1
|
Lim AHW, Zobel J, Bills M, Hsieh W, Crouch B, Joshi R, Thomson JE, Neo E, Kuan LL, Safaeian R, Tse E, Rayner CK, Ruszkiewicz A, Singhal N, Bartholomeusz D, Nguyen NQ. The Impact of Combined Chemotherapy and Intra-Tumoural Injection of Phosphorus-32 Microparticles on Vascularity in Locally Advanced Pancreatic Carcinoma. Cancers (Basel) 2024; 16:3412. [PMID: 39410031 PMCID: PMC11475738 DOI: 10.3390/cancers16193412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Poor intra-tumoural vascularity contributes to a lack of response to chemotherapy in pancreatic cancers. Preliminary data suggest that the addition of endoscopic ultrasound (EUS)-guided intra-tumoural injection of phosphorus-32 (32P) microparticles to standard chemotherapy is potentially beneficial in locally advanced pancreatic cancer (LAPC). We aimed to assess changes in pancreatic tumour vascularity following 32P implantation, using contrast-enhanced EUS (CE-EUS). METHODS This was a prospective single-centre trial from January 2022 to 2024 of patients with unresectable, non-metastatic LAPC undergoing standard FOLFIRINOX chemotherapy and 32P implantation. We performed CE-EUS pre-implantation after two chemotherapy cycles and 4 and 12 weeks after implantation. Time-intensity curves were analysed for 90 s after IV contrast bolus to ascertain peak intensity and intensity gain. RESULTS A total of 20 patients underwent 32P implantation, with 15 completing 12-week follow-up. The technical success of 32P implantation was 100%. The median primary tumour size reduced from 32 mm (IQR 27.5-38.75) pre-implantation to 24 mm (IQR 16-26) 12 weeks post-implantation (p < 0.001). Five patients (25%) had tumour downstaging, and four underwent resections. The baseline (pre-implantation, post-chemotherapy) median intensity gain of contrast enhancement within the tumour was 32.15 (IQR 18.08-54.35). This increased to 46.85 (IQR 35.05-76.6; p = 0.007) and 66.3 (IQR 54.7-76.3; p = 0.001) at 4 weeks and 12 weeks post-implantation, respectively. Over a median follow-up of 11.2 months (IQR 7.8-12.8), 15/20 (75%) of patients remained alive, with 3/20 (15%) demonstrating local disease progression. Overall survival was not significantly different between patients with or without an increased intensity of 10 a.u. or more at 12 weeks post-implantation. CONCLUSION This is the first clinical study to demonstrate treatment-induced increased vascularity within pancreatic primary tumours, which followed 32P implantation and FOLFIRINOX chemotherapy. Larger comparative trials are warranted.
Collapse
Affiliation(s)
- Amanda Huoy Wen Lim
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
| | - Joshua Zobel
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
| | - Madison Bills
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.B.); (W.H.); (B.C.)
| | - William Hsieh
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.B.); (W.H.); (B.C.)
| | - Benjamin Crouch
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.B.); (W.H.); (B.C.)
| | - Rohit Joshi
- Medical Oncology, Lyell McEwin Hospital, Adelaide, SA 5112, Australia;
| | - John-Edwin Thomson
- Department of Hepatobiliary Surgery, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (J.-E.T.); (E.N.)
| | - EuLing Neo
- Department of Hepatobiliary Surgery, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (J.-E.T.); (E.N.)
| | - Li Lian Kuan
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital, Adelaide, SA 5011, Australia;
| | - Romina Safaeian
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
| | - Christopher K. Rayner
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
| | - Andrew Ruszkiewicz
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
- Surgical Pathology, SA Pathology, Adelaide, SA 5000, Australia
- Centre of Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia
| | - Nimit Singhal
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
- Oncology, Royal Adelaide Hospital, Adelaide, SA 5081, Australia
| | - Dylan Bartholomeusz
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.B.); (W.H.); (B.C.)
| | - Nam Quoc Nguyen
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Road, Adelaide, SA 5000, Australia; (A.H.W.L.); (J.Z.); (R.S.); (E.T.); (C.K.R.); (D.B.)
- School Medicine, The University of Adelaide, Adelaide, SA 5005, Australia; (A.R.); (N.S.)
| |
Collapse
|
2
|
Rickard AG, Mowery YM, Bassil A, Rouse DC, Williams NT, Charity T, Belloni R, Crouch B, Ramanujam N, Stevenson D, Castillo R, Blocker S, Epel B, Kotecha M, Palmer GM. Evaluating Tumor Hypoxia Radiosensitization Via Electron Paramagnetic Resonance Oxygen Imaging (EPROI). Mol Imaging Biol 2024; 26:435-447. [PMID: 37721686 DOI: 10.1007/s11307-023-01855-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/30/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE Tumor hypoxia contributes to aggressive phenotypes and diminished therapeutic responses to radiation therapy (RT) with hypoxic tissue being 3-fold less radiosensitive than normoxic tissue. A major challenge in implementing hypoxic radiosensitizers is the lack of a high-resolution imaging modality that directly quantifies tissue-oxygen. The electron paramagnetic resonance oxygen-imager (EPROI) was used to quantify tumor oxygenation in two murine tumor models: E0771 syngeneic transplant breast cancers and primary p53/MCA soft tissue sarcomas, with the latter autochthonous model better recapitulating the tumor microenvironment in human malignancies. We hypothesized that tumor hypoxia differs between these models. We also aimed to quantify the absolute change in tumor hypoxia induced by the mitochondrial inhibitor papaverine (PPV) and its effect on RT response. PROCEDURES Tumor oxygenation was characterized in E0771 and primary p53/MCA sarcomas via EPROI, with the former model also being quantified indirectly via diffuse reflectance spectroscopy (DRS). After confirming PPV's effect on hypoxic fraction (via EPROI), we compared the effect of 0 versus 2 mg/kg PPV prior to 20 Gy on tumor growth delay and survival. RESULTS Hypoxic sarcomas were more radioresistant than normoxic sarcomas (p=0.0057, 2-way ANOVA), and high baseline hypoxic fraction was a significant (p=0.0063, Cox Regression Model) hazard in survivability regardless of treatment. Pre-treatment with PPV before RT did not radiosensitize tumors in the sarcoma or E0771 model. In the sarcoma model, EPROI successfully identified baseline hypoxic tumors. DRS quantification of total hemoglobin, saturated hemoglobin, changes in mitochondrial potential and glucose uptake showed no significant difference in E0771 tumors pre- and post-PPV. CONCLUSION EPROI provides 3D high-resolution pO2 quantification; EPR is better suited than DRS to characterize tumor hypoxia. PPV did not radiosensitize E0771 tumors nor p53/MCA sarcomas, which may be related to the complex pattern of vasculature in each tumor. Additionally, understanding model-dependent tumor hypoxia will provide a much-needed foundation for future therapeutic studies with hypoxic radiosensitizers.
Collapse
Affiliation(s)
- Ashlyn G Rickard
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA.
- Department of Head and Neck Surgery & Communication Sciences, Duke University Medical Center, Durham, NC, USA.
| | - Alex Bassil
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Douglas C Rouse
- Division of Laboratory Animal Resources, Duke University School of Medicine, Durham, NC, USA
| | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Theresa Charity
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Rafaela Belloni
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Brian Crouch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nimmi Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Rico Castillo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Stephanie Blocker
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Boris Epel
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- O2M Technologies LLC, Chicago, IL, USA
| | | | - Gregory M Palmer
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
3
|
Bodgi L, Bou-Gharios J, Azzi J, Challita R, Feghaly C, Baalbaki K, Kharroubi H, Chhade F, Geara F, Abou-Kheir W, Ayoub Z. Effect of bisphosphonates and statins on the in vitro radiosensitivity of breast cancer cell lines. Pharmacol Rep 2024; 76:171-184. [PMID: 38151641 DOI: 10.1007/s43440-023-00560-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Early-stage breast cancer is usually treated with breast-conserving surgery followed by adjuvant radiation therapy. Acute skin toxicity is a common radiation-induced side effect experienced by many patients. Recently, a combination of bisphosphonates (zoledronic acid) and statins (pravastatin), or ZOPRA, was shown to radio-protect normal tissues by enhancing DNA double-strand breaks (DSB) repair mechanism. However, there are no studies assessing the effect of ZOPRA on cancerous cells. The purpose of this study is to characterize the in vitro effect of the zoledronic acid (ZO), pravastatin (PRA), and ZOPRA treatment on the molecular and cellular radiosensitivity of breast cancer cell lines. MATERIALS Two breast cancer cell lines, MDA MB 231 and MCF-7, were tested. Cells were treated with different concentrations of pravastatin (PRA), zoledronate (ZO), as well as their ZOPRA combination, before irradiation. Anti-γH2AX and anti-pATM immunofluorescence were performed to study DNA DSB repair kinetics. MTT assay was performed to assess cell proliferation and viability, and flow cytometry was performed to analyze the effect of the drugs on the cell cycle distribution. The clonogenic assay was used to assess cell survival. RESULTS ZO, PRA, and ZOPRA treatments were shown to increase the residual number of γH2AX foci for both cell lines. ZOPRA treatment was also shown to reduce the activity of the ATM kinase in MCF-7. ZOPRA induced a significant decrease in cell survival for both cell lines. CONCLUSIONS Our findings show that pretreatment with ZOPRA can decrease the radioresistance of breast cancer cells at the molecular and cellular levels. The fact that ZOPRA was previously shown to radioprotect normal tissues, makes it a good candidate to become a therapeutic window-widening drug.
