaptation and selection in repair-deficient cells give rise to a model whereby novel single-nucleotide mutations, structural variants and copy number alterations coexist with altered mitotic control to drive chromosomal instability and aneuploidy. Whole-genome sequencing studies support the concept that hypoxia is a critical microenvironmental cofactor alongside the driver mutations in MYC, BCL2, TP53 and PTEN in determining clonal and subclonal evolution in multiple tumour types. We propose that the hypoxic tumour microenvironment selects for unstable tumour clones which survive, propagate and metastasize under reduced immune surveillance. These aggressive features of hypoxic tumour cells underpin resistance to local and systemic therapies and unfavourable outcomes for patients with cancer. Possible ways to counter the effects of hypoxia to block tumour evolution and improve treatment outcomes are described.
Collapse
Affiliation(s)
- Alexandru Suvac
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jack Ashton
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert G Bristow
- Translational Oncogenomics Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK.
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK.
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
Fletcher T, Thompson AJ, Ashrafian H, Darzi A. The measurement and modification of hypoxia in colorectal cancer: overlooked but not forgotten. Gastroenterol Rep (Oxf) 2022; 10:goac042. [PMID: 36032656 PMCID: PMC9406947 DOI: 10.1093/gastro/goac042] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/18/2022] [Accepted: 07/21/2022] [Indexed: 11/14/2022] Open
Abstract
Tumour hypoxia is the inevitable consequence of a tumour's rapid growth and disorganized, inefficient vasculature. The compensatory mechanisms employed by tumours, and indeed the absence of oxygen itself, hinder the ability of all treatment modalities. The clinical consequence is poorer overall survival, disease-free survival, and locoregional control. Recognizing this, clinicians have been attenuating the effect of hypoxia, primarily with hypoxic modification or with hypoxia-activated pro-drugs, and notable success has been demonstrated. However, in the case of colorectal cancer (CRC), there is a general paucity of knowledge and evidence surrounding the measurement and modification of hypoxia, and this is possibly due to the comparative inaccessibility of such tumours. We specifically review the role of hypoxia in CRC and focus on the current evidence for the existence of hypoxia in CRC, the majority of which originates from indirect positron emission topography imaging with hypoxia selective radiotracers; the evidence correlating CRC hypoxia with poorer oncological outcome, which is largely based on the measurement of hypoxia inducible factor in correlation with clinical outcome; the evidence of hypoxic modification in CRC, of which no direct evidence exists, but is reflected in a number of indirect markers; the prognostic and monitoring implications of accurate CRC hypoxia quantification and its potential in the field of precision oncology; and the present and future imaging tools and technologies being developed for the measurement of CRC hypoxia, including the use of blood-oxygen-level-dependent magnetic resonance imaging and diffuse reflectance spectroscopy.
Collapse
Affiliation(s)
- Teddy Fletcher
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| | - Alex J Thompson
- The Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK
| | - Hutan Ashrafian
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| | - Ara Darzi
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| |
Collapse
|
3
|
Boreel DF, Span PN, Heskamp S, Adema GJ, Bussink J. Targeting Oxidative Phosphorylation to Increase the Efficacy of Radio- and Immune-Combination Therapy. Clin Cancer Res 2021; 27:2970-2978. [PMID: 33419779 DOI: 10.1158/1078-0432.ccr-20-3913] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/25/2020] [Accepted: 01/06/2021] [Indexed: 11/16/2022]
Abstract
As tumors grow, they upregulate glycolytic and oxidative metabolism to support their increased and altered energetic demands. These metabolic changes have major effects on the tumor microenvironment. One of the properties leading to this aberrant metabolism is hypoxia, which occurs when tumors outgrow their often-chaotic vasculature. This scarcity of oxygen is known to induce radioresistance but can also have a disrupting effect on the antitumor immune response. Hypoxia inhibits immune effector cell function, while immune cells with a more suppressing phenotype become more active. Therefore, hypoxia strongly affects the efficacy of both radiotherapy and immunotherapy, as well as this therapy combination. Inhibition of oxidative phosphorylation (OXPHOS) is gaining interest for its ability to combat tumor hypoxia, and there are strong indications that this results in a reactivation of the immune response. This strategy decreases oxygen consumption, leading to better oxygenation of hypoxic tumor areas and eventually an increase in immunogenic cell death induced by radio-immunotherapy combinations. Promising preclinical improvements in radio- and immunotherapy efficacy have been observed by the hypoxia-reducing effect of OXPHOS inhibitors and several compounds are currently in clinical trials for their anticancer properties. Here, we will review the pharmacologic attenuation of tumor hypoxia using OXPHOS inhibitors, with emphasis on their impact on the intrinsic antitumor immune response and how this affects the efficacy of (combined) radio- and immunotherapy.
Collapse
Affiliation(s)
- Daan F Boreel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands. .,Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paul N Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
4
|
Rich LJ, Damasco JA, Bulmahn JC, Kutscher HL, Prasad PN, Seshadri M. Photoacoustic and Magnetic Resonance Imaging of Hybrid Manganese Dioxide-Coated Ultra-small NaGdF 4 Nanoparticles for Spatiotemporal Modulation of Hypoxia in Head and Neck Cancer. Cancers (Basel) 2020; 12:cancers12113294. [PMID: 33172178 PMCID: PMC7694772 DOI: 10.3390/cancers12113294] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Tumor hypoxia is a documented negative prognostic factor that contributes to treatment resistance in head and neck cancer. In the present study, we use non-invasive magnetic resonance imaging (MRI) and photoacoustic imaging (PAI) to evaluate the ability of ultra-small manganese dioxide coated nanoparticles to modulate tumor oxygenation in vitro and in vivo. Our results highlight the utility of MRI and PAI in mapping tumor hypoxia and nanoparticle delivery and demonstrate the potential of image-guided nanodelivery in alleviating tumor hypoxia in head and neck cancer. Abstract There is widespread interest in developing agents to modify tumor hypoxia in head and neck squamous cell carcinomas (HNSCC). Here, we report on the synthesis, characterization, and potential utility of ultra-small NaYF4:Nd3+/NaGdF4 nanocrystals coated with manganese dioxide (usNP-MnO2) for spatiotemporal modulation of hypoxia in HNSCC. Using a dual modality imaging approach, we first visualized the release of Mn2+ using T1-weighted magnetic resonance imaging (MRI) and modulation of oxygen saturation (%sO2) using photoacoustic imaging (PAI) in vascular channel phantoms. Combined MRI and PAI performed in patient-derived HNSCC xenografts following local and systemic delivery of the hybrid nanoparticles enabled mapping of intratumoral nanoparticle accumulation (based on T1 contrast enhancement) and improvement in tumor oxygenation (increased %sO2) within the tumor microenvironment. Our results demonstrate the potential of hybrid nanoparticles for the modulation of tumor hypoxia in head and neck cancer. Our findings also highlight the potential of combined MRI-PAI for simultaneous mapping nanoparticle delivery and oxygenation changes in tumors. Such imaging methods could be valuable in the precise selection of patients that are likely to benefit from hypoxia-modifying nanotherapies.
Collapse
Affiliation(s)
- Laurie J. Rich
- Center for Oral Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA;
| | - Jossana A. Damasco
- Department of Chemistry and the Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (J.A.D.); (J.C.B.); (H.L.K.)
| | - Julia C. Bulmahn
- Department of Chemistry and the Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (J.A.D.); (J.C.B.); (H.L.K.)
| | - Hilliard L. Kutscher
- Department of Chemistry and the Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (J.A.D.); (J.C.B.); (H.L.K.)
- Department of Medicine, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
- Department of Anesthesiology, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Paras N. Prasad
- Department of Chemistry and the Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (J.A.D.); (J.C.B.); (H.L.K.)
- Correspondence: (P.N.P.); (M.S.)
| | - Mukund Seshadri
- Center for Oral Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA;
- Department of Dentistry and Maxillofacial Prosthetics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
- Correspondence: (P.N.P.); (M.S.)
| |
Collapse
|
5
|
Yu P, Han X, Yin L, Hui K, Guo Y, Yuan A, Hu Y, Wu J. Artificial Red Blood Cells Constructed by Replacing Heme with Perfluorodecalin for Hypoxia‐Induced Radioresistance. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Peng Yu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Xiaoxue Han
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Lining Yin
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Kangyu Hui
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Yunfei Guo
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Jiangsu Provincial Key Laboratory for Nano TechnologyNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Jiangsu Provincial Key Laboratory for Nano TechnologyNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| |
Collapse
|
6
|
Fix SM, Papadopoulou V, Velds H, Kasoji SK, Rivera JN, Borden MA, Chang S, Dayton PA. Oxygen microbubbles improve radiotherapy tumor control in a rat fibrosarcoma model - A preliminary study. PLoS One 2018; 13:e0195667. [PMID: 29630640 PMCID: PMC5891067 DOI: 10.1371/journal.pone.0195667] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/27/2018] [Indexed: 11/19/2022] Open
Abstract
Cancer affects 39.6% of Americans at some point during their lifetime. Solid tumor microenvironments are characterized by a disorganized, leaky vasculature that promotes regions of low oxygenation (hypoxia). Tumor hypoxia is a key predictor of poor treatment outcome for all radiotherapy (RT), chemotherapy and surgery procedures, and is a hallmark of metastatic potential. In particular, the radiation therapy dose needed to achieve the same tumor control probability in hypoxic tissue as in normoxic tissue can be up to 3 times higher. Even very small tumors (<2-3 mm3) comprise 10-30% of hypoxic regions in the form of chronic and/or transient hypoxia fluctuating over the course of seconds to days. We investigate the potential of recently developed lipid-stabilized oxygen microbubbles (OMBs) to improve the therapeutic ratio of RT. OMBs, but not nitrogen microbubbles (NMBs), are shown to significantly increase dissolved oxygen content when added to water in vitro and increase tumor oxygen levels in vivo in a rat fibrosarcoma model. Tumor control is significantly improved with OMB but not NMB intra-tumoral injections immediately prior to RT treatment and effect size is shown to depend on initial tumor volume on RT treatment day, as expected.
