1
|
Cheng W, He S, Chen Q, Song X, Lu C, Yang H. X-ray Induced Persistent Type I Photodynamic Therapy with Enhanced Hypoxia Tolerance and Chemoradiotherapy. NANO LETTERS 2025; 25:4549-4559. [PMID: 40062435 DOI: 10.1021/acs.nanolett.5c00433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
The hypoxic tumor microenvironment (TME), inadequate penetration depth of Vis/NIR light, and lack of sustaining reactive oxygen species (ROS) production capability of photosensitizers pose significant obstacles to the widespread clinic applications of photodynamic therapy (PDT). Herein, we developed a "persistent type I X-PDT" platform to simultaneously overcome these three limitations. Such a nanoplatform could generate efficient ROS (•OH and O2•-) under X-ray irradiation in both normoxic and hypoxic environments. The ROS production persists in tumor cells for more than 4 h, even after the X-ray source is removed. Notably, the persistent type I X-PDT does not increase the levels of hypoxia-inducible factor-1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF) in tumor cells both in vitro and in vivo. Moreover, to further enhance the radiotherapy efficacy in hypoxic conditions, a Pt (IV) prodrug was also introduced, which can be reduced to cisplatin selectively in tumor cells, functioning not only as a chemodrug but also as a radiosensitizer.
Collapse
Affiliation(s)
- Wei Cheng
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore
| | - Shuai He
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Qiushui Chen
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Xiaorong Song
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Chunhua Lu
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Huanghao Yang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
2
|
Chen MH, Yiu HP, Wang YC, Liu TY, Li C. Multifunctional Nanoparticles as Radiosensitizers to Overcome Hypoxia-Associated Resistance in Cancer Radiotherapy. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 15:37. [PMID: 39791794 PMCID: PMC11723374 DOI: 10.3390/nano15010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/12/2025]
Abstract
Hypoxia, a phenomenon that occurs when the oxygen level in tissues is lower than average, is commonly observed in human solid tumors. For oncological treatment, the hypoxic environment often results in radioresistance and chemoresistance. In this study, a new multifunctional oxygen carrier, carboxymethyl hexanoyl chitosan (CHC) nanodroplets decorated with perfluorohexane (PFH) and superparamagnetic iron oxide (SPIO) nanodroplets (SPIO@PFH-CHC), was developed and investigated. PFH-based oxygen carriers can augment oxygenation within tumor tissues, thereby mitigating radioresistance. Concurrently, oxygenation can cause deoxyribonucleic acid (DNA) damage via oxygen fixation and consequently suppress cancer cell proliferation. Moreover, these pH-sensitive nanodroplets allow higher cellular uptake with minimal cytotoxicity. Two distinctive mechanisms of SPIO@PFH-CHC nanodroplets were found in this study. The SPIO nanoparticles of the SPIO@PFH-CHC nanodroplets can generate hydroxyl radicals (HO•) and other reactive oxygen species (ROS), which is vital to chemodynamic therapy (CDT) via the Fenton reaction. Meanwhile, the higher X-ray absorption among these nanodroplets leads to a local energy surge and causes more extensive deoxyribonucleic acid (DNA) damage via oxygen fixation. This study demonstrates that low cytotoxic SPIO@PFH-CHC nanodroplets can be an efficient radiosensitizer for radiation therapy.
Collapse
Affiliation(s)
- Ming-Hong Chen
- Division of Neurosurgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan;
- Department of Electrical Engineering, Yuan Ze University, Taoyuan City 320315, Taiwan
| | - Hon-Pan Yiu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (H.-P.Y.); (Y.-C.W.)
| | - Yu-Chi Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (H.-P.Y.); (Y.-C.W.)
| | - Tse-Ying Liu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (H.-P.Y.); (Y.-C.W.)
| | - Chuan Li
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (H.-P.Y.); (Y.-C.W.)
| |
Collapse
|
3
|
Ostapowicz J, Ostrowska K, Golusiński W, Kulcenty K, Suchorska WM. Improving therapeutic strategies for Head and Neck Cancer: Insights from 3D hypoxic cell culture models in treatment response evaluation. Adv Med Sci 2024; 69:368-376. [PMID: 39047970 DOI: 10.1016/j.advms.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/04/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Hypoxia in the tumor core negatively affects the outcome of patients with head and neck squamous cell carcinoma (HNSCC). Nevertheless, its role in predicting treatment response requires further exploration. Typically, reduced oxygen levels in the tumor core correlate with diminished efficacy of radiotherapy, chemotherapy, and immunotherapy, which are commonly used for HNSCC patients' treatment. Understanding the mechanistic underpinnings of these varied treatment responses in HNSCC is crucial for enhancing therapeutic outcomes and extending patients' overall survival (OS) rates. Standard monolayer cell culture conditions have major limitations in mimicking tumor physiological features and the complexity of the tumor microenvironment. Three-dimensional (3D) cell cultures enable the recreation of the in vivo tumor attributes, encompassing oxygen and nutrient gradients, cellular morphology, and intracellular connections. It is vital to use the 3D model in treatment response studies to mimic the tumor microenvironment, as evidenced by the decreased sensitivity of 3D structures to anticancer therapy. Accordingly, the aim of the study was to delineate the utility of the 3D models of hypoxic head and neck tumors in drug screening and treatment response studies.
Collapse
Affiliation(s)
- Julia Ostapowicz
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland; Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland; Doctoral School, Poznan University of Medical Sciences, Poznan, Poland.
| | - Kamila Ostrowska
- Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland; Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznan, Poland
| | - Wojciech Golusiński
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznan, Poland
| | - Katarzyna Kulcenty
- Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland
| | - Wiktoria M Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland; Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
4
|
Kaanders JHAM, Bussink J, Aarntzen EHJG, Braam P, Rütten H, van der Maazen RWM, Verheij M, van den Bosch S. [18F]FDG-PET-Based Personalized Radiotherapy Dose Prescription. Semin Radiat Oncol 2023; 33:287-297. [PMID: 37331783 DOI: 10.1016/j.semradonc.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
PET imaging with 2'-deoxy-2'-[18F]fluoro-D-glucose ([18F]FDG) has become one of the pillars in the management of malignant diseases. It has proven value in diagnostic workup, treatment policy, follow-up, and as prognosticator for outcome. [18F]FDG is widely available and standards have been developed for PET acquisition protocols and quantitative analyses. More recently, [18F]FDG-PET is also starting to be appreciated as a decision aid for treatment personalization. This review focuses on the potential of [18F]FDG-PET for individualized radiotherapy dose prescription. This includes dose painting, gradient dose prescription, and [18F]FDG-PET guided response-adapted dose prescription. The current status, progress, and future expectations of these developments for various tumor types are discussed.
Collapse
Affiliation(s)
- Johannes H A M Kaanders
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands..
| | - Johan Bussink
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
| | - Pètra Braam
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Heidi Rütten
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Marcel Verheij
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Sven van den Bosch
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
5
|
Nguyen AT, Kim HK. Recent Developments in PET and SPECT Radiotracers as Radiopharmaceuticals for Hypoxia Tumors. Pharmaceutics 2023; 15:1840. [PMID: 37514026 PMCID: PMC10385036 DOI: 10.3390/pharmaceutics15071840] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Hypoxia, a deficiency in the levels of oxygen, is a common feature of most solid tumors and induces many characteristics of cancer. Hypoxia is associated with metastases and strong resistance to radio- and chemotherapy, and can decrease the accuracy of cancer prognosis. Non-invasive imaging methods such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) using hypoxia-targeting radiopharmaceuticals have been used for the detection and therapy of tumor hypoxia. Nitroimidazoles are bioreducible moieties that can be selectively reduced under hypoxic conditions covalently bind to intracellular macromolecules, and are trapped within hypoxic cells and tissues. Recently, there has been a strong motivation to develop PET and SPECT radiotracers as radiopharmaceuticals containing nitroimidazole moieties for the visualization and treatment of hypoxic tumors. In this review, we summarize the development of some novel PET and SPECT radiotracers as radiopharmaceuticals containing nitroimidazoles, as well as their physicochemical properties, in vitro cellular uptake values, in vivo biodistribution, and PET/SPECT imaging results.
Collapse
Affiliation(s)
- Anh Thu Nguyen
- Department of Nuclear Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Hee-Kwon Kim
- Department of Nuclear Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
6
|
Nguyen AT, Kim HK. Recent Advances of 68Ga-Labeled PET Radiotracers with Nitroimidazole in the Diagnosis of Hypoxia Tumors. Int J Mol Sci 2023; 24:10552. [PMID: 37445730 DOI: 10.3390/ijms241310552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging method extensively applied in the detection and treatment of various diseases. Hypoxia is a common phenomenon found in most solid tumors. Nitroimidazole is a group of bioreducible pharmacophores that selectively accumulate in hypoxic regions of the body. Over the past few decades, many scientists have reported the use of radiopharmaceuticals containing nitroimidazole for the detection of hypoxic tumors. Gallium-68, a positron-emitting radioisotope, has a favorable half-life time of 68 min and can be conveniently produced by 68Ge/68Ga generators. Recently, there has been significant progress in the preparation of novel 68Ga-labeled complexes bearing nitroimidazole moieties for the diagnosis of hypoxia. This review provides a comprehensive overview of the current status of developing 68Ga-labeled radiopharmaceuticals with nitroimidazole moieties, their pharmacokinetics, and in vitro and in vivo studies, as well as PET imaging studies for hypoxic tumors.
Collapse
Affiliation(s)
- Anh Thu Nguyen
- Department of Nuclear Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Hee-Kwon Kim
- Department of Nuclear Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
7
|
Kerr CP, Grudzinski JJ, Nguyen TP, Hernandez R, Weichert JP, Morris ZS. Developments in Combining Targeted Radionuclide Therapies and Immunotherapies for Cancer Treatment. Pharmaceutics 2022; 15:128. [PMID: 36678756 PMCID: PMC9865370 DOI: 10.3390/pharmaceutics15010128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Targeted radionuclide therapy (TRT) and immunotherapy are rapidly growing classes of cancer treatments. Basic, translational, and clinical research are now investigating therapeutic combinations of these agents. In comparison to external beam radiation therapy (EBRT), TRT has the unique advantage of treating all disease sites following intravenous injection and selective tumor uptake and retention-a particularly beneficial property in metastatic disease settings. The therapeutic value of combining radiation therapy with immune checkpoint blockade to treat metastases has been demonstrated in preclinical studies, whereas results of clinical studies have been mixed. Several clinical trials combining TRT and immune checkpoint blockade have been initiated based on preclinical studies combining these with EBRT and/or TRT. Despite the interest in translation of TRT and immunotherapy combinations, many questions remain surrounding the mechanisms of interaction and the optimal approach to clinical implementation of these combinations. This review highlights the mechanisms of interaction between anti-tumor immunity and radiation therapy and the status of basic and translational research and clinical trials investigating combinations of TRT and immunotherapies.