Collapse
Affiliation(s)
- Larry Bodgi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jolie Bou-Gharios
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Joyce Azzi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rafka Challita
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Charbel Feghaly
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Khanom Baalbaki
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hussein Kharroubi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fatima Chhade
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Fady Geara
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Zeina Ayoub
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
4
|
Tawk B, Rein K, Schwager C, Knoll M, Wirkner U, Hörner-Rieber J, Liermann J, Kurth I, Balermpas P, Rödel C, Linge A, Löck S, Lohaus F, Tinhofer I, Krause M, Stuschke M, Grosu AL, Zips D, Combs SE, Belka C, Stenzinger A, Herold-Mende C, Baumann M, Schirmacher P, Debus J, Abdollahi A. DNA-Methylome-Based Tumor Hypoxia Classifier Identifies HPV-Negative Head and Neck Cancer Patients at Risk for Locoregional Recurrence after Primary Radiochemotherapy. Clin Cancer Res 2023; 29:3051-3064. [PMID: 37058257 PMCID: PMC10425733 DOI: 10.1158/1078-0432.ccr-22-3790] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/16/2023] [Accepted: 04/11/2023] [Indexed: 04/15/2023]
Abstract
PURPOSE Tumor hypoxia is a paradigmatic negative prognosticator of treatment resistance in head and neck squamous cell carcinoma (HNSCC). The lack of robust and reliable hypoxia classifiers limits the adaptation of stratified therapies. We hypothesized that the tumor DNA methylation landscape might indicate epigenetic reprogramming induced by chronic intratumoral hypoxia. EXPERIMENTAL DESIGN A DNA-methylome-based tumor hypoxia classifier (Hypoxia-M) was trained in the TCGA (The Cancer Genome Atlas)-HNSCC cohort based on matched assignments using gene expression-based signatures of hypoxia (Hypoxia-GES). Hypoxia-M was validated in a multicenter DKTK-ROG trial consisting of human papillomavirus (HPV)-negative patients with HNSCC treated with primary radiochemotherapy (RCHT). RESULTS Although hypoxia-GES failed to stratify patients in the DKTK-ROG, Hypoxia-M was independently prognostic for local recurrence (HR, 4.3; P = 0.001) and overall survival (HR, 2.34; P = 0.03) but not distant metastasis after RCHT in both cohorts. Hypoxia-M status was inversely associated with CD8 T-cell infiltration in both cohorts. Hypoxia-M was further prognostic in the TCGA-PanCancer cohort (HR, 1.83; P = 0.04), underscoring the breadth of this classifier for predicting tumor hypoxia status. CONCLUSIONS Our findings highlight an unexplored avenue for DNA methylation-based classifiers as biomarkers of tumoral hypoxia for identifying high-risk features in patients with HNSCC tumors. See related commentary by Heft Neal and Brenner, p. 2954.
Collapse
Affiliation(s)
- Bouchra Tawk
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katrin Rein
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Schwager
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maximilian Knoll
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ute Wirkner
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juliane Hörner-Rieber
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Liermann
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ina Kurth
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
| | - Panagiotis Balermpas
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), partner site, Frankfurt, Germany
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Claus Rödel
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), partner site, Frankfurt, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Annett Linge
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz Association and Helmholtz-Zentrum Dresden – Rossendorf (HZDR), Dresden, Germany
| | - Steffen Löck
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Fabian Lohaus
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz Association and Helmholtz-Zentrum Dresden – Rossendorf (HZDR), Dresden, Germany
| | - Ingeborg Tinhofer
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Berlin, Germany
- Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Mechtild Krause
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz Association and Helmholtz-Zentrum Dresden – Rossendorf (HZDR), Dresden, Germany
| | - Martin Stuschke
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Essen, Germany
- Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Anca Ligia Grosu
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Freiburg, Germany
- Department of Radiation Oncology, University of Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Berlin, Germany
- Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany, German Cancer Consortium (DKTK), partner site Tuebingen, Germany
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls Universität Tübingen, Germany
| | - Stephanie E. Combs
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Munich, Germany
- Department of Radiation Oncology, Technische Universität München, Munich, Germany
| | - Claus Belka
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Munich, Germany
- Department of Radiation Oncology, University Hospital Ludwig-Maximilians-University of Munich, Munich, Germany
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Albrecht Stenzinger
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Baumann
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany, and German Cancer Consortium (DKTK), partner site Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Schirmacher
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK), Core Center Heidelberg, Germany
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
5
|
Boopathi E, Den RB, Thangavel C. Innate Immune System in the Context of Radiation Therapy for Cancer. Cancers (Basel) 2023; 15:3972. [PMID: 37568788 PMCID: PMC10417569 DOI: 10.3390/cancers15153972] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Radiation therapy (RT) remains an integral component of modern oncology care, with most cancer patients receiving radiation as a part of their treatment plan. The main goal of ionizing RT is to control the local tumor burden by inducing DNA damage and apoptosis within the tumor cells. The advancement in RT, including intensity-modulated RT (IMRT), stereotactic body RT (SBRT), image-guided RT, and proton therapy, have increased the efficacy of RT, equipping clinicians with techniques to ensure precise and safe administration of radiation doses to tumor cells. In this review, we present the technological advancement in various types of RT methods and highlight their clinical utility and associated limitations. This review provides insights into how RT modulates innate immune signaling and the key players involved in modulating innate immune responses, which have not been well documented earlier. Apoptosis of cancer cells following RT triggers immune systems that contribute to the eradication of tumors through innate and adoptive immunity. The innate immune system consists of various cell types, including macrophages, dendritic cells, and natural killer cells, which serve as key mediators of innate immunity in response to RT. This review will concentrate on the significance of the innate myeloid and lymphoid lineages in anti-tumorigenic processes triggered by RT. Furthermore, we will explore essential strategies to enhance RT efficacy. This review can serve as a platform for researchers to comprehend the clinical application and limitations of various RT methods and provides insights into how RT modulates innate immune signaling.
Collapse
Affiliation(s)
- Ettickan Boopathi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert B. Den
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Chellappagounder Thangavel
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| |
Collapse
|
6
|
Li T, Wang J, Yang Y, Glide-Hurst CK, Wen N, Cai J. Multi-parametric MRI for radiotherapy simulation. Med Phys 2023; 50:5273-5293. [PMID: 36710376 PMCID: PMC10382603 DOI: 10.1002/mp.16256] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 09/10/2022] [Accepted: 12/06/2022] [Indexed: 01/31/2023] Open
Abstract
Magnetic resonance imaging (MRI) has become an important imaging modality in the field of radiotherapy (RT) in the past decade, especially with the development of various novel MRI and image-guidance techniques. In this review article, we will describe recent developments and discuss the applications of multi-parametric MRI (mpMRI) in RT simulation. In this review, mpMRI refers to a general and loose definition which includes various multi-contrast MRI techniques. Specifically, we will focus on the implementation, challenges, and future directions of mpMRI techniques for RT simulation.