Collapse
Affiliation(s)
- Samantha M. Fix
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and NC State University, Chapel Hill, NC, United States of America
| | - Hunter Velds
- Department of Mechanical Engineering, University of Colorado, Boulder, Colorado, United States of America
| | - Sandeep K. Kasoji
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and NC State University, Chapel Hill, NC, United States of America
| | - Judith N. Rivera
- Department of Radiation Oncology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Mark A. Borden
- Department of Radiation Oncology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Sha Chang
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and NC State University, Chapel Hill, NC, United States of America
- Department of Radiation Oncology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Paul A. Dayton
- Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and NC State University, Chapel Hill, NC, United States of America
| |
Collapse
|
7
|
Yasui H, Kubota N, Nishizumi J, Sakai Y, Yamamori T, Inanami O. Preclinical study on hypoxic radiosensitizing effects of glycididazole in comparison with those of doranidazole in vitro and in vivo. Oncol Lett 2018; 15:1993-1998. [PMID: 29434899 DOI: 10.3892/ol.2017.7481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/20/2017] [Indexed: 11/06/2022] Open
Abstract
To overcome the radioresistance of hypoxic cells in solid tumor, numerous types of radiosensitizers specifically against them have been developed. Glycididazole has a chemical structure in which two metronidazole forms are combined, and is widely used as a hypoxic radiosensitizer in China. However, a detailed investigation of its radiosensitizing properties has not been performed. The present study reported a comparative assessment of glycididazole and doranidazole, another hypoxic radiosensitizer. All experiments were performed using the murine squamous cell carcinoma cell line SCCVII. Prior to X-irradiation, the cells were treated with the test drugs at concentrations of 10 mM and 200 mg/kg in vitro and in vivo, respectively. Uptake and their intratumor chemical forms were analyzed by high performance liquid chromatography (HPLC). Both drugs enhanced the reproductive cell death induced by X-irradiation under hypoxia. However, the growth delay assay of the transplanted tumor revealed the combination of X-irradiation and glycididazole showed a similar antitumor effect to that of X-irradiation alone, whereas doranidazole significantly sensitized the cells to X-irradiation. HPLC analysis revealed that incorporated glycididazole was decomposed to metronidazole and was therefore present at a lower concentration compared with that of doranidazole. The decomposition of glycididazole to metronidazole reduced its radiosensitizing efficiency in vivo. Elucidation of the kinetics of drugs containing metabolizable chemical forms is necessary for the optimization of clinical treatment.
Collapse
Affiliation(s)
- Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan.,Central Institute of Isotope Science, Hokkaido University, Sapporo, Hokkaido 060-0815, Japan
| | - Nobuo Kubota
- R&D Laboratories, Pola Pharma Inc., Yokohama, Kanagawa 244-0812, Japan
| | - Junko Nishizumi
- R&D Laboratories, Pola Pharma Inc., Yokohama, Kanagawa 244-0812, Japan
| | - Yuri Sakai
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Tohru Yamamori
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Division of Veterinary Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| |
Collapse
|
8
|
Wang H, Mu X, He H, Zhang XD. Cancer Radiosensitizers. Trends Pharmacol Sci 2017; 39:24-48. [PMID: 29224916 DOI: 10.1016/j.tips.2017.11.003] [Citation(s) in RCA: 356] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023]
Abstract
Radiotherapy (RT) is a mainstay treatment for many types of cancer, although it is still a large challenge to enhance radiation damage to tumor tissue and reduce side effects to healthy tissue. Radiosensitizers are promising agents that enhance injury to tumor tissue by accelerating DNA damage and producing free radicals. Several strategies have been exploited to develop highly effective and low-toxicity radiosensitizers. In this review, we highlight recent progress on radiosensitizers, including small molecules, macromolecules, and nanomaterials. First, small molecules are reviewed based on free radicals, pseudosubstrates, and other mechanisms. Second, nanomaterials, such as nanometallic materials, especially gold-based materials that have flexible surface engineering and favorable kinetic properties, have emerged as promising radiosensitizers. Finally, emerging macromolecules have shown significant advantages in RT because these molecules can be combined with biological therapy as well as drug delivery. Further research on the mechanisms of radioresistance and multidisciplinary approaches will accelerate the development of radiosensitizers.
Collapse
Affiliation(s)
- Hao Wang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Number 238, Baidi Road, Tianjin 300192, China; These authors have contributed equally
| | - Xiaoyu Mu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China; These authors have contributed equally
| | - Hua He
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiao-Dong Zhang
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China; Tianjin Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, China.
| |
Collapse
|
9
|
Gaustad JV, Simonsen TG, Andersen LMK, Rofstad EK. Vascular abnormalities and development of hypoxia in microscopic melanoma xenografts. J Transl Med 2017; 15:241. [PMID: 29183378 PMCID: PMC5706333 DOI: 10.1186/s12967-017-1347-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/21/2017] [Indexed: 01/10/2023] Open
Abstract
Background Studies investigating the oxygenation status and the development of hypoxia in microscopic tumors are sparse. The purpose of this study was to measure the extent of hypoxia in microscopic melanoma xenografts and to search for possible mechanisms leading to the development of hypoxia in these tumors. Methods A-07, D-12, R-18, and U-25 human melanoma xenografts grown in dorsal window chambers or as flank tumors were used as preclinical tumor models. Morphologic and functional parameters of vascular networks were assessed with intravital microscopy, and the expression of angiogenesis-related genes was assessed with quantitative PCR. Microvessels, pericytes, and the extent of hypoxia were assessed by immunohistochemistry in microscopic tumors by using CD31, αSMA, and pimonidazole as markers, and the extent of radiobiological hypoxia was assessed in macroscopic flank tumors. Results Macroscopic R-18 and U-25 tumors showed extensive hypoxia, whereas macroscopic A-07 and D-12 tumors were less hypoxic. R-18 and U-25 tumors developed hypoxic regions before they reached a size of 2–3 mm in diameter, whereas A-07 and D-12 tumors of similar size did not show hypoxic regions. The development of hypoxic regions was not caused by low vessel density, but was rather a result of inadequate vascular function. Inadequate vascular function was not caused by low vessel diameters or long vessel segments, but was associated with poor vascular pericyte coverage. Poor pericyte coverage was associated with the expression of eight angiogenesis-related genes. Conclusions Two of the four investigated melanoma models developed hypoxic regions in microscopic tumors, and the development of hypoxia was associated with poor vascular pericyte coverage and inadequate vascular function.
Collapse
Affiliation(s)
- Jon-Vidar Gaustad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0310, Oslo, Norway.
| | - Trude G Simonsen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Lise Mari K Andersen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Einar K Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| |
Collapse
|
10
|
Morrison RA, Rybak-Smith MJ, Thompson JM, Thiebaut B, Hill MA, Townley HE. Efficacy of radiosensitizing doped titania nanoparticles under hypoxia and preparation of an embolic microparticle. Int J Nanomedicine 2017; 12:3851-3863. [PMID: 28572729 PMCID: PMC5441663 DOI: 10.2147/ijn.s127341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The aim of this study was to develop a manufacturing protocol for large-scale production of doped titania radiosensitizing nanoparticles (NPs) to establish their activity under hypoxia and to produce a multimodal radiosensitizing embolic particle for cancer treatment. We have previously shown that radiosensitizing NPs can be synthesized from titania doped with rare earth elements, especially gadolinium. To translate this technology to the clinic, a crucial step is to find a suitable, scalable, high-throughput method. Herein, we have described the use of flame spray pyrolysis (FSP) to generate NPs from titanium and gadolinium precursors to produce titania NPs doped with 5 at% gadolinium. The NPs were fully characterized, and their capacity to act as radiosensitizers was confirmed by clonogenic assays. The integrity of the NPs in vitro was also ascertained due to the potentially adverse effects of free gadolinium in the body. The activity of the NPs was then studied under hypoxia since this is often a barrier to effective radiotherapy. In vitro radiosensitization experiments were performed with both the hypoxia mimetics deferoxamine and cobalt chloride and also under true hypoxia (oxygen concentration of 0.2%). It was shown that the radiosensitizing NPs were able to cause a significant increase in cell death even after irradiation under hypoxic conditions such as those found in tumors. Subsequently, the synthesized NPs were used to modify polystyrene embolization microparticles. The NPs were sintered to the surface of the microparticles by heating at 230°C for 15 minutes. This resulted in a good coverage of the surface and to generate embolization particles that were shown to be radiosensitizing. Such multimodal particles could therefore result in occlusion of the tumor blood vessels in conjunction with localized reactive oxygen species generation, even under hypoxic conditions such as those found in the center of tumors.