Collapse
Affiliation(s)
- Caroline P. Kerr
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joseph J. Grudzinski
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Thanh Phuong Nguyen
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Reinier Hernandez
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jamey P. Weichert
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary S. Morris
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
8
|
Chidlow G, Chan WO, Wood JPM, Casson RJ. Investigations into photoreceptor energy metabolism during experimental retinal detachment. Front Cell Neurosci 2022; 16:1036834. [PMID: 36467607 PMCID: PMC9716104 DOI: 10.3389/fncel.2022.1036834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/01/2022] [Indexed: 08/27/2023] Open
Abstract
Retinal detachment is a sight-threatening disorder, which occurs when the photoreceptors are separated from their vascular supply. The aim of the present study was to shed light on photoreceptor energy metabolism during experimental detachment in rats. Retinal detachment was induced in the eyes of rats via subretinal injection of sodium hyaluronate. Initially, we investigated whether detachment caused hypoxia within photoreceptors, as evaluated by the exogenous and endogenous biomarkers pimonidazole and HIF-1α, as well as by qPCR analysis of HIF target genes. The results showed no unequivocal staining for pimonidazole or HIF-1α within any detached retina, nor upregulation of HIF target genes, suggesting that any reduction in pO2 is of insufficient magnitude to produce hypoxia-induced covalent protein adducts or HIF-1α stabilisation. Subsequently, we analysed expression of cellular bioenergetic enzymes in photoreceptors during detachment. We documented loss of mitochondrial, and downregulation of glycolytic enzymes during detachment, indicating that photoreceptors have reduced energetic requirements and/or capacity. Given that detachment did not cause widespread hypoxia, but did result in downregulated expression of bioenergetic enzymes, we hypothesised that substrate insufficiency may be critical in terms of pathogenesis, and that boosting metabolic inputs may preserve photoreceptor bioenergetic production and, protect against their degeneration. Thus, we tested whether supplementation with the bioavailable energy substrate pyruvate mitigated rod and cone injury and degeneration. Despite protecting photoreceptors in culture from nutrient deprivation, pyruvate failed to protect against apoptotic death of rods, loss of cone opsins, and loss of inner segment mitochondria, in situ, when evaluated at 3 days after detachment. The regimen was also ineffective against cumulative photoreceptor deconstruction and degeneration when evaluated after 4 weeks. Retinal metabolism, particularly the bioenergetic profiles and pathological responses of the various cellular subtypes still presents a considerable knowledge gap that has important clinical consequences. While our data do not support the use of pyruvate supplementation as a means of protecting detached photoreceptors, they do provide a foundation and motivation for future research in this area.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia
| | | | | | | |
Collapse
|
9
|
Wu P, Han J, Gong Y, Liu C, Yu H, Xie N. Nanoparticle-Based Drug Delivery Systems Targeting Tumor Microenvironment for Cancer Immunotherapy Resistance: Current Advances and Applications. Pharmaceutics 2022; 14:pharmaceutics14101990. [PMID: 36297426 PMCID: PMC9612242 DOI: 10.3390/pharmaceutics14101990] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/06/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer immunotherapy has shown impressive anti-tumor activity in patients with advanced and early-stage malignant tumors, thus improving long-term survival. However, current cancer immunotherapy is limited by barriers such as low tumor specificity, poor response rate, and systemic toxicities, which result in the development of primary, adaptive, or acquired resistance. Immunotherapy resistance has complex mechanisms that depend on the interaction between tumor cells and the tumor microenvironment (TME). Therefore, targeting TME has recently received attention as a feasibility strategy for re-sensitizing resistant neoplastic niches to existing cancer immunotherapy. With the development of nanotechnology, nanoplatforms possess outstanding features, including high loading capacity, tunable porosity, and specific targeting to the desired locus. Therefore, nanoplatforms can significantly improve the effectiveness of immunotherapy while reducing its toxic and side effects on non-target cells that receive intense attention in cancer immunotherapy. This review explores the mechanisms of tumor microenvironment reprogramming in immunotherapy resistance, including TAMs, CAFs, vasculature, and hypoxia. We also examined whether the application of nano-drugs combined with current regimens is improving immunotherapy clinical outcomes in solid tumors.
Collapse
Affiliation(s)
- Peijie Wu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jun Han
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yanju Gong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Chao Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Han Yu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Correspondence: (H.Y.); (N.X.); Tel.:+86-158-8455-5293 (N.X.)
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
- Correspondence: (H.Y.); (N.X.); Tel.:+86-158-8455-5293 (N.X.)
| |
Collapse
|
10
|
Xu XX, Chen SY, Yi NB, Li X, Chen SL, Lei Z, Cheng DB, Sun T. Research progress on tumor hypoxia-associative nanomedicine. J Control Release 2022; 350:829-840. [PMID: 36100192 DOI: 10.1016/j.jconrel.2022.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 12/17/2022]
Abstract
Hypoxia at the solid tumor site is generally related to the unrestricted proliferation and metabolism of cancerous cells, which can cause tumor metastasis and aggravate tumor progression. Besides, hypoxia plays a substantial role in tumor treatment, and it is one of the main reasons that malignant tumors are difficult to cure and have a poor prognosis. On account of the tumor specific hypoxic environment, many hypoxia-associative nanomedicine have been proposed for tumor treatment. Considering the enhanced targeting effect, designing hypoxia-associative nanomedicine can not only minimize the adverse effects of drugs on normal tissues, but also achieve targeted therapy at the lesion site. Mostly, there can be three strategies for the treatment of hypoxic tumor, including improvement of hypoxic environment, hypoxia responsive drug release and hypoxia activated prodrug. The review describes the design principle and applications of tumor hypoxia-associative nanomedicine in recent years, and also explores its development trends in solid tumor treatment. Moreover, this review presents the current limitations of tumor hypoxia-associative nanomedicine in chemotherapy, radiotherapy, photodynamic therapy, sonodynamic therapy and immunotherapy, which may provide a reference for clinic translation of tumor hypoxia-associative nanomedicine.
Collapse
Affiliation(s)
- Xiao-Xue Xu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Yi Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Ning-Bo Yi
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Xin Li
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Lin Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| |
Collapse
|
11
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
12
|
Ruiter LN, van Dijk BAC, Bruggink AH, Doornaert PAH, Philippens MEP, de Bree R, van Gils CH, Willems SM. Association of histological features with laryngeal squamous cell carcinoma recurrences: a population-based study of 1502 patients in the Netherlands. BMC Cancer 2022; 22:444. [PMID: 35459142 PMCID: PMC9034596 DOI: 10.1186/s12885-022-09533-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/12/2022] [Indexed: 11/12/2022] Open
Abstract
Background Recurrences remain an important problem in laryngeal squamous cell carcinoma. Little has been described about histological characteristics of the primary laryngeal tumor that may be associated with recurrences. Identifying risk factors for recurrences might help in adapting treatment or follow-up. Using real-life population-based data, we aimed to identify histological features of the primary tumor associated with recurrences and overall survival. Material and methods Demographic, clinical and treatment information on all first primary invasive laryngeal tumors diagnosed in 2010–2014 (N = 3705) were extracted from the population-based nationwide Netherlands cancer registry (NCR) and linked to PALGA, the nationwide Dutch pathology registry, to obtain data on histological factors and recurrences. For a random 1502 patients histological information i.e., keratinization, perineural invasion (PNI+), vascular invasion (VI+), growth pattern, degree of differentiation, extracapsular spread (ECS+), cartilage- and bone invasion and extralaryngeal extension, was manually extracted from narrative pathology reports and analyzed for locoregional recurrence and overall survival using cox regression analysis. Results In total, 299 patients developed a locoregional recurrence and 555 patients died. Keratinization (HR = 0.96 (95%CI: 0.68–1.34) p = 0.79), two or three adverse characteristics (PNI+, VI+, non-cohesive growth) (HR = 1.38 (95% CI: 0.63–3.01) p = 0.42), and ECS+ (HR = 1.38 (95% CI: 0.48–4.02) p = 0.55) were not associated to recurrence. For death, also no significant association was found. Conclusion In this population-based real-life dataset on laryngeal carcinoma in the Netherlands, histological factors were not associated with locoregional recurrences or overall survival, but future studies should investigate the role of these features in treatment decisions. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09533-0.
Collapse
Affiliation(s)
- Lilian N Ruiter
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.
| | - Boukje A C van Dijk
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, the Netherlands.,Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Annette H Bruggink
- Nationwide Network and Registry of Histo- and Cytopathology in the Netherlands (PALGA Foundation), De Bouw 123, Houten, 3991 SZ, the Netherlands
| | - Patricia A H Doornaert
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands
| | - Marielle E P Philippens
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands
| | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands
| | - Carla H van Gils
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands
| | - Stefan M Willems
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, 3584 CX, the Netherlands.,Present address: Department of Pathology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9713 GZ, the Netherlands
| |
Collapse
|
13
|
Swartz JE, Smits HJG, Philippens MEP, de Bree R, H A M Kaanders J, Willems SM. Correlation and colocalization of HIF-1α and pimonidazole staining for hypoxia in laryngeal squamous cell carcinomas: A digital, single-cell-based analysis. Oral Oncol 2022; 128:105862. [PMID: 35447566 DOI: 10.1016/j.oraloncology.2022.105862] [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: 02/22/2022] [Revised: 03/31/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Tumor hypoxia results in worse local control and patient survival. We performed a digital, single-cell-based analysis to compare two biomarkers for hypoxia (hypoxia-inducible factor 1-alpha [HIF-1α] and pimonidazole [PIMO]) and their effect on outcome in laryngeal cancer patients treated with accelerated radiotherapy with or without carbogen breathing and nicotinamide (AR versus ARCON). MATERIALS AND METHODS Immunohistochemical staining was performed for HIF-1α and PIMO in consecutive sections of 44 laryngeal cancer patients randomized between AR and ARCON. HIF-1α expression and PIMO-binding were correlated using digital image analysis in QuPath. High-density areas for each biomarker were automatically annotated and staining overlap was analyzed. Kaplan-Meier survival analyses for local control, regional control and disease-free survival were performed to predict a response benefit of ARCON over AR alone for each biomarker. RESULTS 106 Tissue fragments of 44 patients were analyzed. A weak, significant positive correlation was observed between HIF-1α and PIMO positivity on fragment level, but not on patient level. A moderate strength correlation (r = 0.705, p < 0.001) was observed between the number of high-density staining areas for both biomarkers. Staining overlap was poor. HIF-1α expression, PIMO-binding or a combination could not predict a response benefit of ARCON over AR. CONCLUSION Digital image analysis to compare positive cell fractions and staining overlap between two hypoxia biomarkers using open-source software is feasible. Our results highlight that there are distinct differences between HIF-1α and PIMO as hypoxia biomarkers and therefore suggest co-existence of different forms of hypoxia within a single tumor.
Collapse
Affiliation(s)
- Justin E Swartz
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Hilde J G Smits
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johannes H A M Kaanders
- Department of Radiation Oncology, University Medical Center Nijmegen, Nijmegen, the Netherlands
| | - Stefan M Willems
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
14
|
van Lith SAM, Huizing FJ, Franssen GM, Hoeben BAW, Lok J, Doulkeridou S, Boerman OC, Gotthardt M, van Bergen En Henegouwen PMP, Bussink J, Heskamp S. Novel VHH-Based Tracers with Variable Plasma Half-Lives for Imaging of CAIX-Expressing Hypoxic Tumor Cells. Mol Pharm 2022; 19:3511-3520. [PMID: 35044182 PMCID: PMC9533306 DOI: 10.1021/acs.molpharmaceut.1c00841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
![]()
Hypoxic areas are
present in the majority of solid tumors, and
hypoxia is associated with resistance to therapies and poor outcomes.