Collapse
Affiliation(s)
- Tian Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jihong Wang
- Department of Radiation Physics, Division of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Yingli Yang
- Department of Radiology, Ruijin Hospital, Shanghai Jiaotong Univeristy School of Medicine, Shanghai, China
- SJTU-Ruijing-UIH Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Carri K Glide-Hurst
- Department of Radiation Oncology, University of Wisconsin, Madison, Wisconsin, USA
| | - Ning Wen
- Department of Radiology, Ruijin Hospital, Shanghai Jiaotong Univeristy School of Medicine, Shanghai, China
- SJTU-Ruijing-UIH Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- The Global Institute of Future Technology, Shanghai Jiaotong University, Shanghai, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
7
|
Krzykawska-Serda M, Szczygieł D, Gaweł S, Drzał A, Szczygieł M, Kmieć MM, Mackiewicz A, Kieda C, Elas M. Oxygen therapeutic window induced by myo-inositol trispyrophosphate (ITPP)-Local pO2 study in murine tumors. PLoS One 2023; 18:e0285318. [PMID: 37167239 PMCID: PMC10174508 DOI: 10.1371/journal.pone.0285318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/19/2023] [Indexed: 05/13/2023] Open
Abstract
Hypoxia, an inevitable feature of locally advanced solid tumors, has been known as an adverse prognostic factor, a driver of an aggressive phenotype, and an unfavorable factor in therapies. Myo-inositol trispyrophosphate (ITPP) is a hemoglobin modifier known to both increase O2 release and normalize microvasculature. Our goal was to measure the tumor oxygen partial pressure dynamic changes and timing of the therapeutic window after ITPP systemic administration. Two syngeneic tumor models in mice, B16 melanoma and 4T1 breast carcinoma, were used, with varying ITPP dose schedules. Tissue oxygenation level was measured over several days in situ in live animals by Electron Paramagnetic Resonance oximetry with implanted OxyChip used as a constant sensor of the local pO2 value. Both B16 and 4T1 tumors became more normoxic after ITPP treatment, with pO2 levels elevated by 10-20 mm Hg compared to the control. The increase in pO2 was either transient or sustained, and the underlying mechanism relied on shifting hypoxic tumor areas to normoxia. The effect depended on ITPP delivery intervals regarding the tumor type and growth rate. Moreover, hypoxic tumors before treatment responded better than normoxic ones. In conclusion, the ITPP-generated oxygen therapeutic window may be valuable for anti-tumor therapies requiring oxygen, such as radio-, photo- or immunotherapy. Furthermore, such a combinatory treatment can be especially beneficial for hypoxic tumors.
Collapse
Affiliation(s)
- Martyna Krzykawska-Serda
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, Kraków, Poland
| | - Dariusz Szczygieł
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, Kraków, Poland
| | - Szymon Gaweł
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, Kraków, Poland
| | - Agnieszka Drzał
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, Kraków, Poland
| | - Małgorzata Szczygieł
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, Kraków, Poland
| | - Maciej M Kmieć
- Department of Radiology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Greater Poland Cancer Centre, Poznan University of Medical Sciences, Chair of Medical Biotechnology, Poznan, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine -National Research Institute, Warsaw, Poland
- Center for Molecular Biophysics UPR 4301 CNRS, 45071, Orleans, France
- Department of Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Martyna Elas
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
8
|
Hartford AC, Gill GS, Ravi D, Tosteson TD, Li Z, Russo G, Eskey CJ, Jarvis LA, Simmons NE, Evans LT, Williams BB, Gladstone DJ, Roberts DW, Buckey JC. Sensitizing brain metastases to stereotactic radiosurgery using hyperbaric oxygen: A proof-of-principle study. Radiother Oncol 2022; 177:179-184. [PMID: 36404528 PMCID: PMC10827304 DOI: 10.1016/j.radonc.2022.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 11/07/2022]
Abstract
PURPOSE Increased oxygen levels may enhance the radiosensitivity of brain metastases treated with stereotactic radiosurgery (SRS). This project administered hyperbaric oxygen (HBO) prior to SRS to assess feasibility, safety, and response. METHODS 38 patients were studied, 19 with 25 brain metastases treated with HBO prior to SRS, and 19 historical controls with 27 metastases, matched for histology, GPA, resection status, and lesion size. Outcomes included time from HBO to SRS, quality-of-life (QOL) measures, local control, distant (brain) metastases, radionecrosis, and overall survival. RESULTS The average time from HBO chamber to SRS beam-on was 8.3 ± 1.7 minutes. Solicited adverse events (AEs) were comparable between HBO and control patients; no grade III or IV serious AEs were observed. Radionecrosis-free survival (RNFS), radionecrosis-free survival before whole-brain radiation therapy (WBRT) (RNBWFS), local recurrence-free survival before WBRT (LRBWFS), distant recurrence-free survival before WBRT (DRBWFS), and overall survival (OS) were not significantly different for HBO patients and controls on Kaplan-Meier analysis, though at 1-year estimated survival rates trended in favor of SRS + HBO: RNFS - 83% vs 60%; RNBWFS - 78% vs 60%; LRBWFS - 95% vs 78%; DRBWFS - 61% vs 57%; and OS - 73% vs 56%. Multivariate Cox models indicated no significant association between HBO treatment and hazards of RN, local or distant recurrence, or mortality; however, these did show statistically significant associations (p < 0.05) for: local recurrence with higher volume, radionecrosis with tumor resection, overall survival with resection, and overall survival with higher GPA. CONCLUSION Addition of HBO to SRS for brain metastases is feasible without evident decrement in radiation necrosis and other clinical outcomes.
Collapse
Affiliation(s)
- Alan C Hartford
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA.
| | - Gobind S Gill
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Divya Ravi
- Dartmouth Cancer Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Tor D Tosteson
- Dartmouth Cancer Center, One Medical Center Drive, Lebanon, NH 03756, USA.
| | - Zhongze Li
- Dartmouth Cancer Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Gregory Russo
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Clifford J Eskey
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Lesley A Jarvis
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Nathan E Simmons
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Linton T Evans
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Benjamin B Williams
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - David J Gladstone
- Dartmouth Cancer Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - David W Roberts
- Dartmouth Cancer Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Jay C Buckey
- Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| |
Collapse
|
9
|
Moss DY, McCann C, Kerr EM. Rerouting the drug response: Overcoming metabolic adaptation in KRAS-mutant cancers. Sci Signal 2022; 15:eabj3490. [PMID: 36256706 DOI: 10.1126/scisignal.abj3490] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mutations in guanosine triphosphatase KRAS are common in lung, colorectal, and pancreatic cancers. The constitutive activity of mutant KRAS and its downstream signaling pathways induces metabolic rewiring in tumor cells that can promote resistance to existing therapeutics. In this review, we discuss the metabolic pathways that are altered in response to treatment and those that can, in turn, alter treatment efficacy, as well as the role of metabolism in the tumor microenvironment (TME) in dictating the therapeutic response in KRAS-driven cancers. We highlight metabolic targets that may provide clinical opportunities to overcome therapeutic resistance and improve survival in patients with these aggressive cancers.
Collapse
Affiliation(s)
- Deborah Y Moss
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| | - Christopher McCann
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| | - Emma M Kerr
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE Northern Ireland, UK
| |
Collapse
|
10
|
Radiation Treatment Timing and Dose Delivery: Effects on Bladder Cancer Cells in 3D in Vitro Culture. RADIATION 2022. [DOI: 10.3390/radiation2040025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
While radical cystectomy remains the primary treatment of choice for bladder cancer, increased evidence supports the use of bladder-preservation strategies based on adjuvant radiotherapy. This highlights the need for a better understanding of bladder cancer radiosensitivity to different types of treatment deliveries. The purpose of this study is to analyze the effect of treatment time, dose and fractionation on the number and sizes of grown three-dimensional (3D) bladder cancer spheres, and to assess the capacity of the linear-quadratic model in describing the response of cells cultured in 3D. 3D MatrigelTM-based cultures were employed to enrich for cancer stem cells (CSCs) from three human bladder cancer cell lines, RT4, T24 and UM-UC-3. Three single dose radiation treatments were performed at different time points after plating, and sphere number and sizes were assessed. Anti-CD44 immunofluorescence, clonogenic assay and anti-γH2AX staining were also performed to analyze the cell lines’ radiosensitivity. The radiosensitivity of spheres was dependent on the treatment timing after plating. Current linear quadratic dose fractionation models were shown to over-estimate radiosensitivity in 3D models. Our results showed the importance of treatment timing on the radio-response of bladder cancer spheres. We also demonstrated that bladder cancer spheres are more resistant to dose-fractionation than the estimation from the theoretical linear-quadratic model.
Collapse
|
11
|
Wang W, Wang X, Tao F, Hu K, Zhang J, Wu J, You L, Zhao W. Fluorinated Hyaluronic Acid Encapsulated Perfluorocarbon Nanoparticles as Tumor-Targeted Oxygen Carriers to Enhance Radiotherapy. Mol Pharm 2022; 19:3948-3958. [PMID: 36194775 DOI: 10.1021/acs.molpharmaceut.2c00432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The efficacy of radiotherapy is significantly constricted by tumor hypoxia. To overcome this obstacle, one promising approach is to use the perfluorocarbon-based O2 carriers combined with hyperoxic respiration to relieve tumor hypoxia. However, this passively transported oxygen carrier during hyperoxic respiration is prone to cause systemic oxidative stress and toxicity, which further limits its clinical application. Herein, we fabricate O2@PFC@FHA NPs for safe and specific oxygen delivery into tumors by using the fluorinated hyaluronic acid to encapsulate O2-saturated perfluorocarbon. Due to the interaction between HA and CD44 receptors, more FHA@PFC NPs accumulated in the tumor and the O2@PFC@FHA NPs significantly relieved tumor hypoxia. Notably, RT plus O2@PFC@FHA NPs resulted in almost threefold therapeutic improvement compared with RT without obvious systemic toxicity. Therefore, the O2@FHA@PFC NPs may have great potential to enhance the therapeutic efficacy of radiotherapy in the clinic.