Collapse
Affiliation(s)
| | | | - James M Thompson
- Gray Laboratories, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford
| | | | - Mark A Hill
- Gray Laboratories, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford
| | - Helen E Townley
- Department of Engineering Science.,Nuffield Department of Obstetrics and Gynaecology, John Radcliffe Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
11
|
Xu J, Chen A, Xiao J, Jiang Z, Tian Y, Tang Q, Cao P, Dai Y, Krainik A, Shen J. Evaluation of tumour vascular distribution and function using immunohistochemistry and BOLD fMRI with carbogen inhalation. Clin Radiol 2016; 71:1255-1262. [PMID: 27170218 DOI: 10.1016/j.crad.2016.04.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 02/18/2016] [Accepted: 04/06/2016] [Indexed: 10/21/2022]
Abstract
AIM To evaluate oxygenation changes in rat subcutaneous C6 gliomas using blood-oxygen-level dependent (BOLD) functional magnetic resonance imaging (fMRI) combined with non-haemodynamic response function (non-HRF) analysis. MATERIALS AND METHODS BOLD fMRI were performed during carbogen inhalation in 20 Wistar rats bearing gliomas. Statistical maps of spatial oxygenation changes were computed by a dedicated non-HRF analysis algorithm. Three types of regions of interest (ROIs) were defined: (1) maximum re-oxygenation zone (ROImax), (2) re-oxygenation zones that were less than the maximum re-oxygenation (ROInon-max), and (3) zones without significant re-oxygenation (ROInone). The values of percent BOLD signal change (PSC), percent enhancement (ΔSI), and significant re-oxygenation (T) were extracted from each ROI. Tumours were sectioned for histology using the fMRI scan orientation and were stained with haematoxylin and eosin and CD105. The number of microvessels (MVN) in each ROI was counted. Differences and correlations among the values for T, PSC, ΔSI, and MVN were determined. RESULTS After carbogen inhalation, the PSC significantly increased in the ROImax areas (p<0.01) located in the tumour parenchyma. No changes occurred in any of the ROInone areas (20/20). Some changes occurred in a minority of the ROInon-max areas (3/60) corresponding to tumour necrosis. MVN and PSC (R=0.59, p=0.01) were significantly correlated in the ROImax areas. In the ROInon-max areas, MVN was significantly correlated with PSC (R=0.55, p=0.00) and ΔSI (R=0.37, p=0.00). CONCLUSIONS Statistical maps obtained via BOLD fMRI with non-HRF analysis can assess the re-oxygenation of gliomas.
Collapse
Affiliation(s)
- J Xu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - A Chen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - J Xiao
- Department of Radiology, The Central Hospital of Wuhan, Wuhan, China
| | - Z Jiang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China; Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.
| | - Y Tian
- Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China; Suzhou Key Laboratory for Radiation Oncology, Suzhou, China
| | - Q Tang
- Department of Radiology, Wuxi People's Hospital, Wuxi, China
| | - P Cao
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Y Dai
- Magnetic Resonance Imaging Institute for Biomedical Research, Wayne State University, Detroit, MI, USA
| | - A Krainik
- Department of Neuroradiology and MRI, CHU Grenoble-IFR1, Grenoble, France
| | - J Shen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
A role for dynamic contrast-enhanced magnetic resonance imaging in predicting tumour radiation response. Br J Cancer 2016; 114:1206-11. [PMID: 27140315 PMCID: PMC4891499 DOI: 10.1038/bjc.2016.110] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/25/2016] [Accepted: 03/22/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Dynamic contrast-enhanced (DCE) MRI may provide prognostic insights into tumour radiation response. This study examined quantitative DCE MRI parameters in rat tumours, as potential biomarkers of tumour growth delay following single high-dose irradiation. METHODS Dunning R3327-AT1 prostate tumours were evaluated by DCE MRI following intravenous injection of Gd-DTPA. The next day tumours were irradiated (single dose of 30 Gy), while animals breathed air (n=4) or oxygen (n=4); two animals were non-irradiated controls. Growth was followed and tumour volume-quadrupling time (T4) was compared with pre-irradiation DCE assessments. RESULTS Irradiation caused significant tumour growth delay (T4 ranged from 28 to 48 days for air-breathing rats, and 40 to 75 days for oxygen-breathing rats) compared with the controls (T4=7 to 9 days). A strong correlation was observed between T4 and extravascular-extracellular volume fraction (ve) irrespective of the gas inhaled during irradiation. There was also a correlation between T4 and volume transfer constant (K(trans)) for the air-breathing group alone. CONCLUSIONS The data provide rationale for expanded studies of other tumour sites, types and progressively patients, and are potentially significant, as many patients undergo contrast-enhanced MRI as part of treatment planning.
Collapse
|
13
|
Hassan Metwally MA, Ali R, Kuddu M, Shouman T, Strojan P, Iqbal K, Prasad R, Grau C, Overgaard J. IAEA-HypoX. A randomized multicenter study of the hypoxic radiosensitizer nimorazole concomitant with accelerated radiotherapy in head and neck squamous cell carcinoma. Radiother Oncol 2015; 116:15-20. [DOI: 10.1016/j.radonc.2015.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/10/2015] [Accepted: 04/10/2015] [Indexed: 11/26/2022]
|
14
|
Horn D, Hess J, Freier K, Hoffmann J, Freudlsperger C. Targeting EGFR-PI3K-AKT-mTOR signaling enhances radiosensitivity in head and neck squamous cell carcinoma. Expert Opin Ther Targets 2015; 19:795-805. [PMID: 25652792 DOI: 10.1517/14728222.2015.1012157] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Head and neck squamous cell carcinoma (HNSCC) is frequently characterized by high resistance to radiotherapy, which critically depends on both altered signaling pathways within tumor cells and their dynamic interaction with the tumor microenvironment. AREAS COVERED This review covers EGFR-phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT)-mechanistic target of rapamycin (mTOR) signaling in HNSCC. The role of each pathway node in radioresistance is discussed. Preclinical and clinical innovative aspects of targeting EGFR-PI3K-AKT and mTOR are demonstrated. Ongoing clinical trials and future perspectives are presented. EXPERT OPINION Different cellular signaling pathways seem to mediate radioresistance in advanced HNSCC and various molecular targeted therapies are currently being investigated to sensitize tumor cells to radiotherapy. Recently, new insights in the mutational landscape of HNSCC unraveled critical alterations in putative oncogenes and tumor suppressor genes and have emphasized the importance of PI3K and the corresponding upstream and downstream signaling pathways in pathogenesis and treatment response. The frequent activation of the EGFR-PI3K-AKT-mTOR pathway in HNSCC and its implication in the context of radiosensitivity make this pathway one of the most promising targets in the therapy of HNSCC patients. Clinical studies targeting EGFR and mTOR in combination with radiotherapy are under investigation.
Collapse
Affiliation(s)
- Dominik Horn
- University Hospital Heidelberg, Department of Oral and Maxillofacial Surgery , Im Neuenheimer Feld 400, 69120 Heidelberg , Germany +49 0 6221 56 38462 ; +49 0 6221 56 4222 ;
| | | | | | | | | |
Collapse
|
15
|
Kelly CJ, Hussien K, Fokas E, Kannan P, Shipley RJ, Ashton TM, Stratford M, Pearson N, Muschel RJ. Regulation of O2 consumption by the PI3K and mTOR pathways contributes to tumor hypoxia. Radiother Oncol 2014; 111:72-80. [PMID: 24631147 PMCID: PMC4070024 DOI: 10.1016/j.radonc.2014.02.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 01/10/2014] [Accepted: 02/09/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inhibitors of the phosphatidylinositol 3-kinase (PI3K) and the mammalian target of rapamycin (mTOR) pathway are currently in clinical trials. In addition to antiproliferative and proapoptotic effects, these agents also diminish tumor hypoxia. Since hypoxia is a major cause of resistance to radiotherapy, we sought to understand how it is regulated by PI3K/mTOR inhibition. METHODS Whole cell, mitochondrial, coupled and uncoupled oxygen consumption were measured in cancer cells after inhibition of PI3K (Class I) and mTOR by pharmacological means or by RNAi. Mitochondrial composition was assessed by immunoblotting. Hypoxia was measured in spheroids, in tumor xenografts and predicted with mathematical modeling. RESULTS Inhibition of PI3K and mTOR reduced oxygen consumption by cancer cell lines is predominantly due to reduction of mitochondrial respiration coupled to ATP production. Hypoxia in tumor spheroids was reduced, but returned after removal of the drug. Murine tumors had increased oxygenation even in the absence of average perfusion changes or tumor necrosis. CONCLUSIONS Targeting the PI3K/mTOR pathway substantially reduces mitochondrial oxygen consumption thereby reducing tumor hypoxia. These alterations in tumor hypoxia should be considered in the design of clinical trials using PI3K/mTOR inhibitors, particularly in conjunction with radiotherapy.
Collapse
Affiliation(s)
- Catherine J Kelly
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, UK
| | - Kamila Hussien
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, UK
| | - Emmanouil Fokas
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, UK
| | - Pavitra Kannan
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, UK
| | | | - Thomas M Ashton
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, UK
| | - Michael Stratford
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, UK
| | | | - Ruth J Muschel
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, UK.
| |
Collapse
|
16
|
Navarro-Teulon I, Lozza C, Pèlegrin A, Vivès E, Pouget JP. General overview of radioimmunotherapy of solid tumors. Immunotherapy 2013; 5:467-87. [PMID: 23638743 DOI: 10.2217/imt.13.34] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Radioimmunotherapy (RIT) represents an attractive tool for the treatment of local and/or diffuse tumors with radiation. In RIT, cytotoxic radionuclides are delivered by monoclonal antibodies that specifically target tumor-associated antigens or the tumor microenvironment. While RIT has been successfully employed for the treatment of lymphoma, mostly with radiolabeled antibodies against CD20 (Bexxar(®); Corixa Corp., WA, USA and Zevalin(®); Biogen Idec Inc., CA, USA and Schering AG, Berlin, Germany), its use in solid tumors is more challenging and, so far, few trials have progressed beyond Phase II. This review provides an update on antibody-radionuclide conjugates and their use in RIT. It also discusses possible optimization strategies to improve the clinical response by considering biological, radiobiological and physical features.
Collapse
|
17
|
Krainik A, Villien M, Troprès I, Attyé A, Lamalle L, Bouvier J, Pietras J, Grand S, Le Bas JF, Warnking J. Functional imaging of cerebral perfusion. Diagn Interv Imaging 2013; 94:1259-78. [PMID: 24011870 DOI: 10.1016/j.diii.2013.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The functional imaging of perfusion enables the study of its properties such as the vasoreactivity to circulating gases, the autoregulation and the neurovascular coupling. Downstream from arterial stenosis, this imaging can estimate the vascular reserve and the risk of ischemia in order to adapt the therapeutic strategy. This method reveals the hemodynamic disorders in patients suffering from Alzheimer's disease or with arteriovenous malformations revealed by epilepsy. Functional MRI of the vasoreactivity also helps to better interpret the functional MRI activation in practice and in clinical research.