A transmembrane protein that is upregulated by tumor cells that have
adapted to hypoxic conditions is carbonic anhydrase IX (CAIX). Therefore,
noninvasive imaging of CAIX could be of prognostic value, and it could
steer treatment strategies. The aim of this study was to compare variants
of CAIX-binding VHH B9, with and without a C-terminal albumin-binding
domain with varying affinity (ABDlow and ABDhigh), for SPECT imaging of CAIX expression. The binding affinity and
internalization of the various B9-variants were analyzed using SK-RC-52
cells. Biodistribution studies were performed in mice with subcutaneous
SCCNij153 human head and neck cancer xenografts. Tracer uptake was
determined by ex vivo radioactivity counting and
visualized by SPECT/CT imaging. Furthermore, autoradiography images
of tumor sections were spatially correlated with CAIX immunohistochemistry.
B9-variants demonstrated a similar moderate affinity for CAIX in vitro. Maximal tumor uptake and acceptable tumor-to-blood
ratios were found in the SCCNij153 model at 4 h post injection for
[111In]In-DTPA-B9 (0.51 ± 0.08%ID/g and 8.1 ±
0.85, respectively), 24 h post injection for [111In]In-DTPA-B9-ABDlow (2.39 ± 0.44%ID/g and 3.66 ± 0.81, respectively)
and at 72 h post injection for [111In]In-DTPA-B9-ABDhigh (8.7 ± 1.34%ID/g and 2.43 ± 0.15, respectively). An excess of unlabeled monoclonal anti-CAIX antibody efficiently
inhibited tumor uptake of [111In]In-DTPA-B9, while only
a partial reduction of [111In]In-DTPA-B9-ABDlow and [111In]In-DTPA-B9-ABDhigh uptake was found.
Immunohistochemistry and autoradiography images showed colocalization
of all B9-variants with CAIX expression; however, [111In]In-DTPA-B9-ABDlow and [111In]In-DTPA-B9-ABDhigh also
accumulated in non-CAIX expressing regions. Tumor uptake of [111In]In-DTPA-B9-ABDlow and [111In]In-DTPA-B9-ABDhigh, but not of [111In]In-DTPA-B9, could be visualized
with SPECT/CT imaging. In conclusion, [111In]In-DTPA-B9
has a high affinity to CAIX and shows specific targeting to CAIX in
head and neck cancer xenografts. The addition of ABD prolonged plasma
half-life, increased tumor uptake, and enabled SPECT/CT imaging. This
uptake was, however, partly CAIX- independent, precluding the ABD-tracers
for use in hypoxia quantification in this tumor type.
Collapse
Affiliation(s)
- Sanne A M van Lith
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Fokko J Huizing
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands.,Department of Radiation Oncology, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Gerben M Franssen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Bianca A W Hoeben
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands.,Department of Radiation Oncology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Jasper Lok
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Sofia Doulkeridou
- Department of Cell Biology, University of Utrecht, Utrecht, 3584 GE, The Netherlands
| | - Otto C Boerman
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | | | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| |
Collapse
|
15
|
Cortes AC, Nishiofuku H, Polak U, Minhaj AA, Lopez MS, Kichikawa K, Qayyum A, Whitley EM, Avritscher R. Effect of bead size and doxorubicin loading on tumor cellular injury after transarterial embolization and chemoembolization in a rat model of hepatocellular carcinoma. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 39:102465. [PMID: 34571240 PMCID: PMC9206412 DOI: 10.1016/j.nano.2021.102465] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/08/2021] [Accepted: 09/01/2021] [Indexed: 01/03/2023]
Abstract
Embolic agents used in transarterial embolization for intermediate stage hepatocellular carcinoma reduce blood flow into tumors and can deliver anticancer drugs. Tumor blood supply can be interrupted using doxorubicin-eluting beads (DEB-TACE) or non-loaded beads (TAE) of different calibers. In this preclinical study, we characterized the extent of remaining stressed tumor cells after treatment, hypoxia within the surviving tumor regions, and inflammatory immune cell infiltrates after embolization with 40-60 or 70-150 μm with non-loaded or doxorubicin-loaded beads at 3 and 7 days after treatment. TAE-treated tumors had more stressed and surviving tumor cells after 3 days, irrespective of bead size, compared with DEB-TACE-treated tumors. Hypoxic stress of residual cells increased after treatment with 70-150 μm beads without or with doxorubicin. Treatment with DEB-TACE of 70-150 μm resulted in increased inflammation and proliferation in the adjacent parenchyma. Inflammatory cell infiltrates were reduced at the periphery of tumors treated with 40-60 μm DEB-TACE.
Collapse
Affiliation(s)
- Andrea C Cortes
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hideyuki Nishiofuku
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX,Department of Radiology, IVR Center, Nara Medical University, Kashihara, Japan
| | - Urszula Polak
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Adeeb A Minhaj
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mirtha S Lopez
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kimihiko Kichikawa
- Department of Radiology, IVR Center, Nara Medical University, Kashihara, Japan
| | - Aliya Qayyum
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elizabeth M. Whitley
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rony Avritscher
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX,Corresponding author at: Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX. (R. Avritscher)
| |
Collapse
|
16
|
Schaner PE, Williams BB, Chen EY, Pettus JR, Schreiber WA, Kmiec MM, Jarvis LA, Pastel DA, Zuurbier RA, DiFlorio-Alexander RM, Paydarfar JA, Gosselin BJ, Barth RJ, Rosenkranz KM, Petryakov SV, Hou H, Tse D, Pletnev A, Flood AB, Wood VA, Hebert KA, Mosher RE, Demidenko E, Swartz HM, Kuppusamy P. First-In-Human Study in Cancer Patients Establishing the Feasibility of Oxygen Measurements in Tumors Using Electron Paramagnetic Resonance With the OxyChip. Front Oncol 2021; 11:743256. [PMID: 34660306 PMCID: PMC8517507 DOI: 10.3389/fonc.2021.743256] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/07/2021] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE The overall objective of this clinical study was to validate an implantable oxygen sensor, called the 'OxyChip', as a clinically feasible technology that would allow individualized tumor-oxygen assessments in cancer patients prior to and during hypoxia-modification interventions such as hyperoxygen breathing. METHODS Patients with any solid tumor at ≤3-cm depth from the skin-surface scheduled to undergo surgical resection (with or without neoadjuvant therapy) were considered eligible for the study. The OxyChip was implanted in the tumor and subsequently removed during standard-of-care surgery. Partial pressure of oxygen (pO2) at the implant location was assessed using electron paramagnetic resonance (EPR) oximetry. RESULTS Twenty-three cancer patients underwent OxyChip implantation in their tumors. Six patients received neoadjuvant therapy while the OxyChip was implanted. Median implant duration was 30 days (range 4-128 days). Forty-five successful oxygen measurements were made in 15 patients. Baseline pO2 values were variable with overall median 15.7 mmHg (range 0.6-73.1 mmHg); 33% of the values were below 10 mmHg. After hyperoxygenation, the overall median pO2 was 31.8 mmHg (range 1.5-144.6 mmHg). In 83% of the measurements, there was a statistically significant (p ≤ 0.05) response to hyperoxygenation. CONCLUSIONS Measurement of baseline pO2 and response to hyperoxygenation using EPR oximetry with the OxyChip is clinically feasible in a variety of tumor types. Tumor oxygen at baseline differed significantly among patients. Although most tumors responded to a hyperoxygenation intervention, some were non-responders. These data demonstrated the need for individualized assessment of tumor oxygenation in the context of planned hyperoxygenation interventions to optimize clinical outcomes.
Collapse
Affiliation(s)
- Philip E. Schaner
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Benjamin B. Williams
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Eunice Y. Chen
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Jason R. Pettus
- Department of Pathology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Wilson A. Schreiber
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Maciej M. Kmiec
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Lesley A. Jarvis
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - David A. Pastel
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Rebecca A. Zuurbier
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Roberta M. DiFlorio-Alexander
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Joseph A. Paydarfar
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Benoit J. Gosselin
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Richard J. Barth
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Kari M. Rosenkranz
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Sergey V. Petryakov
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Huagang Hou
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Dan Tse
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Alexandre Pletnev
- Department of Chemistry, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Ann Barry Flood
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Victoria A. Wood
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Kendra A. Hebert
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Robyn E. Mosher
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Eugene Demidenko
- Department of Biomedical Data Science, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Harold M. Swartz
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Periannan Kuppusamy
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
- Department of Chemistry, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| |
Collapse
|
17
|
Telarovic I, Wenger RH, Pruschy M. Interfering with Tumor Hypoxia for Radiotherapy Optimization. J Exp Clin Cancer Res 2021; 40:197. [PMID: 34154610 PMCID: PMC8215813 DOI: 10.1186/s13046-021-02000-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/30/2021] [Indexed: 12/11/2022] Open
Abstract
Hypoxia in solid tumors is an important predictor of treatment resistance and poor clinical outcome. The significance of hypoxia in the development of resistance to radiotherapy has been recognized for decades and the search for hypoxia-targeting, radiosensitizing agents continues. This review summarizes the main hypoxia-related processes relevant for radiotherapy on the subcellular, cellular and tissue level and discusses the significance of hypoxia in radiation oncology, especially with regard to the current shift towards hypofractionated treatment regimens. Furthermore, we discuss the strategies to interfere with hypoxia for radiotherapy optimization, and we highlight novel insights into the molecular pathways involved in hypoxia that might be utilized to increase the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Irma Telarovic
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
| |
Collapse
|
18
|
Eskiizmir G, Çalıbaşı Koçal G, Uysal T, Ellidokuz H, Başpınar Y. Serum hypoxia-inducible factor-2: A candidate prognostic biomarker for laryngeal cancer. Clin Otolaryngol 2021; 46:1172-1183. [PMID: 33908192 DOI: 10.1111/coa.13789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/16/2021] [Accepted: 04/03/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To determine the serum hypoxia-inducible factor-1, -2 and -3 (HIF-1, -2 and -3) levels in patients with laryngeal neoplasm, and to investigate their role in differential diagnosis, prediction of tumour characteristic and extension, and prognosis and survival. STUDY DESIGN Prospective, cohort study at a tertiary referral centre. SETTINGS The study was conducted in a tertiary medical centre. PARTICIPANTS Patients with benign, premalignant and malignant laryngeal neoplasms were included. Sixty-four patients with a laryngeal neoplasm were enrolled. MAIN OUTCOME MEASURES Serum HIF-1, -2 and -3 levels were measured from blood samples that were drawn before treatment, using ELISA. RESULTS A statistically significant difference between benign (HIF-1, -2, -3:4046,1 pg/mL; 2581,5 pg/mL; 1321,0 pg/mL), premalignant (HIF-1, -2, -3:3630,3 pg/mL; 3229,7 pg/mL; 2549,8 pg/mL) and malignant (HIF-1, -2, -3:3576,7 pg/mL; 2595,8 pg/mL; 1106,3 pg/mL) laryngeal neoplasms was not detected when serum HIF-1, -2 and -3 levels were compared. However, high serum HIF-2 level adversely affected survival and locoregional control and had more than 7-fold increase in hazard ratio. Moreover, serum HIF-2 was an independent prognostic factor for 2-year overall, disease-free, distant metastasis-free survival and locoregional control. CONCLUSION This is the first clinical study in which the diagnostic, predictive and prognostic roles of hypoxia-related biomolecules were examined in laryngeal neoplasms. Hypoxia-inducible factor-2 is a prognostic factor in larynx cancer irrespective of treatment modality.