Collapse
Affiliation(s)
- Wenguang Wang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Xingli Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Feng Tao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Kaiyuan Hu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Junying Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, Nanjing 210093, China.,Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
| | - Linjun You
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Weijun Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
12
|
Fletcher T, Thompson AJ, Ashrafian H, Darzi A. The measurement and modification of hypoxia in colorectal cancer: overlooked but not forgotten. Gastroenterol Rep (Oxf) 2022; 10:goac042. [PMID: 36032656 PMCID: PMC9406947 DOI: 10.1093/gastro/goac042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/18/2022] [Accepted: 07/21/2022] [Indexed: 11/14/2022] Open
Abstract
Tumour hypoxia is the inevitable consequence of a tumour's rapid growth and disorganized, inefficient vasculature. The compensatory mechanisms employed by tumours, and indeed the absence of oxygen itself, hinder the ability of all treatment modalities. The clinical consequence is poorer overall survival, disease-free survival, and locoregional control. Recognizing this, clinicians have been attenuating the effect of hypoxia, primarily with hypoxic modification or with hypoxia-activated pro-drugs, and notable success has been demonstrated. However, in the case of colorectal cancer (CRC), there is a general paucity of knowledge and evidence surrounding the measurement and modification of hypoxia, and this is possibly due to the comparative inaccessibility of such tumours. We specifically review the role of hypoxia in CRC and focus on the current evidence for the existence of hypoxia in CRC, the majority of which originates from indirect positron emission topography imaging with hypoxia selective radiotracers; the evidence correlating CRC hypoxia with poorer oncological outcome, which is largely based on the measurement of hypoxia inducible factor in correlation with clinical outcome; the evidence of hypoxic modification in CRC, of which no direct evidence exists, but is reflected in a number of indirect markers; the prognostic and monitoring implications of accurate CRC hypoxia quantification and its potential in the field of precision oncology; and the present and future imaging tools and technologies being developed for the measurement of CRC hypoxia, including the use of blood-oxygen-level-dependent magnetic resonance imaging and diffuse reflectance spectroscopy.
Collapse
Affiliation(s)
- Teddy Fletcher
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| | - Alex J Thompson
- The Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK
| | - Hutan Ashrafian
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| | - Ara Darzi
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| |
Collapse
|
13
|
Ionizing radiation induced DNA damage via ROS production in nano ozonized oil treated B-16 melanoma and OV-90 ovarian cells. Biochem Biophys Res Commun 2022; 615:143-149. [PMID: 35623299 DOI: 10.1016/j.bbrc.2022.05.030] [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: 04/19/2022] [Revised: 05/03/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022]
Abstract
In this study, we aimed to investigate ozonized oil nanoemulsions (OZNEs) as a radiosensitizer within B-16 melanoma and OV-90 ovarian cells under X-ray irradiation in vitro. Radiation sensitivity of OZNE treated B-16 melanoma cells and OV-90 ovarian cells were evaluated by performing cell cycle analysis, Reactive Oxygen Species (ROS) and ɣ-H2AX assays by flow cytometry. OZNEs induced G0-1 phase arrest of B-16 melanoma cells for all radiation doses and G2/M arrest for 8 Gy and 15 Gy doses. OZNE treated B-16 melanoma and OV-90 ovarian cells induced DNA damage via the increase in ROS production, as well as significant increase in the expression of ɣ-H2AX under even low doses of radiation (2 Gy). Thus, OZNEs are suggested to help to optimize cancer RT as a radiosensitizer and further studies will significantly outperform recent advances in this field.
Collapse
|
14
|
Shen R, Peng L, Zhou W, Wang D, Jiang Q, Ji J, Hu F, Yuan H. Anti-angiogenic nano-delivery system promotes tumor vascular normalizing and micro-environment reprogramming in solid tumor. J Control Release 2022; 349:550-564. [PMID: 35841997 DOI: 10.1016/j.jconrel.2022.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/27/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
Aberrant tumor vasculature leads to the malignant tumor microenvironment (TME) for tumor progression. Research has found temporary tumor vascular normalization after treated with low-dose anti-angiogenic agents, however, has paid little attention to prolonging the normalization window and its further influence on tumor tissue. Based on the dose- and time-dependent effect of anti-angiogenic agents, we developed V@LDL NPs, a nano-delivery system sustainedly releasing Vandetanib, an anti-VEGFR2 inhibitor, to control the dose of drug to the normalizing level, and prove its stable tumor vascular normalizing effect in 4 T1 breast cancer model. Furthermore, long-term normalized vasculature could improve tumor perfusion, then provide a circulation to reverse abnormalities in TME, such as hypoxia and heterogeneity, and also inhibit tumor progression. Our findings demonstrate that stable tumor vascular normalization could be a considerable strategy for long-term change to remodel TME and probably result in a therapeutic benefit to anti-cancer treatment, which could be achieved by anti-angiogenic nano-delivery system.
Collapse
Affiliation(s)
- Ruoyu Shen
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Lijun Peng
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Wentao Zhou
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Ding Wang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Qi Jiang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Jian Ji
- Department of Polymer Science and Engineering, Zhejiang University, 38 Zhe Da Road, Hangzhou 310027, Zhejiang Province, People's Republic of China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China.
| |
Collapse
|
15
|
Lefèvre AC, Alsner J, Sørensen BS, Tramm T, Toustrup K, Overgaard J, Spindler KLG. Hypoxia and local tumour control in squamous cell carcinoma of the anus - a hypothesis-generating study. Acta Oncol 2022; 61:1132-1135. [PMID: 35730972 DOI: 10.1080/0284186x.2022.2089591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Anna Cecilie Lefèvre
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Alsner
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Brita Singers Sørensen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Trine Tramm
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Kasper Toustrup
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Karen-Lise Garm Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
16
|
Dewhirst MW, Oleson JR, Kirkpatrick J, Secomb TW. Accurate Three-Dimensional Thermal Dosimetry and Assessment of Physiologic Response Are Essential for Optimizing Thermoradiotherapy. Cancers (Basel) 2022; 14:1701. [PMID: 35406473 PMCID: PMC8997141 DOI: 10.3390/cancers14071701] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Numerous randomized trials have revealed that hyperthermia (HT) + radiotherapy or chemotherapy improves local tumor control, progression free and overall survival vs. radiotherapy or chemotherapy alone. Despite these successes, however, some individuals fail combination therapy; not every patient will obtain maximal benefit from HT. There are many potential reasons for failure. In this paper, we focus on how HT influences tumor hypoxia, since hypoxia negatively influences radiotherapy and chemotherapy response as well as immune surveillance. Pre-clinically, it is well established that reoxygenation of tumors in response to HT is related to the time and temperature of exposure. In most pre-clinical studies, reoxygenation occurs only during or shortly after a HT treatment. If this were the case clinically, then it would be challenging to take advantage of HT induced reoxygenation. An important question, therefore, is whether HT induced reoxygenation occurs in the clinic that is of radiobiological significance. In this review, we will discuss the influence of thermal history on reoxygenation in both human and canine cancers treated with thermoradiotherapy. Results of several clinical series show that reoxygenation is observed and persists for 24-48 h after HT. Further, reoxygenation is associated with treatment outcome in thermoradiotherapy trials as assessed by: (1) a doubling of pathologic complete response (pCR) in human soft tissue sarcomas, (2) a 14 mmHg increase in pO2 of locally advanced breast cancers achieving a clinical response vs. a 9 mmHg decrease in pO2 of locally advanced breast cancers that did not respond and (3) a significant correlation between extent of reoxygenation (as assessed by pO2 probes and hypoxia marker drug immunohistochemistry) and duration of local tumor control in canine soft tissue sarcomas. The persistence of reoxygenation out to 24-48 h post HT is distinctly different from most reported rodent studies. In these clinical series, comparison of thermal data with physiologic response shows that within the same tumor, temperatures at the higher end of the temperature distribution likely kill cells, resulting in reduced oxygen consumption rate, while lower temperatures in the same tumor improve perfusion. However, reoxygenation does not occur in all subjects, leading to significant uncertainty about the thermal-physiologic relationship. This uncertainty stems from limited knowledge about the spatiotemporal characteristics of temperature and physiologic response. We conclude with recommendations for future research with emphasis on retrieving co-registered thermal and physiologic data before and after HT in order to begin to unravel complex thermophysiologic interactions that appear to occur with thermoradiotherapy.