Collapse
Affiliation(s)
- A Krainik
- Clinique universitaire de neuroradiologie et IRM, CHU de Grenoble, CS 10217, 38043 Grenoble cedex, France; Inserm U836, université Joseph-Fourier, site santé, chemin Fortuné-Ferrini, 38706 La Tronche cedex, France; UMS IRMaGe, unité IRM 3T recherche, CHU de Grenoble, CS 10217, 38043 Grenoble cedex 9, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Jones LW, Fels DR, West M, Allen JD, Broadwater G, Barry WT, Wilke LG, Masko E, Douglas PS, Dash RC, Povsic TJ, Peppercorn J, Marcom PK, Blackwell KL, Kimmick G, Turkington TG, Dewhirst MW. Modulation of circulating angiogenic factors and tumor biology by aerobic training in breast cancer patients receiving neoadjuvant chemotherapy. Cancer Prev Res (Phila) 2013; 6:925-37. [PMID: 23842792 DOI: 10.1158/1940-6207.capr-12-0416] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Aerobic exercise training (AET) is an effective adjunct therapy to attenuate the adverse side-effects of adjuvant chemotherapy in women with early breast cancer. Whether AET interacts with the antitumor efficacy of chemotherapy has received scant attention. We carried out a pilot study to explore the effects of AET in combination with neoadjuvant doxorubicin-cyclophosphamide (AC+AET), relative to AC alone, on: (i) host physiology [exercise capacity (VO2 peak), brachial artery flow-mediated dilation (BA-FMD)], (ii) host-related circulating factors [circulating endothelial progenitor cells (CEP) cytokines and angiogenic factors (CAF)], and (iii) tumor phenotype [tumor blood flow ((15)O-water PET), tissue markers (hypoxia and proliferation), and gene expression] in 20 women with operable breast cancer. AET consisted of three supervised cycle ergometry sessions/week at 60% to 100% of VO2 peak, 30 to 45 min/session, for 12 weeks. There was significant time × group interactions for VO2 peak and BA-FMD, favoring the AC+AET group (P < 0.001 and P = 0.07, respectively). These changes were accompanied by significant time × group interactions in CEPs and select CAFs [placenta growth factor, interleukin (IL)-1β, and IL-2], also favoring the AC+AET group (P < 0.05). (15)O-water positron emission tomography (PET) imaging revealed a 38% decrease in tumor blood flow in the AC+AET group. There were no differences in any tumor tissue markers (P > 0.05). Whole-genome microarray tumor analysis revealed significant differential modulation of 57 pathways (P < 0.01), including many that converge on NF-κB. Data from this exploratory study provide initial evidence that AET can modulate several host- and tumor-related pathways during standard chemotherapy. The biologic and clinical implications remain to be determined.
Collapse
Affiliation(s)
- Lee W Jones
- Duke Cancer Institute, Duke University Medical Center, Box 3085, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Reoxygenation using a novel CO2 therapy decreases the metastatic potential of osteosarcoma cells. Exp Cell Res 2013; 319:1988-1997. [PMID: 23727023 DOI: 10.1016/j.yexcr.2013.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 05/01/2013] [Accepted: 05/21/2013] [Indexed: 11/22/2022]
Abstract
Osteosarcoma is the most common primary solid malignant bone tumor. Despite substantial improvements in surgery and chemotherapy, metastasis remains a major cause of fatal outcomes, and the molecular mechanisms of metastasis are still poorly understood. Hypoxia, which is common in malignant tumors including osteosarcoma, increases expressions of hypoxia inducible factor (HIF)-1α, matrix metalloproteinase (MMP)-2 and MMP-9, and can induce invasiveness. As we previously showed a novel transcutaneous CO2 application to decrease HIF-1α expression and induce apoptosis in malignant fibrous histiocytoma, we hypothesize that transcutaneous CO2 application could suppress metastatic potential of osteosarcoma by improving hypoxic conditions. Here, we examined the effects of transcutaneous CO2 application on apoptosis, and development of pulmonary metastasis using a highly metastatic osteosarcoma cell line, LM8. Transcutaneous CO2 application significantly decreased tumor growth and pulmonary metastasis in LM8 cells. Apoptotic activity increased, and intratumoral hypoxia was improved with decreased expressions of HIF-1α, MMP-2 and MMP-9, significantly, in the CO2-treated tumors. In conclusion, we found that transcutaneous CO2 application can induce tumor cell apoptosis and might suppress pulmonary metastasis by improvement of hypoxic conditions with decreased expressions of HIF-1α and MMPs in highly metastatic osteosarcoma cell. These findings strongly indicate that this novel transcutaneous CO2 therapy could be a therapeutic breakthrough for osteosarcoma patients.
Collapse
|
20
|
Yasui H, Asanuma T, Kino J, Yamamori T, Meike S, Nagane M, Kubota N, Kuwabara M, Inanami O. The prospective application of a hypoxic radiosensitizer, doranidazole to rat intracranial glioblastoma with blood brain barrier disruption. BMC Cancer 2013; 13:106. [PMID: 23496909 PMCID: PMC3599813 DOI: 10.1186/1471-2407-13-106] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 03/03/2013] [Indexed: 11/24/2022] Open
Abstract
Background Glioblastoma is one of the intractable cancers and is highly resistant to ionizing radiation. This radioresistance is partly due to the presence of a hypoxic region which is widely found in advanced malignant gliomas. In the present study, we evaluated the effectiveness of the hypoxic cell sensitizer doranidazole (PR-350) using the C6 rat glioblastoma model, focusing on the status of blood brain barrier (BBB). Methods Reproductive cell death in the rat C6 glioma cell line was determined by means of clonogenic assay. An intracranial C6 glioma model was established for the in vivo experiments. To investigate the status of the BBB in C6 glioma bearing brain, we performed the Evans blue extravasation test. Autoradiography with [14C]-doranidazole was performed to examine the distribution of doranidazole in the glioma tumor. T2-weighted MRI was employed to examine the effects of X-irradiation and/or doranidazole on tumor growth. Results Doranidazole significantly enhanced radiation-induced reproductive cell death in vitro under hypoxia, but not under normoxia. The BBB in C6-bearing brain was completely disrupted and [14C]-doranidazole specifically penetrated the tumor regions. Combined treatment with X-irradiation and doranidazole significantly inhibited the growth of C6 gliomas. Conclusions Our results revealed that BBB disruption in glioma enables BBB-impermeable radiosensitizers to penetrate and distribute in the target region. This study is the first to propose that in malignant glioma the administration of hydrophilic hypoxic radiosensitizers could be a potent strategy for improving the clinical outcome of radiotherapy without side effects.
Collapse
Affiliation(s)
- Hironobu Yasui
- Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo, Hokkaido, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lawrence YR, Vikram B, Dignam JJ, Chakravarti A, Machtay M, Freidlin B, Takebe N, Curran WJ, Bentzen SM, Okunieff P, Coleman CN, Dicker AP. NCI-RTOG translational program strategic guidelines for the early-stage development of radiosensitizers. J Natl Cancer Inst 2013; 105:11-24. [PMID: 23231975 PMCID: PMC3536642 DOI: 10.1093/jnci/djs472] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 09/15/2012] [Accepted: 10/02/2012] [Indexed: 12/21/2022] Open
Abstract
The addition of chemotherapeutic agents to ionizing radiation has improved survival in many malignancies. Cure rates may be further improved by adding novel targeted agents to current radiotherapy or radiochemotherapy regimens. Despite promising laboratory data, progress in the clinical development of new drugs with radiation has been limited. To define and address the problems involved, a collaborative effort between individuals within the translational research program of the Radiation Oncology Therapy Group and the National Cancer Institute was established. We discerned challenges to drug development with radiation including: 1) the limited relevance of preclinical work, 2) the pharmaceutical industry's diminished interest, and 3) the important individual skills and institutional commitments required to ensure a successful program. The differences between early-phase trial designs with and without radiation are noted as substantial. The traditional endpoints for early-phase clinical trials-acute toxicity and maximum-tolerated dose-are of limited value when combining targeted agents with radiation. Furthermore, response rate is not a useful surrogate marker of activity in radiation combination trials.Consequently, a risk-stratified model for drug-dose escalation with radiation is proposed, based upon the known and estimated adverse effects. The guidelines discuss new clinical trial designs, such as the time-to-event continual reassessment method design for phase I trials, randomized phase II "screening" trials, and the use of surrogate endpoints, such as pathological response. It is hoped that by providing a clear pathway, this article will accelerate the rate of drug development with radiation.
Collapse
|
22
|
Kwan JJ, Kaya M, Borden MA, Dayton PA. Theranostic oxygen delivery using ultrasound and microbubbles. Theranostics 2012; 2:1174-84. [PMID: 23382774 PMCID: PMC3563146 DOI: 10.7150/thno.4410] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 06/08/2012] [Indexed: 11/17/2022] Open
Abstract
Means to overcome tumor hypoxia have been the subject of clinical investigations since the 1960's; however these studies have yet to find a treatment which is widely accepted. It has been known for nearly a century that hypoxic cells are more resistant to radiotherapy than aerobic cells, and tumor hypoxia is a major factor leading to the resistance of tumors to radiation treatment as well as several cytotoxic agents. In this manuscript, the application of ultrasound combined with oxygen-carrier microbubbles is demonstrated as a method to locally increase dissolved oxygen. Microbubbles can also be imaged by ultrasound, thus providing the opportunity for image-guided oxygen delivery. Simulations of gas diffusion and microbubble gas exchange show that small amounts (down to 5 vol%) of a low-solubility osmotic gas can substantially increase microbubble persistence and therefore production rates and stability of oxygen-carrier microbubbles. Simulations also indicate that the lipid shell can be engineered with long-chain lipids to increase oxygen payload during in vivo transit. Experimental results demonstrate that the application of ultrasound to destroy the microbubbles significantly enhances the local oxygen release. We propose this technology as an application for ultrasound image-guided release of oxygen directly to hypoxic tissue, such as tumor sites to enhance radiotherapy.