Collapse
Affiliation(s)
- Görkem Eskiizmir
- Department of Otolaryngology-Head Neck Surgery, Manisa Celal Bayar University, Manisa, Turkey
| | | | - Tuğba Uysal
- Institute of Oncology, Dokuz Eylül University, Izmir, Turkey
| | - Hülya Ellidokuz
- Institute of Oncology, Dokuz Eylül University, Izmir, Turkey
| | | |
Collapse
|
19
|
Wang L, Wang H, Shen K, Park H, Zhang T, Wu X, Hu M, Yuan H, Chen Y, Wu Z, Wang Q, Li Z. Development of Novel 18F-PET Agents for Tumor Hypoxia Imaging. J Med Chem 2021; 64:5593-5602. [PMID: 33901402 DOI: 10.1021/acs.jmedchem.0c01962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tumor hypoxia is a major factor responsible for tumor progression, metastasis, invasion, and treatment resistance, leading to low local tumor control and recurrence after radiotherapy in cancers. Here,18F-positron emission tomography (PET) probes are developed for visualizing viable hypoxic cells in biopsies. Pimonidazole derivatives and nitroimidazole-based agents bearing sulfonyl linkers were evaluated. A small-animal PET study showed that the tumor uptake of [18F]-23 [poly(ethylene glycols) (PEG)-sulfonyl linker] of 3.36 ± 0.29%ID/g was significantly higher (P < 0.01) than that of [18F]-20 (piperazine-linker tracer, 2.55 ± 0.49%ID/g) at 2 h postinjection in UPPL tumors. The tumor-to-muscle uptake ratio of [18F]-23 (2.46 ± 0.48 at 2 h pi) was well improved compared with that of [18F]-FMISO (1.25 ± 0.14 at 2 h pi). A comparable distribution pattern was observed between ex vivo autoradiography of [18F]-23 and pimonidazole staining of the neighboring slice, indicating that [18F]-23 is a promising PET agent for hypoxia imaging.
Collapse
Affiliation(s)
- Li Wang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States.,Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, China
| | - Hui Wang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Kun Shen
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Hyejin Park
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Tao Zhang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Xuedan Wu
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Mei Hu
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, China
| | - Hong Yuan
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Yue Chen
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, China
| | - Zhanhong Wu
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Qiu Wang
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Zibo Li
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27514, United States
| |
Collapse
|
20
|
D'Alonzo RA, Gill S, Rowshanfarzad P, Keam S, MacKinnon KM, Cook AM, Ebert MA. In vivo noninvasive preclinical tumor hypoxia imaging methods: a review. Int J Radiat Biol 2021; 97:593-631. [PMID: 33703994 DOI: 10.1080/09553002.2021.1900943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/28/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022]
Abstract
Tumors exhibit areas of decreased oxygenation due to malformed blood vessels. This low oxygen concentration decreases the effectiveness of radiation therapy, and the resulting poor perfusion can prevent drugs from reaching areas of the tumor. Tumor hypoxia is associated with poorer prognosis and disease progression, and is therefore of interest to preclinical researchers. Although there are multiple different ways to measure tumor hypoxia and related factors, there is no standard for quantifying spatial and temporal tumor hypoxia distributions in preclinical research or in the clinic. This review compares imaging methods utilized for the purpose of assessing spatio-temporal patterns of hypoxia in the preclinical setting. Imaging methods provide varying levels of spatial and temporal resolution regarding different aspects of hypoxia, and with varying advantages and disadvantages. The choice of modality requires consideration of the specific experimental model, the nature of the required characterization and the availability of complementary modalities as well as immunohistochemistry.
Collapse
Affiliation(s)
- Rebecca A D'Alonzo
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, Australia
| | - Suki Gill
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, Australia
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Pejman Rowshanfarzad
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, Australia
| | - Synat Keam
- School of Medicine, The University of Western Australia, Crawley, Australia
| | - Kelly M MacKinnon
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, Australia
| | - Alistair M Cook
- School of Medicine, The University of Western Australia, Crawley, Australia
| | - Martin A Ebert
- School of Physics, Mathematics and Computing, The University of Western Australia, Crawley, Australia
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Nedlands, Australia
- 5D Clinics, Claremont, Australia
| |
Collapse
|
21
|
Wu P, Gao W, Su M, Nice EC, Zhang W, Lin J, Xie N. Adaptive Mechanisms of Tumor Therapy Resistance Driven by Tumor Microenvironment. Front Cell Dev Biol 2021; 9:641469. [PMID: 33732706 PMCID: PMC7957022 DOI: 10.3389/fcell.2021.641469] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/05/2021] [Indexed: 02/05/2023] Open
Abstract
Cancer is a disease which frequently has a poor prognosis. Although multiple therapeutic strategies have been developed for various cancers, including chemotherapy, radiotherapy, and immunotherapy, resistance to these treatments frequently impedes the clinical outcomes. Besides the active resistance driven by genetic and epigenetic alterations in tumor cells, the tumor microenvironment (TME) has also been reported to be a crucial regulator in tumorigenesis, progression, and resistance. Here, we propose that the adaptive mechanisms of tumor resistance are closely connected with the TME rather than depending on non-cell-autonomous changes in response to clinical treatment. Although the comprehensive understanding of adaptive mechanisms driven by the TME need further investigation to fully elucidate the mechanisms of tumor therapeutic resistance, many clinical treatments targeting the TME have been successful. In this review, we report on recent advances concerning the molecular events and important factors involved in the TME, particularly focusing on the contributions of the TME to adaptive resistance, and provide insights into potential therapeutic methods or translational medicine targeting the TME to overcome resistance to therapy in clinical treatment.
Collapse
Affiliation(s)
- Peijie Wu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wei Gao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Miao Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Wenhui Zhang
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jie Lin
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
22
|
Abstract
Radiopharmaceutical therapy or targeted radionuclide therapy (TRT) is a well-established class of cancer therapeutics that includes a growing number of FDA-approved drugs and a promising pipeline of experimental therapeutics. Radiobiology is fundamental to a mechanistic understanding of the therapeutic capacity of these agents and their potential toxicities. However, the field of radiobiology has historically focused on external beam radiation. Critical differences exist between TRT and external beam radiotherapy with respect to dosimetry, dose rate, linear energy transfer, duration of treatment delivery, fractionation, range, and target volume. These distinctions simultaneously make it difficult to extrapolate from the radiobiology of external beam radiation to that of TRT and pose considerable challenges for preclinical and clinical studies investigating TRT. Here, we discuss these challenges and explore the current understanding of the radiobiology of radiopharmaceuticals.
Collapse
Affiliation(s)
- Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Andrew Z Wang
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University, Palo Alto, CA.
| |
Collapse
|
23
|
Imaging Hypoxia. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00074-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
24
|
Wang S, Fu Z, Wang Y, Sun Y, Cui L, Wang C, Liu Q, Shao D, Wang Y, Wen N. Correlation of carbonic anhydrase 9 (CA9) with pathological T-stage and prognosis in patients with oral tongue squamous cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1521. [PMID: 33313266 PMCID: PMC7729320 DOI: 10.21037/atm-20-7144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background We explored the mechanisms underlying tumorigenesis in oral tongue squamous cell carcinoma (OTSCC) with the goal of uncovering prognostic molecular biomarkers. Methods An mRNA sequencing dataset was obtained from The Cancer Genome Atlas (TCGA) database, and differentially expressed genes (DEGs) were selected using R language software packages. Functional enrichment analysis was conducted with DAVID software and protein-protein interaction (PPI) networks were constructed using the STRING database. The relationship between hub genes and overall survival (OS) was evaluated by Kaplan-Meier analysis and Cox proportional hazard regression models. Expression of the candidate gene, carbonic anhydrase 9 (CA9), was verified by real-time RT-PCR, western blotting, and immunohistochemistry. Results DEGs (n=581) were obtained from 11 OTSCC samples and corresponding adjacent non-tumor tissues. Gene ontology (GO) analysis revealed that most DEGs were implicated in anterior/posterior pattern specification, embryonic skeletal system morphogenesis, and multicellular organism development, and pathway analysis suggested that DEGs were associated with neuroactive ligand-receptor interaction, calcium signaling pathway and transcriptional misregulation in the cancer. A PPI network consisting of 301 nodes and 2011 edges was constructed and 71 hub genes, with high degrees of connectivity in the network, were identified. Kaplan-Meier analysis of the hub genes indicated that high expression of CA9, LHX1, and KISS1R and low expression of CCKAR were associated with poor OS in OTSCC; however, only CA9 was a significant prognostic factor influencing survival in OTSCC on multivariate analysis. High expression of CA9 was associated with poor pathological T-stage. CA9 tumor specificity was confirmed using the Gene Expression Omnibus (GEO) database and further molecular tests. Conclusions We identified key DEGs that may assist in the molecular understanding of OTSCC. CA9 warrants further exploration as potential prognostic biomarker and therapeutic target in OTSCC.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China.,Department of Stomatology, Huangdao District Central Hospital, Qingdao, China
| | - Zhiguang Fu
- Department of Tumor Radiotherapy, Air Force Medical Center, PLA, Beijing, China
| | - Yizhu Wang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yaping Sun
- Department of Stomatology, Huangdao District Central Hospital, Qingdao, China
| | - Lei Cui
- Department of Stomatology, Huangdao District Central Hospital, Qingdao, China
| | - Chunfang Wang
- Department of Stomatology, Huangdao District Central Hospital, Qingdao, China
| | - Qiaoling Liu
- Department of Oncology, Huangdao District Central Hospital, Qingdao, China
| | - Dan Shao
- Department of Stomatology, Huangdao District Central Hospital, Qingdao, China
| | - Yu Wang
- Department of Oncology, Air Force Medical Center, PLA, Beijing, China
| | - Ning Wen
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Fang Y, Ouyang L, Zhang T, Wang C, Lu B, Sun W. Optimizing Bifurcated Channels within an Anisotropic Scaffold for Engineering Vascularized Oriented Tissues. Adv Healthc Mater 2020; 9:e2000782. [PMID: 32790048 DOI: 10.1002/adhm.202000782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/20/2020] [Indexed: 11/07/2022]
Abstract
Despite progress in engineering both vascularized tissues and oriented tissues, the fabrication of 3D vascularized oriented tissues remains a challenge due to an inability to successfully integrate vascular and anisotropic structures that can support mass transfer and guide cell alignment, respectively. More importantly, there is a lack of an effective approach to guiding the scaffold design bearing both structural features. Here, an approach is presented to optimize the bifurcated channels within an anisotropic scaffold based on oxygen transport simulation and biological experiments. The oxygen transport simulation is performed using the experimentally measured effective oxygen diffusion coefficient and hydraulic permeability of the anisotropic scaffolds, which are also seeded with muscle precursor cells and cultured in a custom-made perfusion bioreactor. Symmetric bifurcation model is used as fractal unit to design the channel network based on biomimetic principles. The bifurcation level of channel network is further optimized based on the oxygen transport simulation, which is then validated by DNA quantification assay and pimonidazole immunostaining. This study provides a practical guide to optimizing bifurcated channels in anisotropic scaffolds for oriented tissue engineering.