Collapse
Affiliation(s)
- Mark W Dewhirst
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - James R Oleson
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - John Kirkpatrick
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
17
|
Wegge M, Dok R, Nuyts S. Hypoxia and Its Influence on Radiotherapy Response of HPV-Positive and HPV-Negative Head and Neck Cancer. Cancers (Basel) 2021; 13:5959. [PMID: 34885069 PMCID: PMC8656584 DOI: 10.3390/cancers13235959] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cancers are a heterogeneous group of cancers that arise from the upper aerodigestive tract. Etiologically, these tumors are linked to alcohol/tobacco abuse and infections with high-risk human papillomavirus (HPV). HPV-positive HNSCCs are characterized by a different biology and also demonstrate better therapy response and survival compared to alcohol/tobacco-related HNSCCs. Despite this advantageous therapy response and the clear biological differences, all locally advanced HNSCCs are treated with the same chemo-radiotherapy schedules. Although we have a better understanding of the biology of both groups of HNSCC, the biological factors associated with the increased radiotherapy response are still unclear. Hypoxia, i.e., low oxygen levels because of an imbalance between oxygen demand and supply, is an important biological factor associated with radiotherapy response and has been linked with HPV infections. In this review, we discuss the effects of hypoxia on radiotherapy response, on the tumor biology, and the tumor microenvironment of HPV-positive and HPV-negative HNSCCs by pointing out the differences between these two tumor types. In addition, we provide an overview of the current strategies to detect and target hypoxia.
Collapse
Affiliation(s)
- Marilyn Wegge
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven, 3000 Leuven, Belgium; (M.W.); (R.D.)
| | - Rüveyda Dok
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven, 3000 Leuven, Belgium; (M.W.); (R.D.)
| | - Sandra Nuyts
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven, 3000 Leuven, Belgium; (M.W.); (R.D.)
- Department of Radiation Oncology, Leuven Cancer Institute, UZ Leuven, 3000 Leuven, Belgium
| |
Collapse
|
18
|
Yang X, Wang Y, Mao T, Wang Y, Liu R, Yu L, Ding J. An oxygen-enriched thermosensitive hydrogel for the relief of a hypoxic tumor microenvironment and enhancement of radiotherapy. Biomater Sci 2021; 9:7471-7482. [PMID: 34617528 DOI: 10.1039/d1bm01280k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The rapid proliferation of tumor cells and tortuous vasculature in solid tumors often bring about a hypoxic tumor microenvironment, which renders tumor cells more resistant to many cancer treatments, including radiotherapy. In this study, an injectable and thermosensitive composite hydrogel composed of perfluorooctanoic acid (PFOA) modified monomethoxy poly(ethylene glycol)-poly(D,L-lactide-co-glycolide) (mPEG-PLGA-PFOA) and perfluorooctyl bromide (PFOB) that presented a thermoreversible sol-gel transition upon heating was developed to deliver exogenous oxygen for the relief of tumor hypoxia and enhancement of radiotherapy. The fluorinated modification of copolymers significantly increased the stability of PFOB in the mPEG-PLGA-PFOA aqueous solution owing to the fluorophilic interaction between PFOB and PFOA-modified copolymers. The introduction of PFOB not only efficiently heightened the oxygen loading capacity of the composite hydrogel, but also endowed it with excellent X-ray opacity, allowing the visual observation of the hydrogel via micro-CT imaging. After peritumoral injection of the oxygen-enriched composite hydrogel, the continuous supply of oxygen effectively relieved tumor hypoxia and down-regulated the expression of hypoxia-inducible factor-1α. Profiting from this, the hyposensitivity of tumor cells to radiation was successfully reversed, and the tumor growth in mice was significantly suppressed and the survival of mice was prolonged when combined with multiple X-ray exposure. As a result, the oxygen-enriched composite hydrogel shows a great potential for radiosensitization to improve the radiotherapeutic efficacy.
Collapse
Affiliation(s)
- Xiaowei Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital, Fudan University, Shanghai 200438, China.
| | - Yaoben Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital, Fudan University, Shanghai 200438, China.
| | - Tianjiao Mao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital, Fudan University, Shanghai 200438, China.
| | - Yang Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital, Fudan University, Shanghai 200438, China.
| | - Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital, Fudan University, Shanghai 200438, China.
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital, Fudan University, Shanghai 200438, China.
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital, Fudan University, Shanghai 200438, China.
| |
Collapse
|
19
|
Togni L, Mascitti M, Sartini D, Campagna R, Pozzi V, Salvolini E, Offidani A, Santarelli A, Emanuelli M. Nicotinamide N-Methyltransferase in Head and Neck Tumors: A Comprehensive Review. Biomolecules 2021; 11:1594. [PMID: 34827592 PMCID: PMC8615955 DOI: 10.3390/biom11111594] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/31/2022] Open
Abstract
The head and neck tumors (HNT) are a heterogeneous group of diseases ranging from benign to malignant lesions, with distinctive molecular and clinical behaviors. Several studies have highlighted the presence of an altered metabolic phenotype in HNT, such as the upregulation of nicotinamide N-methyltransferase (NNMT). However, its biological effects have not been completely disclosed and the role of NNMT in cancer cell metabolism remains unclear. Therefore, this comprehensive review aims to evaluate the available literature regarding the biological, diagnostic, and prognostic role of NNMT in HNT. NNMT was shown to be significantly overexpressed in all of the evaluated HNT types. Moreover, its upregulation has been correlated with cancer cell migration and adverse clinical outcomes, such as high-pathological stage, lymph node metastasis, and locoregional recurrences. However, in oral squamous cell carcinoma (OSCC) these associations are still debated, and several studies have failed to demonstrate the prognostic significance of NNMT. The shRNA-mediated gene silencing efficiently suppressed the NNMT gene expression and exhibited a clear inhibitory effect on cell proliferation, promoting the expression of apoptosis-related proteins and modulating the cell cycle. NNMT could represent a new molecular biomarker and a new target of molecular-based therapy, although further studies on larger patient cohorts are needed to explore its biological role in HNT.
Collapse
Affiliation(s)
- Lucrezia Togni
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Marco Mascitti
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Davide Sartini
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Roberto Campagna
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Valentina Pozzi
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Eleonora Salvolini
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| | - Annamaria Offidani
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy;
| | - Andrea Santarelli
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
- Dentistry Clinic, National Institute of Health and Science of Aging, IRCCS INRCA, 60126 Ancona, Italy
| | - Monica Emanuelli
- Department of Clinical, Specialistic and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (L.T.); (M.M.); (D.S.); (R.C.); (V.P.); (E.S.); (M.E.)
| |
Collapse
|
20
|
Alghamdi M, Gumbleton M, Newland B. Local delivery to malignant brain tumors: potential biomaterial-based therapeutic/adjuvant strategies. Biomater Sci 2021; 9:6037-6051. [PMID: 34357362 DOI: 10.1039/d1bm00896j] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most aggressive malignant brain tumor and is associated with a very poor prognosis. The standard treatment for newly diagnosed patients involves total tumor surgical resection (if possible), plus irradiation and adjuvant chemotherapy. Despite treatment, the prognosis is still poor, and the tumor often recurs within two centimeters of the original tumor. A promising approach to improving the efficacy of GBM therapeutics is to utilize biomaterials to deliver them locally at the tumor site. Local delivery to GBM offers several advantages over systemic administration, such as bypassing the blood-brain barrier and increasing the bioavailability of the therapeutic at the tumor site without causing systemic toxicity. Local delivery may also combat tumor recurrence by maintaining sufficient drug concentrations at and surrounding the original tumor area. Herein, we critically appraised the literature on local delivery systems based within the following categories: polymer-based implantable devices, polymeric injectable systems, and hydrogel drug delivery systems. We also discussed the negative effect of hypoxia on treatment strategies and how one might utilize local implantation of oxygen-generating biomaterials as an adjuvant to enhance current therapeutic strategies.
Collapse
Affiliation(s)
- Majed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Faculty of Pharmacy, King Abdulaziz University, Jeddah, 22522, Kingdom of Saudi Arabia
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| |
Collapse
|
21
|
Magnussen AL, Mills IG. Vascular normalisation as the stepping stone into tumour microenvironment transformation. Br J Cancer 2021; 125:324-336. [PMID: 33828258 PMCID: PMC8329166 DOI: 10.1038/s41416-021-01330-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/17/2021] [Accepted: 02/17/2021] [Indexed: 02/01/2023] Open
Abstract
A functional vascular system is indispensable for drug delivery and fundamental for responsiveness of the tumour microenvironment to such medication. At the same time, the progression of a tumour is defined by the interactions of the cancer cells with their surrounding environment, including neovessels, and the vascular network continues to be the major route for the dissemination of tumour cells in cancer, facilitating metastasis. So how can this apparent conflict be reconciled? Vessel normalisation-in which redundant structures are pruned and the abnormal vasculature is stabilised and remodelled-is generally considered to be beneficial in the course of anti-cancer treatments. A causality between normalised vasculature and improved response to medication and treatment is observed. For this reason, it is important to discern the consequence of vessel normalisation on the tumour microenvironment and to modulate the vasculature advantageously. This article will highlight the challenges of controlled neovascular remodelling and outline how vascular normalisation can shape disease management.