Collapse
Affiliation(s)
- James J Kwan
- Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309, USA
| | | | | | | |
Collapse
|
23
|
Ning S, Bednarski M, Oronsky B, Scicinski J, Saul G, Knox SJ. Dinitroazetidines are a novel class of anticancer agents and hypoxia-activated radiation sensitizers developed from highly energetic materials. Cancer Res 2012; 72:2600-8. [PMID: 22589277 DOI: 10.1158/0008-5472.can-11-2303] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In an effort to develop cancer therapies that maximize cytotoxicity, while minimizing unwanted side effects, we studied a series of novel compounds based on the highly energetic heterocyclic scaffold, dinitroazetidine. In this study, we report the preclinical validation of 1-bromoacetyl-3,3-dinitroazetidine (ABDNAZ), a representative lead compound currently in a phase I clinical trial in patients with cancer. In tumor cell culture, ABDNAZ generated reactive free radicals in a concentration- and time-dependent manner, modulating intracellular redox status and triggering apoptosis. When administered to mice as a single agent, ABDNAZ exhibited greater cytotoxicity than cisplatin or tirapazamine under hypoxic conditions. However, compared with cisplatin, ABDNAZ was better tolerated at submaximal doses, yielding significant tumor growth inhibition in the absence of systemic toxicity. Similarly, when combined with radiation, ABDNAZ accentuated antitumor efficacy along with the therapeutic index. Toxicity studies indicated that ABDNAZ was not myelosuppressive and no dose-limiting toxicity was apparent following daily administration for 14 days. Taken together, our findings offer preclinical proof-of-concept for ABDNAZ as a promising new anticancer agent with a favorable toxicity profile, either as a chemotherapeutic agent or a radiosensitizer.
Collapse
Affiliation(s)
- Shoucheng Ning
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
24
|
Han J, Yu M, Dai M, Li H, Xiu R, Liu Q. Decreased expression of MDR1 in PEG-conjugated hemoglobin solution combined cisplatin treatment in a tumor xenograft model. ACTA ACUST UNITED AC 2012; 40:239-44. [PMID: 22432540 DOI: 10.3109/10731199.2012.663385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study aims to examine the contribution of PEG-conjugated hemoglobin combined with cisplatin to the expression of HIF-1α and MDR1 in a tumor xenograft model. Cervical carcinoma models were assigned to 4 groups and treated respectively: group 1(control); group 2, cisplatin; group 3, PEG-Hb; group 4 cisplatin plus PEG-Hb. 4 weeks later, tumor volume and MVD was significantly decreased in group 4 compared with other groups. Lower expression of HIF-1α and MDR1 were detected in group4. Taken together, our data indicated that PEG-Hb plus cisplatin can promote tumor tissue oxygenation and enhance the chemotherapy sensitivity. HIF-1α regulated MDR1 pathway correlated with this process.
Collapse
Affiliation(s)
- Jianqun Han
- Institute of Microcirculation, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Key Laboratory of Microcirculation Ministry of Health, Beijing, China
| | | | | | | | | | | |
Collapse
|
25
|
Hoogsteen IJ, Marres HAM, van den Hoogen FJA, Rijken PFJW, Lok J, Bussink J, Kaanders JHAM. Expression of EGFR under tumor hypoxia: identification of a subpopulation of tumor cells responsible for aggressiveness and treatment resistance. Int J Radiat Oncol Biol Phys 2012; 84:807-14. [PMID: 22420963 DOI: 10.1016/j.ijrobp.2012.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 12/15/2011] [Accepted: 01/03/2012] [Indexed: 01/14/2023]
Abstract
PURPOSE Overexpression of epidermal growth factor receptor (EGFR) and tumor hypoxia have been shown to correlate with worse outcome in several types of cancer including head-and-neck squamous cell carcinoma. Little is known about the combination and possible interactions between the two phenomena. METHODS AND MATERIALS In this study, 45 cases of histologically confirmed squamous cell carcinomas of the head and neck were analyzed. All patients received intravenous infusions of the exogenous hypoxia marker pimonidazole prior to biopsy. Presence of EGFR, pimonidazole binding, and colocalization between EGFR and tumor hypoxia were examined using immunohistochemistry. RESULTS Of all biopsies examined, respectively, 91% and 60% demonstrated EGFR- and pimonidazole-positive areas. A weak but significant association was found between the hypoxic fractions of pimonidazole (HFpimo) and EGFR fractions (F-EGFR) and between F-EGFR and relative vascular area. Various degrees of colocalization between hypoxia and EGFR were found, increasing with distance from the vasculature. A high fraction of EGFR was correlated with better disease-free and metastasis-free survival, whereas a high degree of colocalization correlated with poor outcome. CONCLUSIONS Colocalization of hypoxia and EGFR was demonstrated in head-and-neck squamous cell carcinomas, predominantly at longer distances from vessels. A large amount of colocalization was associated with poor outcome, which points to a survival advantage of hypoxic cells that are also able to express EGFR. This subpopulation of tumor cells might be indicative of tumor aggressiveness and be partly responsible for treatment resistance.
Collapse
Affiliation(s)
- Ilse J Hoogsteen
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
26
|
Gospodarowicz M. Combination therapy: hypoxia modification with radiotherapy for bladder cancer. Nat Rev Clin Oncol 2011; 8:129-30. [PMID: 21283126 DOI: 10.1038/nrclinonc.2011.5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Mary Gospodarowicz
- Princess Margaret Hospital, University of Toronto, 610 University Avenue, Toronto, ON M5G 2M9, Canada.
| |
Collapse
|
27
|
Yaromina A, Thames H, Zhou X, Hering S, Eicheler W, Dörfler A, Leichtner T, Zips D, Baumann M. Radiobiological hypoxia, histological parameters of tumour microenvironment and local tumour control after fractionated irradiation. Radiother Oncol 2010; 96:116-22. [DOI: 10.1016/j.radonc.2010.04.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 04/07/2010] [Accepted: 04/25/2010] [Indexed: 01/09/2023]
|
28
|
Hoeben BA, Kaanders JH, Franssen GM, Troost EG, Rijken PF, Oosterwijk E, Dongen GAV, Oyen WJ, Boerman OC, Bussink J. PET of Hypoxia with 89Zr-Labeled cG250-F(ab′)2 in Head and Neck Tumors. J Nucl Med 2010; 51:1076-83. [DOI: 10.2967/jnumed.109.073189] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
29
|
Simonsen TG, Gaustad JV, Rofstad EK. Development of hypoxia in a preclinical model of tumor micrometastases. Int J Radiat Oncol Biol Phys 2010; 76:879-88. [PMID: 20159362 DOI: 10.1016/j.ijrobp.2009.09.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 08/27/2009] [Accepted: 09/26/2009] [Indexed: 11/16/2022]
Abstract
PURPOSE Hypoxic regions have been shown to be a characteristic feature of a wide variety of human primary tumors, whereas the oxygenation status of subclinical micrometastases is in general unknown. The development of hypoxia in a xenograft model of microscopic metastases was investigated in this study. METHODS AND MATERIALS U-25-GFP human melanomas growing in dorsal window chamber preparations in BALB/c nu/nu mice were used as a preclinical model of micrometastases. Tumor blood supply time and morphologic parameters of the vascular network were determined from first-pass imaging movies and vascular maps recorded by use of 155-kDa tetramethylrhodamine isothiocyanate-labeled dextran as a vascular tracer. Tumor hypoxia was assessed from immunohistochemical preparations of the imaged tissue by use of pimonidazole as a hypoxia marker. RESULTS Nearly half of the tumors had developed hypoxic regions when they reached a diameter of 2 to 3 mm. Tumors with multiple hypoxic foci showed a low growth rate, low blood flow velocity, high vessel tortuosity, high vessel segment length, and high vascular density, whereas tumors with a single hypoxic region showed a high growth rate, high blood flow velocity, low vessel tortuosity, low vessel segment length, and low vascular density. The tumors with hypoxic regions did not differ from those without hypoxia in any single parameter. CONCLUSIONS U-25-GFP xenograft models of vascularized human tumor micrometastases may develop hypoxic regions as a consequence of two distinctly different morphologic abnormalities in the vascular network: high resistance against blood flow (i.e., high vessel tortuosity and high vessel segment length) or low vascular density.
Collapse
Affiliation(s)
- Trude G Simonsen
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | | | | |
Collapse
|
30
|
Yaromina A, Eckardt A, Zips D, Eicheler W, Schuetze C, Thames H, Baumann M. Core needle biopsies for determination of the microenvironment in individual tumours for longitudinal radiobiological studies. Radiother Oncol 2009; 92:460-5. [DOI: 10.1016/j.radonc.2009.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 07/16/2009] [Accepted: 07/18/2009] [Indexed: 10/20/2022]
|
31
|
The PI3-K/AKT-Pathway and Radiation Resistance Mechanisms in Non-small Cell Lung Cancer. J Thorac Oncol 2009; 4:761-7. [DOI: 10.1097/jto.0b013e3181a1084f] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
32
|
Abstract
Hypoxia imaging has applications in functional recovery in ischemic events such as stroke and myocardial ischemia, but especially in tumors in which hypoxia can be predictive of treatment response and overall prognosis. Recently there has been development of imaging agents utilizing positron emission tomography for non-invasive imaging of hypoxia. Many of these PET agents have come to the forefront of hypoxia imaging. Halogenated PET nitroimidazole imaging agents labeled with (18)F (t(1/2) = 110 m) and (124)I (t(1/2) = 110 m) have been under investigation for the last 25 years, with radiometal agents ((64)Cu-ATSM) being developed more recently. This review focuses on these positron emission tomography imaging agents for hypoxia.