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, P. R. China
- "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, P. R. China
| | - Liliang Ouyang
- Department of Materials, Imperial College London, London, SW7 2AZ, UK
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, P. R. China
- "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, P. R. China
| | - Chengjin Wang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, P. R. China
- "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, P. R. China
| | - Bingchuan Lu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, P. R. China
- "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, P. R. China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, P. R. China
- "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, P. R. China
- Department of Mechanical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
26
|
Okuyan D, Aydogan Turkoglu S, Kockar F. Carbonic anhydrase III is a new target of HIF1α in prostate cancer model. Gene 2020; 762:145034. [PMID: 32777521 DOI: 10.1016/j.gene.2020.145034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/09/2020] [Accepted: 08/04/2020] [Indexed: 12/30/2022]
Abstract
Carbonic Anhydrase III (CAIII) belongs to a member of the alpha Carbonic Anhydrase (CA) family. Although some CA members are strongly up-regulated by HIF1-α, it is not known about the transcriptional regulation of CAIII in prostate cancer cells, PCa. Therefore, we aimed to identify regulatory regions important for the regulation of CAIII gene under hypoxic conditions in human prostate cancer cells (PC3). The present study, for the first time, demonstrated that the chemically mimicked hypoxic condition led to the induced CAIII mRNA and protein expression in prostate cancer cells. Transcriptional regulation of CAIII was investigated by transient transfection assay that indicates that the most active promoter activity was in the region of P2 -699/+86. Hypoxic condition also upregulates the basal activity of for P1;-941/+86 and P2;-699/+86 constructs containing putative Hypoxia Response Element (HRE) region located in -268/-252. EMSA analysis of HRE located in -268/-252 bases, showed one DNA-protein binding complexes. Competition assays indicated this complex is resulted from HIF1α interactions. In addition, site-directed mutagenesis of potential HIF1α binding sites diminished a DNA-protein complex. These findings suggest that CAIII is a hypoxia-regulated gene and valuable for targeting of prostate cancer tumors in hypoxic condition.
Collapse
Affiliation(s)
- Derya Okuyan
- Balikesir University, Faculty of Science and Literature, Department of Molecular Biology and Genetics, Balıkesir, Turkey
| | - Sumeyye Aydogan Turkoglu
- Balikesir University, Faculty of Science and Literature, Department of Molecular Biology and Genetics, Balıkesir, Turkey
| | - Feray Kockar
- Balikesir University, Faculty of Science and Literature, Department of Molecular Biology and Genetics, Balıkesir, Turkey.
| |
Collapse
|
27
|
Qian J, Yu X, Li B, Fei Z, Huang X, Luo P, Zhang L, Zhang Z, Lou J, Wang H. In vivo Monitoring of Oxygen Levels in Human Brain Tumor Between Fractionated Radiotherapy Using Oxygen-enhanced MR Imaging. Curr Med Imaging 2020; 16:427-432. [PMID: 32410542 DOI: 10.2174/1573405614666180925144814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/19/2018] [Accepted: 09/11/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND It was known that the response of tumor cells to radiation is closely related to tissue oxygen level and fractionated radiotherapy allows reoxygenation of hypoxic tumor cells. Non-invasive mapping of tissue oxygen level may hold great importance in clinic. OBJECTIVE The aim of this study is to evaluate the role of oxygen-enhanced MR imaging in the detection of tissue oxygen levels between fractionated radiotherapy. METHODS A cohort of 10 patients with brain metastasis was recruited. Quantitative oxygen enhanced MR imaging was performed prior to, 30 minutes and 22 hours after first fractionated radiotherapy. RESULTS The ΔR1 (the difference of longitudinal relaxivity between 100% oxygen breathing and air breathing) increased in the ipsilateral tumor site and normal tissue by 242% and 152%, respectively, 30 minutes after first fractionated radiation compared to pre-radiation levels. Significant recovery of ΔR1 in the contralateral normal tissue (p < 0.05) was observed 22 hours compared to 30 minutes after radiation levels. CONCLUSION R1-based oxygen-enhanced MR imaging may provide a sensitive endogenous marker for oxygen changes in the brain tissue between fractionated radiotherapy.
Collapse
Affiliation(s)
- Junchao Qian
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China.,Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xiang Yu
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Bingbing Li
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Zhenle Fei
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Xiang Huang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Peng Luo
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Liwei Zhang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Zhiming Zhang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Jianjun Lou
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China
| | - Hongzhi Wang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei Cancer Hospital, Hefei 230031, China.,Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
28
|
Huizing FJ, Garousi J, Lok J, Franssen G, Hoeben BAW, Frejd FY, Boerman OC, Bussink J, Tolmachev V, Heskamp S. CAIX-targeting radiotracers for hypoxia imaging in head and neck cancer models. Sci Rep 2019; 9:18898. [PMID: 31827111 PMCID: PMC6906415 DOI: 10.1038/s41598-019-54824-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-induced carbonic anhydrase IX (CAIX) expression is a prognostic marker in solid tumors. In recent years many radiotracers have been developed, but a fair comparison of these compounds is not possible because of the diversity in tumor models and other experimental parameters. In this study we performed a direct in vivo comparison of three promising CAIX targeting radiotracers in xenografted head and neck cancer models. The biodistribution of [111In]In-DOTA-ZCAIX:2 was directly compared with [111In]In-DTPA-G250-F(ab′)2 and [111In] In-DTPA-G250 in female BALB/C nu/nu mice bearing two HNSCC xenografts with different levels of CAIX expression. In vivo biodistribution was quantified by means of microSPECT/CT scans and ex vivo biodistribution was determined with the use of a γ-counter. Tumors were snap frozen and sections were stained for CAIX expression, vessels, hypoxia (pimonidazole) and tumor blood perfusion. Tracer uptake was significantly higher in SSCNij153 tumors compared to SCCNij185 tumors for [111In]In-DOTA-HE3-ZCAIX:2: 0.32 ± 0.03 versus 0.18 ± 0.01%ID/g,(p = 0.003) 4 h p.i., for [111In]In-DTPA-girentuximab-F(ab′)2: 3.0 ± 0.5%ID/g and 1.2 ± 0.1%ID/g (p = 0.03), 24 h p.i. and for [111In]In-DTPA-girentuximab: 30 ± 2.1%ID/g and 7.0 ± 1.0%ID/g (p = 0.0002) 72 h p.i. SPECT imaging with both [111In]In-DTPA-girentuximab-F(ab′)2 and [111In]In-DTPA-girentuximab showed a clear difference in tracer distribution between the two tumor models. The whole IgG, i.e. [111In]In-DTPA-girentuximab, showed the highest tumor-to-muscle ratio. We showed that different CAIX-targeting radiotracers can discriminate a low CAIX-expressing tumor from a high CAIX-expressing head and neck cancer xenografts model. In these hypoxic head and neck xenograft models [111In]In-DTPA-girentuximab showed the most promising results.
Collapse
Affiliation(s)
- Fokko J Huizing
- Departments of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jasper Lok
- Departments of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben Franssen
- Departments of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bianca A W Hoeben
- Departments of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Affibody AB, Solna, Sweden
| | - Otto C Boerman
- Departments of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan Bussink
- Departments of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sandra Heskamp
- Departments of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
29
|
Olcina MM, Balanis NG, Kim RK, Aksoy BA, Kodysh J, Thompson MJ, Hammerbacher J, Graeber TG, Giaccia AJ. Mutations in an Innate Immunity Pathway Are Associated with Poor Overall Survival Outcomes and Hypoxic Signaling in Cancer. Cell Rep 2019; 25:3721-3732.e6. [PMID: 30590044 PMCID: PMC6405289 DOI: 10.1016/j.celrep.2018.11.093] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/01/2018] [Accepted: 11/27/2018] [Indexed: 12/18/2022] Open
Abstract
Complement-mediated cytotoxicity may act as a selective pressure for tumor overexpression of complement regulators. We hypothesize that the same selective pressure could lead to complement alterations at the genetic level. We find that, when analyzed as a pathway, mutations in complement genes occur at a relatively high frequency and are associated with changes in overall survival across a number of cancer types. Analysis of pathways expressed in patients with complement mutations that are associated with poor overall survival reveals crosstalk between complement and hypoxia in colorectal cancer. The importance of this crosstalk is highlighted by two key findings: hypoxic signaling is increased in tumors harboring complement mutations, and hypoxic tumor cells are resistant to complement-mediated cytotoxicity due, in part, to hypoxia-induced expression of complement regulator CD55. The range of strategies employed by tumors to dysregulate the complement system testifies to the importance of this pathway in tumor progression.
Collapse
Affiliation(s)
- Monica M Olcina
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA.
| | - Nikolas G Balanis
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ryan K Kim
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - B Arman Aksoy
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Julia Kodysh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael J Thompson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jeff Hammerbacher
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Poor outcome in hypoxic endometrial carcinoma is related to vascular density. Br J Cancer 2019; 120:1037-1044. [PMID: 31011231 PMCID: PMC6738053 DOI: 10.1038/s41416-019-0461-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/01/2019] [Accepted: 04/04/2019] [Indexed: 01/05/2023] Open
Abstract
Background Identification of endometrial carcinoma (EC) patients at high risk of recurrence is lacking. In this study, the prognostic role of hypoxia and angiogenesis was investigated in EC patients. Methods Tumour slides from EC patients were stained by immunofluorescence for carbonic anhydrase IX (CAIX) as hypoxic marker and CD34 for assessment of microvessel density (MVD). CAIX expression was determined in epithelial tumour cells, with a cut-off of 1%. MVD was assessed according to the Weidner method. Correlations with disease-specific survival (DSS), disease-free survival (DFS) and distant disease-free survival (DDFS) were calculated using Kaplan–Meier curves and Cox regression analysis. Results Sixty-three (16.4%) of 385 ECs showed positive CAIX expression with high vascular density. These ECs had a reduced DSS compared to tumours with either hypoxia or high vascular density (log-rank p = 0.002). Multivariable analysis showed that hypoxic tumours with high vascular density had a reduced DSS (hazard ratio [HR] 3.71, p = 0.002), DDFS (HR 2.68, p = 0.009) and a trend for reduced DFS (HR 1.87, p = 0.054). Conclusions This study has shown that adverse outcome in hypoxic ECs is seen in the presence of high vascular density, suggesting an important role of angiogenesis in the metastatic process of hypoxic EC. Differential adjuvant treatment might be indicated for these patients.
Collapse
|
31
|
Chen Y, Hou Y, Yang Y, Pan M, Wang J, Wang W, Zuo Y, Cong J, Wang X, Mu N, Zhang C, Gong B, Hou J, Wang S, Xu L. Gene expression changes in cervical squamous cancers following neoadjuvant interventional chemoembolization. Clin Chim Acta 2019; 493:79-86. [PMID: 30772336 DOI: 10.1016/j.cca.2019.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/21/2019] [Accepted: 02/13/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND The efficacy of therapy for cervical cancer is related to the alteration of multiple molecular events and signaling networks during treatment. The aim of this study was to evaluate gene expression alterations in advanced cervical cancers before- and after-trans-uterine arterial chemoembolization- (TUACE). METHODS Gene expression patterns in three squamous cell cervical cancers before- and after-TUACE were determined using microarray technique. Changes in AKAP12 and CA9 genes following TUACE were validated by quantitative real-time PCR. RESULTS Unsupervised cluster analysis revealed that the after-TUACE samples clustered together, which were separated from the before-TUACE samples. Using a 2-fold threshold, we identified 1131 differentially expressed genes that clearly discriminate after-TUACE tumors from before-TUACE tumors, including 209 up-regulated genes and 922 down-regulated genes. Pathway analysis suggests these genes represent diverse functional categories. Results from real-time PCR confirmed the expression changes detected by microarray. CONCLUSIONS Gene expression signature significantly changes during TUACE therapy of cervical cancer. Theses alterations provide useful information for the development of novel treatment strategies for cervical cancers on the molecular level.