Collapse
Affiliation(s)
- Anette L Magnussen
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.
- Patrick G Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast, UK.
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway.
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
22
|
Lodhi T, Song YP, West C, Hoskin P, Choudhury A. Hypoxia and its Modification in Bladder Cancer: Current and Future Perspectives. Clin Oncol (R Coll Radiol) 2021; 33:376-390. [PMID: 33762140 DOI: 10.1016/j.clon.2021.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023]
Abstract
Radiotherapy plays an essential role in the curative treatment of muscle-invasive bladder cancer (MIBC). Hypoxia affects the response to MIBC radiotherapy, limiting radiocurability. Likewise, hypoxia influences MIBC genetic instability and malignant progression being associated with metastatic disease and a worse prognosis. Hypoxia identification in MIBC enables treatment stratification and the promise of improved survival. The most promising methods are histopathological markers such as necrosis; biomarkers of protein expression such as HIF-1α, GLUT-1 and CAIX; microRNAs; and novel mRNA signatures. Although hypoxia modification can take different forms, the gold standard remains carbogen and nicotinamide, which improve local control rates in bladder preservation and absolute overall survival with no significant increase in late toxicity. This is an exciting time for evolving therapies such as bioreductive agents, novel oxygen delivery techniques, immunotherapy and poly (ADP-ribose) polymerase 1 (PARP) inhibitors, all in development and representing upcoming trends in MIBC hypoxia modification. Whatever the future holds for hypoxia-modified radiotherapy, there is no doubt of its importance in MIBC. mRNA signatures provide an ideal platform for the selection of those with hypoxic tumours but are yet to qualified and integrated into the clinic. Future interventional trials will require biomarker stratification to ensure optimal treatment response to improve outcomes for patients with MIBC.
Collapse
Affiliation(s)
- T Lodhi
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Y P Song
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - C West
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, The Christie NHS Foundation Trust, Manchester, UK
| | - P Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, The Christie NHS Foundation Trust, Manchester, UK; Cancer Centre, Mount Vernon Hospital, Northwood, UK
| | - A Choudhury
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
23
|
Otazo R, Lambin P, Pignol JP, Ladd ME, Schlemmer HP, Baumann M, Hricak H. MRI-guided Radiation Therapy: An Emerging Paradigm in Adaptive Radiation Oncology. Radiology 2020; 298:248-260. [PMID: 33350894 DOI: 10.1148/radiol.2020202747] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Radiation therapy (RT) continues to be one of the mainstays of cancer treatment. Considerable efforts have been recently devoted to integrating MRI into clinical RT planning and monitoring. This integration, known as MRI-guided RT, has been motivated by the superior soft-tissue contrast, organ motion visualization, and ability to monitor tumor and tissue physiologic changes provided by MRI compared with CT. Offline MRI is already used for treatment planning at many institutions. Furthermore, MRI-guided linear accelerator systems, allowing use of MRI during treatment, enable improved adaptation to anatomic changes between RT fractions compared with CT guidance. Efforts are underway to develop real-time MRI-guided intrafraction adaptive RT of tumors affected by motion and MRI-derived biomarkers to monitor treatment response and potentially adapt treatment to physiologic changes. These developments in MRI guidance provide the basis for a paradigm change in treatment planning, monitoring, and adaptation. Key challenges to advancing MRI-guided RT include real-time volumetric anatomic imaging, addressing image distortion because of magnetic field inhomogeneities, reproducible quantitative imaging across different MRI systems, and biologic validation of quantitative imaging. This review describes emerging innovations in offline and online MRI-guided RT, exciting opportunities they offer for advancing research and clinical care, hurdles to be overcome, and the need for multidisciplinary collaboration.
Collapse
Affiliation(s)
- Ricardo Otazo
- From the Departments of Medical Physics (R.O.) and Radiology (R.O., H.H.), Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065; The D-Lab, Department of Precision Medicine, Department of Radiology & Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre, Maastricht, the Netherlands (P.L.); Department of Radiation Oncology, Dalhousie University, Halifax, Canada (J.P.P.); Divisions of Medical Physics in Radiology (M.E.L.), Radiology (H.P.S.), and Radiation Oncology/Radiobiology (M.B.), German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Physics and Astronomy (M.E.L.) and Faculty of Medicine (M.E.L., M.B.), Heidelberg University, Heidelberg, Germany; and OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany (M.B.)
| | - Philippe Lambin
- From the Departments of Medical Physics (R.O.) and Radiology (R.O., H.H.), Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065; The D-Lab, Department of Precision Medicine, Department of Radiology & Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre, Maastricht, the Netherlands (P.L.); Department of Radiation Oncology, Dalhousie University, Halifax, Canada (J.P.P.); Divisions of Medical Physics in Radiology (M.E.L.), Radiology (H.P.S.), and Radiation Oncology/Radiobiology (M.B.), German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Physics and Astronomy (M.E.L.) and Faculty of Medicine (M.E.L., M.B.), Heidelberg University, Heidelberg, Germany; and OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany (M.B.)
| | - Jean-Philippe Pignol
- From the Departments of Medical Physics (R.O.) and Radiology (R.O., H.H.), Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065; The D-Lab, Department of Precision Medicine, Department of Radiology & Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre, Maastricht, the Netherlands (P.L.); Department of Radiation Oncology, Dalhousie University, Halifax, Canada (J.P.P.); Divisions of Medical Physics in Radiology (M.E.L.), Radiology (H.P.S.), and Radiation Oncology/Radiobiology (M.B.), German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Physics and Astronomy (M.E.L.) and Faculty of Medicine (M.E.L., M.B.), Heidelberg University, Heidelberg, Germany; and OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany (M.B.)
| | - Mark E Ladd
- From the Departments of Medical Physics (R.O.) and Radiology (R.O., H.H.), Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065; The D-Lab, Department of Precision Medicine, Department of Radiology & Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre, Maastricht, the Netherlands (P.L.); Department of Radiation Oncology, Dalhousie University, Halifax, Canada (J.P.P.); Divisions of Medical Physics in Radiology (M.E.L.), Radiology (H.P.S.), and Radiation Oncology/Radiobiology (M.B.), German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Physics and Astronomy (M.E.L.) and Faculty of Medicine (M.E.L., M.B.), Heidelberg University, Heidelberg, Germany; and OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany (M.B.)
| | - Heinz-Peter Schlemmer
- From the Departments of Medical Physics (R.O.) and Radiology (R.O., H.H.), Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065; The D-Lab, Department of Precision Medicine, Department of Radiology & Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre, Maastricht, the Netherlands (P.L.); Department of Radiation Oncology, Dalhousie University, Halifax, Canada (J.P.P.); Divisions of Medical Physics in Radiology (M.E.L.), Radiology (H.P.S.), and Radiation Oncology/Radiobiology (M.B.), German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Physics and Astronomy (M.E.L.) and Faculty of Medicine (M.E.L., M.B.), Heidelberg University, Heidelberg, Germany; and OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany (M.B.)
| | - Michael Baumann
- From the Departments of Medical Physics (R.O.) and Radiology (R.O., H.H.), Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065; The D-Lab, Department of Precision Medicine, Department of Radiology & Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre, Maastricht, the Netherlands (P.L.); Department of Radiation Oncology, Dalhousie University, Halifax, Canada (J.P.P.); Divisions of Medical Physics in Radiology (M.E.L.), Radiology (H.P.S.), and Radiation Oncology/Radiobiology (M.B.), German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Physics and Astronomy (M.E.L.) and Faculty of Medicine (M.E.L., M.B.), Heidelberg University, Heidelberg, Germany; and OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany (M.B.)
| | - Hedvig Hricak
- From the Departments of Medical Physics (R.O.) and Radiology (R.O., H.H.), Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065; The D-Lab, Department of Precision Medicine, Department of Radiology & Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre, Maastricht, the Netherlands (P.L.); Department of Radiation Oncology, Dalhousie University, Halifax, Canada (J.P.P.); Divisions of Medical Physics in Radiology (M.E.L.), Radiology (H.P.S.), and Radiation Oncology/Radiobiology (M.B.), German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Physics and Astronomy (M.E.L.) and Faculty of Medicine (M.E.L., M.B.), Heidelberg University, Heidelberg, Germany; and OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany (M.B.)