Collapse
Affiliation(s)
- Suzanne E. Lapi
- Assistant Professor, Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO
| | - Thomas F. Voller
- Research Laboratory Manager, Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO
| | - Michael J. Welch
- Professor, Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO
| |
Collapse
|
33
|
Yu M, Han J, Dai M, Cui P, Li H, Liu Q, Xiu R. Influence of PEG-conjugated hemoglobin on tumor oxygenation and response to chemotherapy. ACTA ACUST UNITED AC 2008; 36:551-61. [PMID: 19065307 DOI: 10.1080/10731190802556674] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Hypoxic tumors are significantly more malignant, metastatic, radio- and chemoresistant. The use of artificial oxygen carriers represents a new approach to the problem of hypoxia. In the present study, female athymic BALB/c nude mice bearing the cervical carcinoma were untreated or treated with cisplatin to determine whether administration of artificial oxygen carrier (PEG-conjugated Hemoglobin, PEG-Hb) could improve the tumor oxygenation and enhance the anti-tumor efficacy of cisplatin. Pimonidazole staining was employed to detect tumor tissue oxygenation status. We found that the application of a higher dose (0.6 g/kg) PEG-Hb could significantly ameliorate the hypoxic condition in cervical carcinoma xenograft models. Co-administration of PEG-Hb (0.6 g/kg) with cisplatin produced significant tumor growth inhibition and pro-apoptotic and anti-proliferative effects as compared to cisplatin alone. These suggest the evaluated PEG-Hb in this experiment has positive effects on cisplatin or cisplatin-based chemotherapy, and further work to optimize its application is warranted.
Collapse
Affiliation(s)
- Minghua Yu
- Institute of Microcirculation, Peking Union Medical College & Chinese Academy of Medical Sciences, 5 Dong Dan San Tiao, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Isohashi F, Endo H, Mukai M, Inoue T, Inoue M. Insulin-like growth factor stimulation increases radiosensitivity of a pancreatic cancer cell line through endoplasmic reticulum stress under hypoxic conditions. Cancer Sci 2008; 99:2395-401. [PMID: 19018773 PMCID: PMC11159351 DOI: 10.1111/j.1349-7006.2008.00970.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Revised: 08/04/2008] [Accepted: 08/13/2008] [Indexed: 01/22/2023] Open
Abstract
Tumor hypoxia is an obstacle to radiotherapy. Radiosensitivity under hypoxic conditions is determined by molecular oxygen levels, as well as by various biological cellular responses. The insulin-like growth factor (IGF) signaling pathway is a widely recognized survival signal that confers radioresistance. However, under hypoxic conditions the role of IGF signaling in radiosensitivity is still poorly understood. Here, we demonstrate that IGF-II stimulation decreases clonogenic survival under hypoxic conditions in the pancreatic cancer cell lines AsPC-1 and Panc-1, and in the human breast cancer cell line MCF-7. IGF treatment under hypoxic conditions suppressed increased radiation sensitivity in these cell lines by pharmacologically inhibiting the phosphoinositide 3-kinase-mammalian target of rapamycin pathway, a major IGF signal-transduction pathway. Meanwhile, IGF-II induced the endoplasmic reticulum stress response under hypoxia, including increased protein levels of CHOP and ATF4, mRNA levels of CHOP, GADD34, and BiP, as well as splicing levels of XBP-1. The response was suppressed by inhibiting phosphoinositide 3-kinase and mammalian target of rapamycin activity. Overexpression of CHOP in AsPC-1 cells increased radiation sensitivity by IGF-II simulation under hypoxic conditions, whereas suppression of CHOP expression levels with small hairpin RNA or a dominant negative form of a proline-rich extensin-like receptor protein kinase in hypoxia decreased IGF-induced radiosensitivity. IGF-induced endoplasmic reticulum stress contributed to radiosensitization independent of cell cycle status. Taken together, IGF stimulation increased radiosensitivity through the endoplasmic reticulum stress response under hypoxic conditions.
Collapse
Affiliation(s)
- Fumiaki Isohashi
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka 537-8511, Japan
| | | | | | | | | |
Collapse
|
35
|
Wijffels KIEM, Hoogsteen IJ, Lok J, Rijken PFJW, Marres HAM, de Wilde PCM, van der Kogel AJ, Kaanders JHAM. No detectable hypoxia in malignant salivary gland tumors: preliminary results. Int J Radiat Oncol Biol Phys 2008; 73:1319-25. [PMID: 18973979 DOI: 10.1016/j.ijrobp.2008.06.1927] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 06/23/2008] [Accepted: 06/23/2008] [Indexed: 10/21/2022]
Abstract
PURPOSE Hypoxia is detected in most solid tumors and is associated with malignant progression and adverse treatment outcomes. However, the oxygenation status of malignant salivary gland tumors has not been previously studied. The aim of this study was to investigate the potential clinical relevance of hypoxia in this tumor type. METHODS AND MATERIALS Twelve patients scheduled for surgical resection of a salivary gland tumor were preoperatively injected with the hypoxia marker pimonidazole and the proliferation marker iododeoxyuridine. Tissue samples of the dissected tumor were immunohistochemically stained for blood vessels, pimonidazole, carbonic anhydrase-IX, glucose transporters-1 and -3 (Glut-1, Glut-3), hypoxia-inducible factor-1alpha, iododeoxyuridine, and epidermal growth factor receptor. The tissue sections were quantitatively assessed by computerized image analysis. RESULTS The tissue material from 8 patients was of sufficient quality for quantitative analysis. All tumors were negative for pimonidazole binding, as well as for carbonic anhydrase-IX, Glut-1, Glut-3, and hypoxia-inducible factor-1alpha. The vascular density was high, with a median value of 285 mm(-2) (range, 209-546). The iododeoxyuridine-labeling index varied from <0.1% to 12.2% (median, 2.2%). Epidermal growth factor receptor expression levels were mostly moderate to high. In one-half of the cases, nuclear expression of epidermal growth factor receptor was observed. CONCLUSION The absence of detectable pimonidazole binding, as well as the lack of expression of hypoxia-associated proteins in all tumors, indicates that malignant salivary gland tumors are generally well oxygenated. It is unlikely that hypoxia is a relevant factor for their clinical behavior and treatment responsiveness.
Collapse
Affiliation(s)
- Karien I E M Wijffels
- Department of Otorhinolaryngology, Head and Neck Surgery, Deventer Hospital, Deventer, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Investigation of the influence of carbon dioxide concentrations on cerebral physiology by susceptibility-weighted magnetic resonance imaging (SWI). Neuroimage 2008; 43:36-43. [PMID: 18678260 DOI: 10.1016/j.neuroimage.2008.07.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 06/26/2008] [Accepted: 07/01/2008] [Indexed: 01/31/2023] Open
|
37
|
Aarts F, Bleichrodt RP, Oyen WJG, Boerman OC. Intracavitary radioimmunotherapy to treat solid tumors. Cancer Biother Radiopharm 2008; 23:92-107. [PMID: 18298333 DOI: 10.1089/cbr.2007.0412] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Radioimmunotherapy (RIT) potentially is an attractive treatment for radiosensitive early-stage solid tumors and as an adjuvant to cytoreductive surgery. Topical administration of RIT may improve the efficacy because higher local concentrations are achieved. We reviewed the results of locally applied radiolabeled monoclonal antibodies for the treatment of solid tumors. Intracavitary RIT in patients with ovarian cancer and glioma showed improved targeting after local administration, as compared to the intravenous administration. In addition, various studies showed the feasibility of locally applied RIT in these patients. In studies that included patients with small-volume disease, adjuvant RIT in ovarian cancer and glioma showed to be at least as effective as standard therapy. The information about RIT for peritoneal carcinomatosis of colorectal origin is scarce, while results from preclinical data are promising. RIT may be applied for other, relatively unexplored indications. Studies on the application of radiolabeled antibodies in early urothelial cell cancer have been performed, showing that intracavitary RIT may hold a promise. Moreover, in patients with malignant pleural mesothelioma or malignant pleural effusion, RIT may play a role in the palliative treatment. Intracavitary RIT limits toxicity and improves tumor targeting. RIT is more effective in patients with small-volume disease of solid cancers. RIT may have potential for palliation in patients with malignant pleural mesothelioma or malignant pleural effusion. The future of RIT may, therefore, not only be in the inclusion in contemporary multimodality treatment, but also in the expansion to palliative treatment.
Collapse
Affiliation(s)
- Frits Aarts
- Department of Surgery, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
38
|
Activation of the PI3-K/AKT pathway and implications for radioresistance mechanisms in head and neck cancer. Lancet Oncol 2008; 9:288-96. [DOI: 10.1016/s1470-2045(08)70073-1] [Citation(s) in RCA: 239] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
39
|
Sonveaux P. Provascular strategy: Targeting functional adaptations of mature blood vessels in tumors to selectively influence the tumor vascular reactivity and improve cancer treatment. Radiother Oncol 2008; 86:300-13. [PMID: 18313779 DOI: 10.1016/j.radonc.2008.01.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 01/30/2008] [Accepted: 01/30/2008] [Indexed: 12/22/2022]
|
40
|
Kisilevsky M, Hudson C, Mardimae A, Wong T, Fisher J. Concentration-dependent vasoconstrictive effect of hyperoxia on hypercarbia-dilated retinal arterioles. Microvasc Res 2008; 75:263-8. [DOI: 10.1016/j.mvr.2007.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 06/28/2007] [Accepted: 07/24/2007] [Indexed: 10/22/2022]
|
41
|
|
42
|
Hoh DJ, Liu CY, Chen JC, Pagnini PG, Yu C, Wang MY, Apuzzo ML. CHAINED LIGHTNING. Neurosurgery 2007; 61:1111-29; discussion 1129-30. [DOI: 10.1227/01.neu.0000306089.22894.4e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Abstract
RADIOSURGERY IS FUNDAMENTALLY the harnessing of energy and delivering it to a focal target for a therapeutic effect. The evolution of radiosurgical technology and practice has served toward refining methodologies for better conformal energy delivery. In the past, this has resulted in developing strategies for improved beam generation and delivery. Ultimately, however, our current instrumentation and treatment modalities may be approaching a practical limit with regard to further optimizing energy containment.