Collapse
Affiliation(s)
- Yonghua Chen
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Yuanyuan Hou
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Ying Yang
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Meixia Pan
- Yantai Yuhuangding Hospital LaiShan Division of Medical College, Qingdao University, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Wenshuang Wang
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Ying Zuo
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Jianglin Cong
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Xiaojie Wang
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Nan Mu
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Chenglin Zhang
- Central Laboratory, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Benjiao Gong
- Central Laboratory, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China
| | - Jianqing Hou
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China.
| | - Shaoguang Wang
- Department of Obstetrics and Gynecology, the Affiliated Yantai Yuhuangding Hospital of Medical College, Qingdao University, Yantai 264000, Shandong, China.
| | - Liping Xu
- Medical College, Qingdao University, Qingdao 266021, Shandong, China
| |
Collapse
|
32
|
Huizing FJ, Hoeben BAW, Franssen GM, Boerman OC, Heskamp S, Bussink J. Quantitative Imaging of the Hypoxia-Related Marker CAIX in Head and Neck Squamous Cell Carcinoma Xenograft Models. Mol Pharm 2018; 16:701-708. [PMID: 30550290 PMCID: PMC6364270 DOI: 10.1021/acs.molpharmaceut.8b00950] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Tumor hypoxia plays a major role
in radio- and chemotherapy resistance
in solid tumors. Carbonic Anhydrase IX (CAIX) is an endogenous hypoxia-related
protein, which is associated with poor patient outcome. The quantitative
assessment of CAIX expression of tumors may steer cancer treatment
by predicting therapy response or patient selection for antihypoxia
or CAIX-targeted treatment. Recently, the single-photon emission computerized
tomography (SPECT) tracer [111In]In-DTPA-girentuximab-F(ab′)2 was developed and validated for targeting CAIX. The aim of
this study was to optimize quantitative microSPECT/CT of CAIX expression in vivo in head and neck tumor models. Athymic mice with
subcutaneous SCCNij153 and SCCNij202 head and neck squamous cell carcinoma
xenografts were injected with [111In]In-DTPA-girentuximab-F(ab′)2. First, the protein dose, timing, and image acquisition settings
were optimized. Tracer uptake was determined by quantitative SPECT, ex vivo radioactivity counting, and by autoradiography of
tumor sections. The same tumor sections were immunohistochemically
stained for CAIX expression and hypoxia. Highest tumor-normal-tissue
contrast was obtained at 24 h after injection of the tracer. A protein
dose of 10 μg resulted in the highest tumor-to-muscle ratio
at 24 h p.i. Ex vivo biodistribution studies showed
a tumor uptake of 3.0 ± 0.6%ID/g and a tumor-to-muscle ratio
of 8.7 ± 1.4 (SCCNij153). Quantitative analysis of the SPECT
images enabled us to distinguish CAIX antigen blocked from nonblocked
tumors, fractions positive for CAIX expression: 0.22 ± 0.02 versus
0.08 ± 0.01 (p < 0.01). Immunohistochemical,
autoradiographic, and microSPECT/CT analyses showed a distinct intratumoral
spatial correlation between localization of the radiotracer and CAIX
expression. Here, we demonstrate that [111In]In-DTPA-girentuximab-F(ab′)2 specifically targets CAIX-expressing cells in head and neck
cancer xenografts. SPECT imaging with indium-labeled girentuximab-F(ab′)2 allows quantitative assessment of the fraction of CAIX positive
tissue in head and neck cancer xenografts. These results indicate
that [111In]In-DTPA-girentuximab-F(ab′)2 is a promising tracer to image hypoxia-related CAIX expression.
Collapse
|
33
|
Desmet CM, Tran LBA, Danhier P, Gallez B. Characterization of a clinically used charcoal suspension for in vivo EPR oximetry. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2018; 32:205-212. [DOI: 10.1007/s10334-018-0704-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/21/2018] [Accepted: 08/31/2018] [Indexed: 12/18/2022]
|
34
|
Olcina MM, Kim RK, Melemenidis S, Graves EE, Giaccia AJ. The tumour microenvironment links complement system dysregulation and hypoxic signalling. Br J Radiol 2018; 92:20180069. [PMID: 29544344 PMCID: PMC6435069 DOI: 10.1259/bjr.20180069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The complement system is an innate immune pathway typically thought of as part of the first line of defence against “non-self” species. In the context of cancer, complement has been described to have an active role in facilitating cancer-associated processes such as increased proliferation, angiogenesis and migration. Several cellular members of the tumour microenvironment express and/or produce complement proteins locally, including tumour cells. Dysregulation of the complement system has been reported in numerous tumours and increased expression of complement activation fragments in cancer patient specimens correlates with poor patient prognosis. Importantly, genetic or pharmacological targeting of complement has been shown to reduce tumour growth in several cancer preclinical models, suggesting that complement could be an attractive therapeutic target. Hypoxia (low oxygen) is frequently found in solid tumours and has a profound biological impact on cellular and non-cellular components of the tumour microenvironment. In this review, we focus on hypoxia since this is a prevailing feature of the tumour microenvironment that, like increased complement, is typically associated with poor prognosis. Furthermore, interesting links between hypoxia and complement have been recently proposed but never collectively reviewed. Here, we explore how hypoxia alters regulation of complement proteins in different cellular components of the tumour microenvironment, as well as the downstream biological consequences of this regulation.
Collapse
Affiliation(s)
- Monica M Olcina
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Ryan K Kim
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Edward E Graves
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
35
|
Featherstone AK, O'Connor JP, Little RA, Watson Y, Cheung S, Babur M, Williams KJ, Matthews JC, Parker GJ. Data-driven mapping of hypoxia-related tumor heterogeneity using DCE-MRI and OE-MRI. Magn Reson Med 2018; 79:2236-2245. [PMID: 28856728 PMCID: PMC5836865 DOI: 10.1002/mrm.26860] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 07/13/2017] [Accepted: 07/13/2017] [Indexed: 01/06/2023]
Abstract
PURPOSE Previous work has shown that combining dynamic contrast-enhanced (DCE)-MRI and oxygen-enhanced (OE)-MRI binary enhancement maps can identify tumor hypoxia. The current work proposes a novel, data-driven method for mapping tissue oxygenation and perfusion heterogeneity, based on clustering DCE/OE-MRI data. METHODS DCE-MRI and OE-MRI were performed on nine U87 (glioblastoma) and seven Calu6 (non-small cell lung cancer) murine xenograft tumors. Area under the curve and principal component analysis features were calculated and clustered separately using Gaussian mixture modelling. Evaluation metrics were calculated to determine the optimum feature set and cluster number. Outputs were quantitatively compared with a previous non data-driven approach. RESULTS The optimum method located six robustly identifiable clusters in the data, yielding tumor region maps with spatially contiguous regions in a rim-core structure, suggesting a biological basis. Mean within-cluster enhancement curves showed physiologically distinct, intuitive kinetics of enhancement. Regions of DCE/OE-MRI enhancement mismatch were located, and voxel categorization agreed well with the previous non data-driven approach (Cohen's kappa = 0.61, proportional agreement = 0.75). CONCLUSION The proposed method locates similar regions to the previous published method of binarization of DCE/OE-MRI enhancement, but renders a finer segmentation of intra-tumoral oxygenation and perfusion. This could aid in understanding the tumor microenvironment and its heterogeneity. Magn Reson Med 79:2236-2245, 2018. © 2017 The Authors Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
Collapse
Affiliation(s)
- Adam K. Featherstone
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
| | - James P.B. O'Connor
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
- Division of Cancer StudiesThe University of ManchesterManchesterUK
- Department of RadiologyChristie NHS Foundation TrustManchesterUK
| | - Ross A. Little
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
| | - Yvonne Watson
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
| | - Sue Cheung
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
| | - Muhammad Babur
- Division of Pharmacy & OptometryThe University of ManchesterManchesterUK
| | - Kaye J. Williams
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
- Division of Pharmacy & OptometryThe University of ManchesterManchesterUK
| | - Julian C. Matthews
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
| | - Geoff J.M. Parker
- Division of Informatics, Imaging & Data SciencesThe University of ManchesterManchesterUK
- CRUK & EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and ManchesterUK
- Bioxydyn LtdManchesterUK
| |
Collapse
|
36
|
Marie-Egyptienne DT, Chaudary N, Kalliomäki T, Hedley DW, Hill RP. Cancer initiating-cells are enriched in the CA9 positive fraction of primary cervix cancer xenografts. Oncotarget 2018; 8:1392-1404. [PMID: 27901496 PMCID: PMC5352063 DOI: 10.18632/oncotarget.13625] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 11/07/2016] [Indexed: 12/27/2022] Open
Abstract
Numerous studies have suggested that Cancer Initiating Cells (CIC) can be identified/enriched in cell populations obtained from solid tumors based on the expression of cell surface marker proteins. We used early passage primary cervix cancer xenografts to sort cells based on the expression of the intrinsic hypoxia marker Carbonic Anhydrase 9 (CA9) and tested their cancer initiation potential by limiting dilution assay. We demonstrated that CICs are significantly enriched in the CA9+ fraction in 5/6 models studied. Analyses of the expression of the stem cell markers Oct4, Notch1, Sca-1 & Bmi1 showed a trend toward an increase in the CA9+ populations, albeit not significant. We present evidence that enhanced autophagy does not play a role in the enhanced growth of the CA9+ cells. Our study suggests a direct in vivo functional link between hypoxic cells and CICs in primary cervix cancer xenografts.