| |
Collapse
|
24
|
Cao X, Allu SR, Jiang S, Gunn Bs JR, Yao PhD C, Xin PhD J, Bruza PhD P, Gladstone ScD DJ, Jarvis Md PhD LA, Tian PhD J, Swartz Md Msph PhD HM, Vinogradov PhD SA, Pogue PhD BW. High-Resolution pO 2 Imaging Improves Quantification of the Hypoxic Fraction in Tumors During Radiation Therapy. Int J Radiat Oncol Biol Phys 2020; 109:603-613. [PMID: 33002542 DOI: 10.1016/j.ijrobp.2020.09.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE The extreme microscopic heterogeneity of tumors makes it difficult to characterize tumor hypoxia. We evaluated how changes in the spatial resolution of oxygen imaging could alter measures of tumor hypoxia and their correlation to radiation therapy response. METHODS AND MATERIALS Cherenkov-Excited Luminescence Imaging in combination with an oxygen probe, Oxyphor PtG4 was used to directly image tumor pO2 distributions with 0.2 mm spatial resolution at the time of radiation delivery. These pO2 images were analyzed with variations of reduced spatial resolution from 0.2 mm to 5 mm, to investigate the influence of how reduced imaging spatial resolution would affect the observed tumor hypoxia. As an in vivo validation test, mice bearing tumor xenografts were imaged for hypoxic fraction and median pO2 to examine the predictive link with tumor response to radiation therapy, while accounting for spatial resolution. RESULTS In transitioning from voxel sizes of 200 μm to 3 mm, the median pO2 values increased by a few mm Hg, and the hypoxic fraction decreased by more than 50%. When looking at radiation-responsive tumors, the median pO2 values changed just a few mm Hg as a result of treatment, and the hypoxic fractions changed by as much as 50%. This latter change, however, could only be seen when sampling was performed with high spatial resolution. Median pO2 or similar quantities obtained from low resolution measurements are commonly used in clinical practice, however these parameters are much less sensitive to changes in the tumor microenvironment than the tumor hypoxic fraction obtained from high-resolution oxygen images. CONCLUSIONS This study supports the hypothesis that for adequate measurements of the tumor response to radiation therapy, oxygen imaging with high spatial resolution is required to accurately characterize the hypoxic fraction.
Collapse
Affiliation(s)
- Xu Cao
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire; Xidian University, Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xi'an, Shaanxi, China
| | - Srinivasa Rao Allu
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Chemistry, School or Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shudong Jiang
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire; Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Jason R Gunn Bs
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire
| | - Cuiping Yao PhD
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire; Xi'an Jiaotong University, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an, Shaanxi, China
| | - Jing Xin PhD
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire; Xi'an Jiaotong University, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an, Shaanxi, China
| | - Petr Bruza PhD
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire
| | - David J Gladstone ScD
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire; Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire; Department of Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Lesley A Jarvis Md PhD
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire; Department of Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Jie Tian PhD
- Xidian University, Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xi'an, Shaanxi, China; CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | | | - Sergei A Vinogradov PhD
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Chemistry, School or Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brian W Pogue PhD
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire; Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|
25
|
Shen H, Cook K, Gee HE, Hau E. Hypoxia, metabolism, and the circadian clock: new links to overcome radiation resistance in high-grade gliomas. J Exp Clin Cancer Res 2020; 39:129. [PMID: 32631383 PMCID: PMC7339573 DOI: 10.1186/s13046-020-01639-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is the cornerstone of treatment of high-grade gliomas (HGGs). It eradicates tumor cells by inducing oxidative stress and subsequent DNA damage. Unfortunately, almost all HGGs recur locally within several months secondary to radioresistance with intricate molecular mechanisms. Therefore, unravelling specific underlying mechanisms of radioresistance is critical to elucidating novel strategies to improve the radiosensitivity of tumor cells, and enhance the efficacy of radiotherapy. This review addresses our current understanding of how hypoxia and the hypoxia-inducible factor 1 (HIF-1) signaling pathway have a profound impact on the response of HGGs to radiotherapy. In addition, intriguing links between hypoxic signaling, circadian rhythms and cell metabolism have been recently discovered, which may provide insights into our fundamental understanding of radioresistance. Cellular pathways involved in the hypoxic response, DNA repair and metabolism can fluctuate over 24-h periods due to circadian regulation. These oscillatory patterns may have consequences for tumor radioresistance. Timing radiotherapy for specific times of the day (chronoradiotherapy) could be beneficial in patients with HGGs and will be discussed.
Collapse
Affiliation(s)
- Han Shen
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, Westmead, New South Wales, 2145, Australia.
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia.
| | - Kristina Cook
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health & Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Harriet E Gee
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, Westmead, New South Wales, 2145, Australia
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
- Department of Radiation Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, New South Wales, Australia
| | - Eric Hau
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, Westmead Institute for Medical Research, Westmead, New South Wales, 2145, Australia
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
- Department of Radiation Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, New South Wales, Australia
- Blacktown Hematology and Cancer Centre, Blacktown Hospital, Blacktown, New South Wales, Australia
| |
Collapse
|
26
|
Kery M, Papandreou I. Emerging strategies to target cancer metabolism and improve radiation therapy outcomes. Br J Radiol 2020; 93:20200067. [PMID: 32462882 DOI: 10.1259/bjr.20200067] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer-specific metabolic changes support the anabolic needs of the rapidly growing tumor, maintain a favorable redox balance, and help cells adapt to microenvironmental stresses like hypoxia and nutrient deprivation. Radiation is extensively applied in a large number of cancer treatment protocols but despite its curative potential, radiation resistance and treatment failures pose a serious problem. Metabolic control of DNA integrity and genomic stability can occur through multiple processes, encompassing cell cycle regulation, nucleotide synthesis, epigenetic regulation of gene activity, and antioxidant defenses. Given the important role of metabolic pathways in oxidative damage responses, it is necessary to assess the potential for tumor-specific radiosensitization by novel metabolism-targeted therapies. Additionally, there are opportunities to identify molecular and functional biomarkers of vulnerabilities to combination treatments, which could then inform clinical decisions. Here, we present a curated list of metabolic pathways in the context of ionizing radiation responses. Glutamine metabolism influences DNA damage responses by mechanisms such as synthesis of nucleotides for DNA repair or of glutathione for ROS detoxification. Repurposed oxygen consumption inhibitors have shown promising radiosensitizing activity against murine model tumors and are now in clinical trials. Production of 2-hydroxy glutarate by isocitrate dehydrogenase1/2 neomorphic oncogenic mutants interferes with the function of α-ketoglutarate-dependent enzymes and modulates Ataxia Telangiectasia Mutated (ATM) signaling and glutathione pools. Radiation-induced oxidative damage to membrane phospholipids promotes ferroptotic cell loss and cooperates with immunotherapies to improve tumor control. In summary, there are opportunities to enhance the efficacy of radiotherapy by exploiting cell-inherent vulnerabilities and dynamic microenvironmental components of the tumor.
Collapse
Affiliation(s)
| | - Ioanna Papandreou
- Department of Radiation Oncology, Wexner Medical Center and Comprehensive Cancer Center The Ohio State University Columbus, OH, USA
| |
Collapse
|
27
|
Kim MM, Parmar HA, Schipper M, Devasia T, Aryal MP, Kesari S, O'Day S, Morikawa A, Spratt DE, Junck L, Mammoser A, Hayman JA, Lawrence TS, Tsien CI, Aiken R, Goyal S, Abrouk N, Trimble M, Cao Y, Lao CD. BRAINSTORM: A Multi-Institutional Phase 1/2 Study of RRx-001 in Combination With Whole Brain Radiation Therapy for Patients With Brain Metastases. Int J Radiat Oncol Biol Phys 2020; 107:478-486. [PMID: 32169409 DOI: 10.1016/j.ijrobp.2020.02.639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 02/19/2020] [Accepted: 02/29/2020] [Indexed: 01/05/2023]
Abstract
PURPOSE To determine the recommended phase 2 dose of RRx-001, a radiosensitizer with vascular normalizing properties, when used with whole-brain radiation therapy (WBRT) for brain metastases and to assess whether quantitative changes in perfusion magnetic resonance imaging (MRI) after RRx-001 correlate with response. METHODS AND MATERIALS Five centers participated in this phase 1/2 trial of RRx-001 given once pre-WBRT and then twice weekly during WBRT. Four dose levels were planned (5 mg/m2, 8.4 mg/m2, 16.5 mg/m2, 27.5 mg/m2). Dose escalation was managed by the time-to-event continual reassessment method algorithm. Linear mixed models were used to correlate change in 24-hour T1, Ktrans (capillary permeability), and fractional plasma volume with change in tumor volume. RESULTS Between 2015 and 2017, 31 patients were enrolled. Two patients dropped out before any therapy. Median age was 60 years (range, 30-76), and 12 were male. The most common tumor types were melanoma (59%) and non-small cell lung cancer (18%). No dose limiting toxicities were observed. The most common severe adverse event was grade 3 asthenia (6.9%, 2 of 29). The median intracranial response rate was 46% (95% confidence interval, 24-68) and median overall survival was 5.2 months (95% confidence interval, 4.5-9.4). No neurologic deaths occurred. Among 10 patients undergoing dynamic contrast-enhanced MRI, a reduction in Vp 24 hours after RRx-001 was associated with reduced tumor volume at 1 and 4 months (P ≤ .01). CONCLUSIONS The addition of RRx-001 to WBRT is well tolerated with favorable intracranial response rates. Because activity was observed across all dose levels, the recommended phase 2 dose is 10 mg twice weekly. A reduction in fractional plasma volume on dynamic contrast-enhanced MRI 24 hours after RRx-001 suggests antiangiogenic activity associated with longer-term tumor response.