In looking forward, several strategies are emerging to circumvent these limitations and improve conformal radiosurgery. Refinement of imaging techniques through functional imaging and nanoprobes for cancer detection may benefit lesion localization and targeting. Methods for enhancing the biological effect while reducing radiation-induced changes are being examined through dose fractionation schedules. Radiosensitizers and photosensitizers are being investigated as agents for modulating the biological response of tissues to radiation and alternative energy forms. Discovery of new energy modalities is being pursued through development of microplanar beams, free electron lasers, and high-intensity focused ultrasound. The exploration of these future possibilities will provide the tools for radiosurgical treatment of a broader spectrum of diseases for the next generation.
Collapse
Affiliation(s)
- Daniel J. Hoh
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Charles Y. Liu
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Joseph C.T. Chen
- Departments of Radiation Oncology and Neurological Surgery, Southern California Permanente Medical Group, Los Angeles, California
| | - Paul G. Pagnini
- Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Cheng Yu
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Michael Y. Wang
- Miller School of Medicine, University of Miami, Miami, Florida
| | - Michael L.J. Apuzzo
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
43
|
Abstract
Since observations from the beginning of the last century, it has become well established that solid tumors may contain oxygen-deficient hypoxic areas and that cells in such areas may cause tumors to become radioresistant. Identifying hypoxic cells in human tumors has improved by the help of new imaging and physiologic techniques, and a substantial amount of data indicates the presence of hypoxia in many types of human tumors, although with a considerable heterogeneity among individual tumors. Controlled clinical trials during the last 40 years have indicated that this source of radiation resistance can be eliminated or modified by normobaric or hyperbaric oxygen or by the use of nitroimidazoles as hypoxic radiation sensitizers. More recently, attention has been given to hypoxic cytotoxins, a group of drugs that selectively or preferably destroys cells in a hypoxic environment. An updated systematic review identified 10,108 patients in 86 randomized trials designed to modify tumor hypoxia in patients treated with curative attempted primary radiation therapy alone. Overall modification of tumor hypoxia significantly improved the effect of radiotherapy, with an odds ratio of 0.77 (95% CI, 0.71 to 0.86) for the outcome of locoregional control and with an associated significant overall survival benefit (odds ratio = 0.87; 95% CI, 0.80 to 0.95). No significant influence was found on the incidence of distant metastases or on the risk of radiation-related complications. Ample data exist to support a high level of evidence for the benefit of hypoxic modification. However, hypoxic modification still has no impact on general clinical practice.
Collapse
Affiliation(s)
- Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus C, Denmark.
| |
Collapse
|
44
|
Wijffels KIEM, Marres HAM, Peters JPW, Rijken PFJW, van der Kogel AJ, Kaanders JHAM. Tumour cell proliferation under hypoxic conditions in human head and neck squamous cell carcinomas. Oral Oncol 2007; 44:335-44. [PMID: 17689286 DOI: 10.1016/j.oraloncology.2007.04.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 02/15/2007] [Accepted: 04/18/2007] [Indexed: 11/17/2022]
Abstract
Two mechanisms of radiotherapy resistance of major importance in head and neck cancer are tumour cell repopulation and hypoxia. Hypoxic tumour cells that retain their clonogenic potential can survive radiation treatment and lead to local recurrences. The aim of this study was to quantify this cellular population in a cohort of human head and neck carcinomas and to investigate the prognostic significance. The proliferation marker iododeoxyuridine (IdUrd) and the hypoxia marker pimonidazole were administered intravenously prior to biopsy taking in patients with stage II-IV squamous cell carcinoma of the head and neck. Triple immunohistochemical staining of blood vessels, IdUrd and pimonidazole was performed and co-localization of IdUrd and pimonidazole was quantitatively assessed by computerized image analysis. The results were related with treatment outcome. Thirty-nine biopsies were analyzed. Tumours exhibited different patterns of proliferation and hypoxia but generally the IdUrd signal was found in proximity to blood vessels whereas pimonidazole binding was predominantly at a distance from vessels. Overall, no correlations were found between proliferative activity and oxygenation status. The fraction of IdUrd-labelled cells positive for pimonidazole ranged from 0% to 16.7% with a mean of 2.4% indicating that proliferative activity was low in hypoxic areas and occurring mainly in the well-oxygenated tumour compartments. IdUrd positive cells in hypoxic areas made up only 0.09% of the total viable tumour cell mass. There were no associations between the magnitude of this cell population and local tumour control or survival. Co-localization between proliferating cells and hypoxia in head and neck carcinomas was quantified using an immunohistochemical triple staining technique combined with a computerized simultaneous analysis of multiple parameters. The proportion of cells proliferating under hypoxic conditions was small and no correlation with treatment outcome could be found.
Collapse
Affiliation(s)
- Karien I E M Wijffels
- Department of Otorhinolaryngology, Head and Neck Surgery, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
45
|
Cairns RA, Papandreou I, Sutphin PD, Denko NC. Metabolic targeting of hypoxia and HIF1 in solid tumors can enhance cytotoxic chemotherapy. Proc Natl Acad Sci U S A 2007; 104:9445-50. [PMID: 17517659 PMCID: PMC1890514 DOI: 10.1073/pnas.0611662104] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2006] [Indexed: 12/26/2022] Open
Abstract
Solid tumors frequently contain large regions with low oxygen concentrations (hypoxia). The hypoxic microenvironment induces adaptive changes to tumor cell metabolism, and this alteration can further distort the local microenvironment. The net result of these tumor-specific changes is a microenvironment that inhibits many standard cytotoxic anticancer therapies and predicts for a poor clinical outcome. Pharmacologic targeting of the unique metabolism of solid tumors could alter the tumor microenvironment to provide more favorable conditions for anti-tumor therapy. Here, we describe a strategy in which the mitochondrial metabolism of tumor cells is increased by pharmacologic inhibition of hypoxia-inducible factor 1 (HIF1) or its target gene pyruvate dehydrogenase kinase 1 (PDK1). This acute increase in oxygen consumption leads to a corresponding decrease in tumor oxygenation. Whereas decreased oxygenation could reduce the effectiveness of some traditional therapies, we show that it dramatically increases the effectiveness of a hypoxia-specific cytotoxin. This treatment strategy should provide a high degree of tumor specificity for increasing the effectiveness of hypoxic cytotoxins, as it depends on the activation of HIF1 and the presence of hypoxia, conditions that are present only in the tumor, and not the normal tissue.
Collapse
Affiliation(s)
- Rob A. Cairns
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305
| | - Ioanna Papandreou
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305
| | - Patrick D. Sutphin
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305
| | - Nicholas C. Denko
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
46
|
Nishimura Y, Nakagawa K, Takeda K, Tanaka M, Segawa Y, Tsujino K, Negoro S, Fuwa N, Hida T, Kawahara M, Katakami N, Hirokawa K, Yamamoto N, Fukuoka M, Ariyoshi Y. Phase I/II trial of sequential chemoradiotherapy using a novel hypoxic cell radiosensitizer, doranidazole (PR-350), in patients with locally advanced non-small-cell lung Cancer (WJTOG-0002). Int J Radiat Oncol Biol Phys 2007; 69:786-92. [PMID: 17512126 DOI: 10.1016/j.ijrobp.2007.04.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2006] [Revised: 03/31/2007] [Accepted: 04/03/2007] [Indexed: 10/23/2022]
Abstract
PURPOSE This Phase I/II trial was conducted to assess the efficacy and safety of PR-350, a novel hypoxic cell radiosensitizer, when administered with thoracic radiation therapy (RT) after induction chemotherapy (CT) for locally advanced non-small-cell lung cancer (NSCLC). METHODS AND MATERIALS Two cycles of cisplatin (80 mg/m(2)) and paclitaxel (180 mg/m(2)), or carboplatin (AUC = 6) and paclitaxel (200 mg/m(2)) were given before RT of 60 Gy in 30 fractions. In the Phase I portion, the starting dosage of PR-350 was 10 daily administrations (2000 mg/m(2)) in combination with RT, and this number was increased in increments of 10 for successive groups to 30 doses. RESULTS In total, 37 patients were enrolled. In Phase I (n = 20), PR-350 could be administered 30 times with concurrent thoracic RT. Thus, in Phase II (n = 17), PR-350 was administered 30 times. The major toxicity was radiation pneumonitis, with Grade 3 or more pneumonitis noted in 6 patients (16%) including 2 with treatment-related deaths. However, no Grade 3 or more esophageal toxicity was noted, and only Grade 1 peripheral neuropathy was noted in 9 patients (24%). For all 37 patients, the median survival time (MST) and the 2-year survival rate were 15.9 months and 24%, respectively. For 18 patients receiving 21 to 30 doses of PR-350, the MST and 2-year survival rate were 20.9 months and 33%, respectively. CONCLUSIONS Thoracic RT combined with 30 daily administrations of PR-350 after induction CT was well tolerated and promising for locally advanced NSCLC.