Collapse
Affiliation(s)
- Delphine Tamara Marie-Egyptienne
- Ontario Cancer Institute/Princess Margaret Cancer Centre, University Health Network and Campbell Family Institute for Cancer Research, Toronto, Ontario, M5G2M9, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Naz Chaudary
- Ontario Cancer Institute/Princess Margaret Cancer Centre, University Health Network and Campbell Family Institute for Cancer Research, Toronto, Ontario, M5G2M9, Canada
| | - Tuula Kalliomäki
- Ontario Cancer Institute/Princess Margaret Cancer Centre, University Health Network and Campbell Family Institute for Cancer Research, Toronto, Ontario, M5G2M9, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - David William Hedley
- Ontario Cancer Institute/Princess Margaret Cancer Centre, University Health Network and Campbell Family Institute for Cancer Research, Toronto, Ontario, M5G2M9, Canada.,Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, M5G2M9, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario, Canada
| | - Richard Peter Hill
- Ontario Cancer Institute/Princess Margaret Cancer Centre, University Health Network and Campbell Family Institute for Cancer Research, Toronto, Ontario, M5G2M9, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Klein C, Dokic I, Mairani A, Mein S, Brons S, Häring P, Haberer T, Jäkel O, Zimmermann A, Zenke F, Blaukat A, Debus J, Abdollahi A. Overcoming hypoxia-induced tumor radioresistance in non-small cell lung cancer by targeting DNA-dependent protein kinase in combination with carbon ion irradiation. Radiat Oncol 2017; 12:208. [PMID: 29287602 PMCID: PMC5747947 DOI: 10.1186/s13014-017-0939-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 12/06/2017] [Indexed: 01/19/2023] Open
Abstract
Background Hypoxia-induced radioresistance constitutes a major obstacle for a curative treatment of cancer. The aim of this study was to investigate effects of photon and carbon ion irradiation in combination with inhibitors of DNA-Damage Response (DDR) on tumor cell radiosensitivity under hypoxic conditions. Methods Human non-small cell lung cancer (NSCLC) models, A549 and H1437, were irradiated with dose series of photon and carbon ions under hypoxia (1% O2) vs. normoxic conditions (21% O2). Clonogenic survival was studied after dual combinations of radiotherapy with inhibitors of DNA-dependent Protein Kinase (DNAPKi, M3814) and ATM serine/threonine kinase (ATMi). Results The OER at 30% survival for photon irradiation of A549 cells was 1.4. The maximal oxygen effect measured as survival ratio was 2.34 at 8 Gy photon irradiation of A549 cells. In contrast, no significant oxygen effect was found after carbon ion irradiation. Accordingly, the relative effect of 6 Gy carbon ions was determined as 3.8 under normoxia and. 4.11 under hypoxia. ATM and DNA-PK inhibitors dose dependently sensitized tumor cells for both radiation qualities. For 100 nM DNAPKi the survival ratio at 4 Gy more than doubled from 1.59 under normoxia to 3.3 under hypoxia revealing a strong radiosensitizing effect under hypoxic conditions. In contrast, this ratio only moderately increased after photon irradiation and ATMi under hypoxia. The most effective treatment was combined carbon ion irradiation and DNA damage repair inhibition. Conclusions Carbon ions efficiently eradicate hypoxic tumor cells. Both, ATMi and DNAPKi elicit radiosensitizing effects. DNAPKi preferentially sensitizes hypoxic cells to radiotherapy. Electronic supplementary material The online version of this article (10.1186/s13014-017-0939-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carmen Klein
- Division of Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Ivana Dokic
- Division of Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Andrea Mairani
- Division of Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany.,National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Stewart Mein
- Division of Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Stephan Brons
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Peter Häring
- Heidelberg Institute of Radiation Oncology (HIRO), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Physics in Radiation Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Haberer
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Oliver Jäkel
- Heidelberg Institute of Radiation Oncology (HIRO), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | - Jürgen Debus
- Division of Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Amir Abdollahi
- Division of Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Germany. .,Heidelberg Institute of Radiation Oncology (HIRO), German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany. .,Division of Molecular and Translational Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Im Neuenheimer Feld 450, 69120, Heidelberg, Germany.
| |
Collapse
|
38
|
Impact of Blood Vessel Quantity and Vascular Expression of CD133 and ICAM-1 on Survival of Glioblastoma Patients. NEUROSCIENCE JOURNAL 2017; 2017:5629563. [PMID: 29250531 PMCID: PMC5698821 DOI: 10.1155/2017/5629563] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 08/07/2017] [Accepted: 10/04/2017] [Indexed: 11/18/2022]
Abstract
Glioblastoma (GB) is the most angiogenic tumor. Nevertheless, antiangiogenic therapy has not shown significant clinical efficacy. The aim of this study was to assess blood vessel characteristics on survival of GB patients. Surgically excised GB tissues were histologically examined for overall proportion of glomeruloid microvascular proliferation (MP) and the total number of blood vessels. Also, immunohistochemical vascular staining intensities of CD133 and ICAM-1 were determined. Vessel parameters were correlated with patients' overall survival. The survival time depended on the number of blood vessels (p = 0.03) but not on the proportion of MP. Median survival times for patients with low (<median) and high (≥median) number of blood vessels were 9.0 months (95% CI: 7.5–10.5) and 12.0 months (95% CI: 9.3–14.7). Also, median survival times for patients with low (<median) and high (≥median) vascular expression level of CD133 were 9.0 months (95% CI: 8.0–10.1) and 12.0 months (95% CI: 10.3–13.7). In contrast, the staining intensity of vascular ICAM-1 did not affect survival. In multivariate analysis, the number of blood vessels emerged as an independent predictor for longer overall survival (HR: 2.4, 95% CI: 1.2–5.0, p = 0.02). For success in antiangiogenic therapy, better understanding about tumor vasculature biology is needed.
Collapse
|
39
|
|
40
|
He L, Zhao C, Li Y, Du G, Liu K, Cui D, Tang L, Wu X, Wen S, Chen H. Antiangiogenic effects of recombinant human endostatin in lung cancers. Mol Med Rep 2017; 17:79-86. [PMID: 29115591 PMCID: PMC5780156 DOI: 10.3892/mmr.2017.7859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/22/2017] [Indexed: 12/31/2022] Open
Abstract
Antiangiogenic therapy, as a new anticancer method, can improve the anticancer effect of traditional therapies. Different antiangiogenic drugs may have different vascular normalization time windows. Whether the antiangiogenic treatment is within the vascular normalization time window is very important in the treatment of cancers. Previous studies have indicated that recombinant human endostatin (rh-ES) can transiently normalize tumor microvessels. Yet the molecular mechanism and the time window of rh-ES remains unclear. The aim of the present study was to explore the optimal time window and molecular mechanism of rh-ES in inhibiting Lewis lung cancer (LLC). By comparatively accessing the changes in microvascular and hypoxic conditions of tumors in host mice treated with rh-ES or saline for different days, the authors aimed to investigate the best administration time of rh-ES treatment on human lung cancers and obtain a better understanding concerning the involved molecular mechanism. A total of 40 C57/BL6 mice with LLC xenografts were randomly divided into normal saline (NS) and rh-ES groups (20 mice/group). 0.2 ml NS or 5 mg/kg rh-ES were administrated via intraperitoneal injection (i.p.) into each mouse each day during the 9-day experiment. A total of 5 mice from each group were sacrificed at day 2, 4, 6 or 9. CA9 and RGS5 expression levels of both groups were compared using immunohistochemistry, reverse transcription-quantitative polymerase chain reaction and ELISA. Rh-ES caused vascular normalization and improved hypoxia at days 4 and 6. Compared with the control (NS) group, both CA9 and RGS5 expression in rh-ES group were significantly decreased at days 4 and 6 (P<0.05), while no significant change between two groups was observed at days 2 and 9. Rh-ES can induce transient tumor vascular normalization and improves tissue hypoxia in LLC tumors. The vascular normalization window is accompanied by the reduction in RGS5 and CA expression.
Collapse
Affiliation(s)
- Lang He
- Department of Oncology, The Fifth People's Hospital of Chengdu, North Sichuan Medical College, Chengdu, Sichuan 611130, P.R. China
| | - Chaofen Zhao
- Department of Medical Oncology, Affiliated Hospital of Guizhou Medical University, Guizhou Cancer Hospital, Guiyang, Guizhou 550004, P.R. China
| | - Yunxiang Li
- Department of Urology, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guocheng Du
- Department of Galactophore, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Dandan Cui
- Department of Oncology, The Fifth People's Hospital of Chengdu, North Sichuan Medical College, Chengdu, Sichuan 611130, P.R. China
| | - Lina Tang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xun Wu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shimin Wen
- Cancer Center, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Hong Chen
- Tumor Department of TCM, Sichuan Cancer Hospital and Institute, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
41
|
Gallez B, Neveu MA, Danhier P, Jordan BF. Manipulation of tumor oxygenation and radiosensitivity through modification of cell respiration. A critical review of approaches and imaging biomarkers for therapeutic guidance. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:700-711. [DOI: 10.1016/j.bbabio.2017.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/05/2017] [Accepted: 01/06/2017] [Indexed: 11/17/2022]
|
42
|
Goda JS, Pachpor T, Basu T, Chopra S, Gota V. Targeting the AKT pathway: Repositioning HIV protease inhibitors as radiosensitizers. Indian J Med Res 2017; 143:145-59. [PMID: 27121513 PMCID: PMC4859124 DOI: 10.4103/0971-5916.180201] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cellular resistance in tumour cells to different therapeutic approaches has been a limiting factor in the curative treatment of cancer. Resistance to therapeutic radiation is a common phenomenon which significantly reduces treatment options and impacts survival. One of the mechanisms of acquiring resistance to ionizing radiation is the overexpression or activation of various oncogenes like the EGFR (epidermal growth factor receptor), RAS (rat sarcoma) oncogene or loss of PTEN (phosphatase and tensin homologue) which in turn activates the phosphatidyl inositol 3-kinase/protein kinase B (PI3-K)/AKT pathway responsible for radiation resistance in various tumours. Blocking the pathway enhances the radiation response both in vitro and in vivo. Due to the differential activation of this pathway (constitutively activated in tumour cells and not in the normal host cells), it is an excellent candidate target for molecular targeted therapy to enhance radiation sensitivity. In this regard, HIV protease inhibitors (HPIs) known to interfere with PI3-K/AKT signaling in tumour cells, have been shown to sensitize various tumour cells to radiation both in vitro and in vivo. As a result, HPIs are now being investigated as possible radiosensitizers along with various chemotherapeutic drugs. This review describes the mechanisms by which PI3-K/AKT pathway causes radioresistance and the role of HIV protease inhibitors especially nelfinavir as a potential candidate drug to target the AKT pathway for overcoming radioresistance and its use in various clinical trials for different malignancies.
Collapse
Affiliation(s)
- Jayant S Goda
- Department of Radiation Oncology; Clinical Biology Laboratory, Department of Radiation Oncology, Advance Centre for Treatment Research & Education in Cancer, Tata Memorial Center, Navi Mumbai, India
| | | | | | | | | |
Collapse
|
43
|
Imai T, Muz B, Yeh CH, Yao J, Zhang R, Azab AK, Wang L. Direct measurement of hypoxia in a xenograft multiple myeloma model by optical-resolution photoacoustic microscopy. Cancer Biol Ther 2017; 18:101-105. [PMID: 28045569 DOI: 10.1080/15384047.2016.1276137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Using photoacoustic microscopy (PAM), we evaluated non-invasively oxygenation and vascularization in vivo due to multiple myeloma (MM) progression. Mice injected with MM.1S-GFP were monitored with a fluorescence microscope for tumor progression. In vivo PAM of the cerebral bone marrow quantified the total oxygen saturation (sO2). At 28 days after the MM cell injection, the total sO2 had decreased by half in the developing tumor regions, while in the non-tumor regions it had decreased by 20% compared with the value at one day post MM injection. The blood vessel density was reduced by 35% in the developing tumor regions, while in the non-tumor regions it was reduced by 8% compared with the value at one day post MM injection. Hence, PAM corroborated the development of hypoxia due to MM progression and demonstrated decreased vascularization surrounding the tumor areas.
Collapse
Affiliation(s)
- Toru Imai
- a Department of Biomedical Engineering , Washington University in St. Louis , St. Louis , MO , USA
| | - Barbara Muz
- b Department of Radiation Oncology , Cancer Biology Division, Washington University in St. Louis School of Medicine , St. Louis , MO , USA
| | - Cheng-Hung Yeh
- a Department of Biomedical Engineering , Washington University in St. Louis , St. Louis , MO , USA
| | - Junjie Yao
- a Department of Biomedical Engineering , Washington University in St. Louis , St. Louis , MO , USA
| | - Ruiying Zhang
- a Department of Biomedical Engineering , Washington University in St. Louis , St. Louis , MO , USA
| | - Abdel Kareem Azab
- b Department of Radiation Oncology , Cancer Biology Division, Washington University in St. Louis School of Medicine , St. Louis , MO , USA
| | - Lihong Wang
- a Department of Biomedical Engineering , Washington University in St. Louis , St. Louis , MO , USA
| |
Collapse
|
44
|
Evaluation of CAIX and CAXII Expression in Breast Cancer at Varied O2 Levels: CAIX is the Superior Surrogate Imaging Biomarker of Tumor Hypoxia. Mol Imaging Biol 2016; 18:219-31. [PMID: 26276155 DOI: 10.1007/s11307-015-0885-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Hypoxia is commonly observed in regions of primary tumors and metastases, and is associated with resistance to treatment, more aggressive tumor phenotypes and poor prognosis. Reliable and validated imaging biomarkers of hypoxia are needed for pre-clinical studies and clinical use. Expression of cell-surface carbonic anhydrases IX and XII (CAIX and CAXII) in tumor cells has been associated with tumor hypoxia. CAIX and CAXII specific antibodies conjugated to fluorescent dye were evaluated for the non-invasive detection of hypoxia in vivo. PROCEDURES Human breast cancer cell lines (MCF10A, DCIS, MCF7, ZR-75.1 and MDA-mb231) were characterized for CAIX and CAXII expression by real-time RT-PCR and immunocytochemistry (ICC) under normoxic and hypoxic conditions. Immunohistochemical (IHC) staining of CAIX, CAXII and the commercially available exogenous hypoxia marker, pimonidazole, was performed using sections of ZR-75.1 and MDA-mb-231 orthotopic breast cancer xenograft tumors from nude mice. In vivo fluorescence imaging of ZR-75.1 tumors in animals housed at varied levels of oxygen was used to quantify the relative uptake of the CAIX and CAXII agents and a commercially available sulfonamide-based agent. Corresponding tumor sections were IHC stained for CAIX, CAXII and pimonidazole. RESULTS CAIX mRNA expression was significantly higher (p < 0.05) in hypoxia for all cell lines, which was in agreement with protein expression by ICC. CAXII expression was mixed, with a modest hypoxia-related increase in two cell lines (p < 0.05) and no change in others. Quantified IHC staining of ZR-75.1 and MDA-mb-231 tumor sections showed that CAIX and CAXII expression was elevated in regions with pimonidazole staining, but CAXII levels were lower than CAIX. Tumor uptake of the CAIX targeted agent, and IHC staining of CAIX and pimonidazole in corresponding tumor sections were correlated, and co-registered, and shown to be significantly elevated by level of oxygenation (p < 0.001): hypoxia > normoxia > hyperoxia. However, the CAXII and sulfonamide agents were not significantly correlated with hypoxia. CONCLUSION These studies suggest that the fluorescently labeled CAIX-specific agent is a more robust indicator of hypoxia in vivo compared to the CAXII-specific agent or the agent specific to the CA active site.
Collapse
|
45
|
Swartz HM. Using Stable Free Radicals to Obtain Unique and Clinically Useful Data In Vivo in Human Subjects. RADIATION PROTECTION DOSIMETRY 2016; 172:3-15. [PMID: 27886997 PMCID: PMC6061194 DOI: 10.1093/rpd/ncw323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/30/2016] [Indexed: 06/06/2023]
Abstract
This paper attempts to: (1) provide a critical overview of the challenges and opportunities to extend electron paramagnetic resonance (EPR) into practical applications in human subjects, based on EPR measurements made in vivo; (2) summarize the clinical applications of EPR for improving treatments in cancer, wound healing and diabetic care, emphasizing EPR's unique capability to measure tissue oxygen repeatedly and with particular sensitivity to hypoxia and (3) summarize the capabilities of in vivo EPR to measure radiation dose for triage and medical guidance after a large-scale radiation exposure. The conclusion is that while still at a relatively early stage of its development and availability, clinical applications of EPR already have demonstrated significant value and the field is likely to grow in both the extent of its applications and its impact on significant problems.
Collapse
Affiliation(s)
- Harold M Swartz
- EPR Center for the Study of Viable Systems at Dartmouth, Department of Radiology, Geisel School of Medicine at Dartmouth, HB 7785 One Medical Center Drive, Lebanon, NH 03756, USA
- Division of Radiation Oncology, Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
46
|
Olcina MM, Giaccia AJ. Reducing radiation-induced gastrointestinal toxicity - the role of the PHD/HIF axis. J Clin Invest 2016; 126:3708-3715. [PMID: 27548524 DOI: 10.1172/jci84432] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an effective treatment strategy for cancer, but a significant proportion of patients experience radiation-induced toxicity due to damage to normal tissue in the irradiation field. The use of chemical or biological approaches aimed at reducing or preventing normal tissue toxicity induced by radiotherapy is a long-held goal. Hypoxia-inducible factors (HIFs) regulate the production of factors that may protect several cellular compartments affected by radiation-induced toxicity. Pharmacological inhibitors of prolyl hydroxylase domain-containing enzymes (PHDs), which result in stabilization of HIFs, have recently been proposed as a new class of radioprotectors. In this review, radiation-induced toxicity in the gastrointestinal (GI) tract and the main cellular compartments studied in this context will be discussed. The effects of PHD inhibition on GI radioprotection will be described in detail.
Collapse
|
47
|
Fortuna L, Relf J, Chang YM, Hibbert A, Martineau H, Garden O. Prevalence of FoxP3 + Cells in Canine Tumours and Lymph Nodes Correlates Positively with Glucose Transporter 1 Expression. J Comp Pathol 2016; 155:171-180. [DOI: 10.1016/j.jcpa.2016.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/19/2016] [Accepted: 06/04/2016] [Indexed: 12/19/2022]
|
48
|
Laurens E, Yeoh SD, Rigopoulos A, O'Keefe GJ, Tochon-Danguy HJ, Chong LW, White JM, Scott AM, Ackermann U. Fluorine-18 radiolabeling of a nitrophenyl sulfoxide and its evaluation in an SK-RC-52 model of tumor hypoxia. J Labelled Comp Radiopharm 2016; 59:416-23. [PMID: 27435268 DOI: 10.1002/jlcr.3426] [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: 04/06/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 11/10/2022]
Abstract
The significance of imaging hypoxia with the positron emission tomography ligand [(18) F]FMISO has been demonstrated in a variety of cancers. However, the slow kinetics of [(18) F]FMISO require a 2-h delay between tracer administration and patient scanning. Labeled chloroethyl sulfoxides have shown faster kinetics and higher contrast than [(18) F]FMISO in a rat model of ischemic stroke. However, these nitrogen mustard analogues are unsuitable for routine production and use in humans. Here, we report on the synthesis and in vitro and in vivo evaluation of a novel sulfoxide, which contains an ester moiety for hydrolysis and subsequent trapping in hypoxic cells. Non-decay corrected yields of radioactivity were 1.18 ± 0.24% (n = 27, 2.5 ± 0.5% decay corrected radiochemical yield) based on K[(18) F]F. The radiotracer did not show any defluorination and did not undergo metabolism in an in vitro assay using S9 liver fractions. Imaging studies using an SK-RC-52 tumor model in BALB/c nude mice have revealed that [(18) F]1 is retained in hypoxic tumors and has similar hypoxia selectivity to [(18) F]FMISO. Because of a three times faster clearance rate than [(18) F]FMISO from normoxic tissue, [(18) F]1 has emerged as a promising new radiotracer for hypoxia imaging.
Collapse
Affiliation(s)
- Evelyn Laurens
- School of Chemistry, The University of Melbourne, Melbourne, Australia.,Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Shinn Dee Yeoh
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | | | - Graeme J O'Keefe
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
| | - Henri J Tochon-Danguy
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia.,Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Lee Wenn Chong
- School of Chemistry, The University of Melbourne, Melbourne, Australia.,Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Jonathan M White
- School of Chemistry, The University of Melbourne, Melbourne, Australia.,Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Andrew M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia.,Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Uwe Ackermann
- Bio21 Institute, The University of Melbourne, Melbourne, Australia.,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia.,Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
49
|
Prognostic Significance of Transverse Relaxation Rate (R2*) in Blood Oxygenation Level-Dependent Magnetic Resonance Imaging in Patients with Invasive Breast Cancer. PLoS One 2016; 11:e0158500. [PMID: 27384310 PMCID: PMC4934782 DOI: 10.1371/journal.pone.0158500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/15/2016] [Indexed: 11/19/2022] Open
Abstract
Objective To examine the relationship between magnetic resonance transverse relaxation rate (R2*) and prognostic factors. Materials and Methods A total of 159 women with invasive ductal carcinomas (IDCs) underwent breast magnetic resonance imaging (MRI) including blood oxygenation level-dependent (BOLD) sequence at 3 T. The distribution of the measured R2* values were analyzed, and the correlation between R2* and various prognostic factors (age, tumor size, histologic grade, lymphovascular invasion, and axillary lymph node status, as well as expression of estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2, p53, and Ki-67) were retrospectively assessed using patient medical records. Results The baseline R2* values of the IDCs were very heterogeneous with wide range among the patients. The mean R2* value was (32.8 ± 14.0) Hz with a median of 29.3 Hz (range 13.5–109.4 Hz). In multivariate analysis, older age was associated with decreased R2* value (P = 0.011) and IDCs with p53-overexpression showed higher R2* values than those without p53-overexpression group (P = 0.031). Other prognostic factors were not significantly correlated with R2* value. Conclusion In this study, R2* values were significantly correlated with age and expression of p53. Further studies are necessary to determine the prognostic value of BOLD-MRI.
Collapse
|
50
|
Horsman MR, Vaupel P. Pathophysiological Basis for the Formation of the Tumor Microenvironment. Front Oncol 2016; 6:66. [PMID: 27148472 PMCID: PMC4828447 DOI: 10.3389/fonc.2016.00066] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/07/2016] [Indexed: 12/27/2022] Open
Abstract
Poor microenvironmental conditions are a characteristic feature of solid tumors. Such conditions occur because the tumor vascular supply, which develops from the normal host vasculature by the process of angiogenesis, is generally inadequate in meeting the oxygen and nutrient demands of the growing tumor mass. Regions of low oxygenation (hypoxia) is believed to be the most critical deficiency, since it has been well documented to play a significant role in influencing the response to conventional radiation and chemotherapy treatments, as well as influencing malignant progression in terms of aggressive growth and recurrence of the primary tumor and its metastatic spread. As a result, significant emphasis has been placed on finding clinically applicable approaches to identify those tumors that contain hypoxia and realistic methods to target this hypoxia. However, most studies consider hypoxia as a single entity, yet we now know that it is multifactorial. Furthermore, hypoxia is often associated with other microenvironmental parameters, such as elevated interstitial fluid pressure, glycolysis, low pH, and reduced bioenergetic status, and these can also influence the effects of hypoxia. Here, we review the various aspects of hypoxia, but also discuss the role of the other microenvironmental parameters associated with hypoxia.
Collapse
Affiliation(s)
- Michael R Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital , Aarhus , Denmark
| | - Peter Vaupel
- Department of Radiooncology and Radiotherapy, Klinikum rechts der Isar, Technische Universität München (TUM) , Munich , Germany
| |
Collapse
|