Collapse
Affiliation(s)
- Yasumasa Nishimura
- Department of Radiation Oncology, Kinki University School of Medicine, Osaka-Sayama, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Corvò R. Evidence-based radiation oncology in head and neck squamous cell carcinoma. Radiother Oncol 2007; 85:156-70. [PMID: 17482300 DOI: 10.1016/j.radonc.2007.04.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Revised: 03/26/2007] [Accepted: 04/04/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND AND PURPOSE Historically, radiation therapy (RT) has been an available treatment option for patients with early resectable head and neck squamous cell carcinoma (HNSCC) and the sole therapy for those with unresectable or inoperable disease. Recently, four noteworthy strategies have emerged for the improvement of therapeutic outcome in the curative treatment of HNSCC: they include the development of altered fractionation radiotherapy, integration of chemotherapy with radiotherapy, incorporation of intensity-modulated radiotherapy and the introduction of targeted biological therapy. These strategies are briefly reviewed in an effort to help interpret evidence-based data and to facilitate clinical-decision making in a clinical context. MATERIALS AND METHODS For patients with early stage HNSCC no level 1 study exists in which radiation therapy is compared with conservative surgery for the evaluation of local control or survival. Only evidence from prospective and retrospective cohort studies is available to evaluate the role external radiotherapy and/or brachytherapy currently play in limited disease. For patients with locally advanced HNSCC the recommendations to address the questions about better treatment in resectable and unresectable tumors are based on more than 100 randomized Phase III trials included in six meta-analyses on chemo-radiotherapy and/or altered fractionation. Data from phase II trials and cohort studies help interpret the advances in intensity-modulated radiotherapy. RESULTS External radiotherapy and/or brachytherapy are crucial treatment options in patients with early stage HNSCC. For patients with locally advanced HNSCC, where outcome with conventional radiotherapy is poor, meta-analyses and collective data showed that loco-regional control may be improved at high level of evidence by altered fractionation radiotherapy, chemo-radiotherapy with concomitant approach or association of selected hypoxic cell radiosensitizer with radiotherapy. For these patients, overall survival may be improved at high level of evidence by concomitant chemo-radiotherapy or hyperfractionated RT delivered with increased total dose. Also EGFR-inhibitors (cetuximab)-radiotherapy strategy offers at a lower level of evidence better loco-regional control and overall survival than radiotherapy alone. Chemo-radiotherapy programs can achieve an improved larynx-function preservation program without the risk of overall survival reduction, for patients with larynx or hypopharynx tumors who are candidates to radical surgery followed by radiotherapy. Recently, strong evidence for an improved outcome for high-risk resected patients has been shown by the use of adjuvant concomitant chemo-radiotherapy. Despite improved results, a higher severe toxicity has been largely evidenced with concomitant chemo-radiotherapy by reducing the gain in the therapeutic index with new treatment strategies. Three-dimensional conformal radiotherapy is the minimal standard of technique in HNSCC: however, as advances are promising, intensity-modulated radiotherapy should be largely implemented. CONCLUSIONS Stepwise improvements in HNSCC non-surgical therapy have shown favorable impact on loco-regional control and overall survival. However, despite hundreds of clinical trials in patients with advanced disease, there is no absolute consensus about patient selection for altered fractionation regimens, type of chemo-radiotherapy association, radiation or chemotherapy dose schedule. Nevertheless, many well-conducted clinical studies have expanded therapy options besides standard radiotherapy and have contributed to defining the evolving standard of care for patients with HNSCC.
Collapse
Affiliation(s)
- Renzo Corvò
- Department of Radiation Oncology, National Cancer Research Institute and University, Genova, Italy.
| |
Collapse
|
48
|
Wardman P. Chemical radiosensitizers for use in radiotherapy. Clin Oncol (R Coll Radiol) 2007; 19:397-417. [PMID: 17478086 DOI: 10.1016/j.clon.2007.03.010] [Citation(s) in RCA: 318] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Accepted: 03/13/2007] [Indexed: 12/21/2022]
Abstract
Radiosensitizers are intended to enhance tumour cell killing while having much less effect on normal tissues. Some drugs target different physiological characteristics of the tumour, particularly hypoxia associated with radioresistance. Oxygen is the definitive hypoxic cell radiosensitizer, the large differential radiosensitivity of oxic vs hypoxic cells being an attractive factor. The combination of nicotinamide to reduce acute hypoxia with normobaric carbogen breathing is showing clinical promise. 'Electron-affinic' chemicals that react with DNA free radicals have the potential for universal activity to combat hypoxia-associated radioresistance; a nitroimidazole, nimorazole, is clinically effective at tolerable doses. Hypoxia-specific cytotoxins, such as tirapazamine, are valuable adjuncts to radiotherapy. Nitric oxide is a potent hypoxic cell radiosensitizer; variations in endogenous levels might have prognostic significance, and routes to deliver nitric oxide specifically to tumours are being developed. In principle, many drugs can be delivered selectively to hypoxic tumours using either reductase enzymes or radiation-produced free radicals to activate drug release from electron-affinic prodrugs. A redox-active agent based on a gadolinium chelate is being evaluated clinically. Pyrimidines substituted with bromine or iodine are incorporated into DNA and enhance free radical damage; fluoropyrimidines act by different mechanisms. A wide variety of drugs that influence the nature or repair of DNA damage are being evaluated in conjunction with radiation; it is often difficult to define the mechanisms underlying chemoradiation regimens. Drugs being evaluated include topoisomerase inhibitors (e.g. camptothecin, topotecan), and the hypoxia-activated anthraquinone AQ4N; alkylating agents include temozolomide. Drugs involved in DNA repair pathways being investigated include the potent poly(ADP ribose)polymerase inhibitor, AG14,361. Proteins involved in cell signalling, such as the Ras family, are attractive targets linked to radioresistance, as are epidermal growth factor receptors and linked kinases (drugs including vandetanib [ZD6,474], cetuximab and gefitinib), and cyclooxygenase-2 (celecoxib). The suppression of radioprotective thiols seems to offer more potential with alkylating agents than with radiotherapy, although it remains a strategy worthy of exploration.
Collapse
Affiliation(s)
- P Wardman
- University of Oxford, Gray Cancer Institute, PO Box 100, Mount Vernon Hospital, Northwood HA6 2JR, UK.
| |
Collapse
|
49
|
Hoogsteen IJ, Marres HAM, van der Kogel AJ, Kaanders JHAM. The hypoxic tumour microenvironment, patient selection and hypoxia-modifying treatments. Clin Oncol (R Coll Radiol) 2007; 19:385-96. [PMID: 17433637 DOI: 10.1016/j.clon.2007.03.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 03/02/2007] [Indexed: 01/18/2023]
Abstract
Tumour hypoxia has been found to be a characteristic feature in many solid tumours. It has been shown to decrease the therapeutic efficacy of radiation treatment, surgery and some forms of chemotherapy. Successful approaches have been developed to counteract this resistance mechanism, although usually at the cost of increased short- and long-term side-effects. New methods for qualitative and quantitative assessment of tumour oxygenation have made it possible to establish the prognostic significance of tumour hypoxia. The ability to determine the degree and extent of hypoxia in solid tumours is not only important prognostically, but also in the selection of patients for hypoxia-modifying treatments. To provide the best attainable quality of life for individual patients it is of increasing importance that tools be developed that allow a better selection of patients for these intensified treatment strategies. Several genes and proteins involved in the response to hypoxia have been identified as potential candidates for future use in predictive assays. Although some markers and combinations have shown potential benefit and are associated with treatment outcome, their clinical usefulness needs to be validated in prospective trials. A review of published studies was carried out, focusing on the assessment of tumour hypoxia, patient selection and the possibilities to overcome hypoxia during treatment.
Collapse
Affiliation(s)
- I J Hoogsteen
- Department of Radiation Oncology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
50
|
Nagasawa H, Uto Y, Kirk KL, Hori H. Design of hypoxia-targeting drugs as new cancer chemotherapeutics. Biol Pharm Bull 2007; 29:2335-42. [PMID: 17142959 DOI: 10.1248/bpb.29.2335] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The tumor microenvironment is now recognized as a major factor that influences not only the response to conventional anti-cancer therapies but also helps define the potential for malignant progression and metastasis. In particular, hypoxia is now considered a fundamentally important characteristic of the tumor microenvironment. Furthermore, discovery of the hypoxia inducible factor 1alpha (HIF-1alpha) has led to a rapidly increasing understanding of the molecular mechanisms involved in tumor hypoxia. This in turn has led to the current extensive interest in the signal molecules related to tumor hypoxia as potential molecular targets for cancer therapeutics. In this paper we give an overview of recent advances in hypoxia research, including cancer treatments that target tumor hypoxia. Progress in the development of hypoxia-targeting drugs will be discussed, including antiangiogenic hypoxic cell radiosensitizers and hypoxic cytotoxins, hypoxia targeting boron carriers and p53-inhibiting bifunctional radiosensitizers. We will also review our own recent research results in these areas. For example, we have found that certain of the 2-nitroimidazole radiosensitizers and heterocycle-N-oxide hypoxic cytotoxins we developed have antiangiogenic activity and antimetastatic activity. We propose that these activities are based on the inhibition of signal transduction mediated by HIF-1alpha. The anti-tumor activities of hypoxia response are considered to be cytostatic (tumor dormancy-inducing) effects in contrast to cytotoxic DNA damaging effects. The combination of these cytostatic effects that are related to radiosensitization with the cytotoxic effects of radiation should improve the prognosis and QOL of patients receiving radiation and lead to an overall response to treatment. Based on these considerations, we developed the antiangiogenic hypoxic cell radiosensitizers, TX-1877, TX-1898 and the hypoxic cytotoxin TX-402 that inhibits the HIF-1alpha pathway We will also discuss our research involved with the development of other drugs to exploit tumor hypoxia, including a hypoxia-targeting boron carrier for boron neutron capture therapy (BNCT) and a p53 inhibiting radiosensitizer.
Collapse
Affiliation(s)
- Hideko Nagasawa
- Laboratory of Pharmaceutical Chemistry, Gifu Pharmaceutical University, Japan.
| | | | | | | |
Collapse
|