Collapse
Affiliation(s)
- Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
| | - Hemant A Parmar
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Matthew Schipper
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Theresa Devasia
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Madhava P Aryal
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Santosh Kesari
- Providence Saint John's Health Center, John Wayne Cancer Institute, Santa Monica, California
| | - Steven O'Day
- Providence Saint John's Health Center, John Wayne Cancer Institute, Santa Monica, California
| | - Aki Morikawa
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Larry Junck
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Aaron Mammoser
- Department of Neurosurgery, Louisiana State University, New Orleans, Louisiana
| | - James A Hayman
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Christina I Tsien
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Robert Aiken
- The Cancer Institute of New Jersey/Rutgers University, New Brunswick, New Jersey
| | - Sharad Goyal
- Department of Radiation Oncology, George Washington University, Washington, DC
| | - Nacer Abrouk
- Clinical Trials Innovations, Mountain View, California
| | | | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Christopher D Lao
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
28
|
Dunne M, Regenold M, Allen C. Hyperthermia can alter tumor physiology and improve chemo- and radio-therapy efficacy. Adv Drug Deliv Rev 2020; 163-164:98-124. [PMID: 32681862 DOI: 10.1016/j.addr.2020.07.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/20/2022]
Abstract
Hyperthermia has demonstrated clinical success in improving the efficacy of both chemo- and radio-therapy in solid tumors. Pre-clinical and clinical research studies have demonstrated that targeted hyperthermia can increase tumor blood flow and increase the perfused fraction of the tumor in a temperature and time dependent manner. Changes in tumor blood circulation can produce significant physiological changes including enhanced vascular permeability, increased oxygenation, decreased interstitial fluid pressure, and reestablishment of normal physiological pH conditions. These alterations in tumor physiology can positively impact both small molecule and nanomedicine chemotherapy accumulation and distribution within the tumor, as well as the fraction of the tumor susceptible to radiation therapy. Hyperthermia can trigger drug release from thermosensitive formulations and further improve the accumulation, distribution, and efficacy of chemotherapy.
Collapse
|
29
|
Garousi J, Huizing FJ, Vorobyeva A, Mitran B, Andersson KG, Leitao CD, Frejd FY, Löfblom J, Bussink J, Orlova A, Heskamp S, Tolmachev V. Comparative evaluation of affibody- and antibody fragments-based CAIX imaging probes in mice bearing renal cell carcinoma xenografts. Sci Rep 2019; 9:14907. [PMID: 31624303 PMCID: PMC6797765 DOI: 10.1038/s41598-019-51445-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Carbonic anhydrase IX (CAIX) is a cancer-associated molecular target for several classes of therapeutics. CAIX is overexpressed in a large fraction of renal cell carcinomas (RCC). Radionuclide molecular imaging of CAIX-expression might offer a non-invasive methodology for stratification of patients with disseminated RCC for CAIX-targeting therapeutics. Radiolabeled monoclonal antibodies and their fragments are actively investigated for imaging of CAIX expression. Promising alternatives are small non-immunoglobulin scaffold proteins, such as affibody molecules. A CAIX-targeting affibody ZCAIX:2 was re-designed with the aim to decrease off-target interactions and increase imaging contrast. The new tracer, DOTA-HE3-ZCAIX:2, was labeled with 111In and characterized in vitro. Tumor-targeting properties of [111In]In-DOTA-HE3-ZCAIX:2 were compared head-to-head with properties of the parental variant, [99mTc]Tc(CO)3-HE3-ZCAIX:2, and the most promising antibody fragment-based tracer, [111In]In-DTPA-G250(Fab’)2, in the same batch of nude mice bearing CAIX-expressing RCC xenografts. Compared to the 99mTc-labeled parental variant, [111In]In-DOTA-HE3-ZCAIX:2 provides significantly higher tumor-to-lung, tumor-to-bone and tumor-to-liver ratios, which is essential for imaging of CAIX expression in the major metastatic sites of RCC. [111In]In-DOTA-HE3-ZCAIX:2 offers significantly higher tumor-to-organ ratios compared with [111In]In-G250(Fab’)2. In conclusion, [111In]In-DOTA-HE3-ZCAIX:2 can be considered as a highly promising tracer for imaging of CAIX expression in RCC metastases based on our results and literature data.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fokko J Huizing
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bogdan Mitran
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ken G Andersson
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Charles Dahlsson Leitao
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Sandra Heskamp
- Department of Radiology and Nuclear medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
30
|
Forster JC, Marcu LG, Bezak E. Approaches to combat hypoxia in cancer therapy and the potential for in silico models in their evaluation. Phys Med 2019; 64:145-156. [PMID: 31515013 DOI: 10.1016/j.ejmp.2019.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/17/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023] Open
Abstract
AIM The negative impact of tumour hypoxia on cancer treatment outcome has been long-known, yet there has been little success combating it. This paper investigates the potential role of in silico modelling to help test emerging hypoxia-targeting treatments in cancer therapy. METHODS A Medline search was undertaken on the current landscape of in silico models that simulate cancer therapy and evaluate their ability to test hypoxia-targeting treatments. Techniques and treatments to combat tumour hypoxia and their current challenges are also presented. RESULTS Hypoxia-targeting treatments include tumour reoxygenation, hypoxic cell radiosensitization with nitroimidazoles, hypoxia-activated prodrugs and molecular targeting. Their main challenges are toxicity and not achieving adequate delivery to hypoxic regions of the tumour. There is promising research toward combining two or more of these techniques. Different types of in silico therapy models have been developed ranging from temporal to spatial and from stochastic to deterministic models. Numerous models have compared the effectiveness of different radiotherapy fractionation schedules for controlling hypoxic tumours. Similarly, models could help identify and optimize new treatments for overcoming hypoxia that utilize novel hypoxia-targeting technology. CONCLUSION Current therapy models should attempt to incorporate more sophisticated modelling of tumour angiogenesis/vasculature and vessel perfusion in order to become more useful for testing hypoxia-targeting treatments, which typically rely upon the tumour vasculature for delivery of additional oxygen, (pro)drugs and nanoparticles.
Collapse
Affiliation(s)
- Jake C Forster
- SA Medical Imaging, Department of Nuclear Medicine, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; Department of Physics, University of Adelaide, North Terrace, Adelaide SA 5005, Australia
| | - Loredana G Marcu
- Faculty of Science, University of Oradea, Oradea 410087, Romania; Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Eva Bezak
- Department of Physics, University of Adelaide, North Terrace, Adelaide SA 5005, Australia; Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide SA 5001, Australia
| |
Collapse
|
31
|
Yu P, Han X, Yin L, Hui K, Guo Y, Yuan A, Hu Y, Wu J. Artificial Red Blood Cells Constructed by Replacing Heme with Perfluorodecalin for Hypoxia‐Induced Radioresistance. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Peng Yu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Xiaoxue Han
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Lining Yin
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Kangyu Hui
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Yunfei Guo
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Jiangsu Provincial Key Laboratory for Nano TechnologyNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School of Nanjing University & School of Life SciencesNanjing University Nanjing 210093 China
- Jiangsu Provincial Key Laboratory for Nano TechnologyNanjing University Nanjing 210093 China
- Institute of Drug R&DNanjing University Nanjing 210093 China
| |
Collapse
|
32
|
Abstract
Data from observational studies indicate that both physical activity as well as exercise (ie, structured physical activity) is associated with reductions in the risk of recurrence and cancer mortality after a diagnosis of certain forms of cancer. Emerging evidence from preclinical studies indicates that physical activity/exercise paradigms regulate intratumoral vascular maturity and perfusion, hypoxia, and metabolism and augments the antitumor immune response. Such responses may, in turn, enhance response to standard anticancer treatments. For instance, exercise improves efficacy of chemotherapeutic agents, and there is rationale to believe that it will also improve radiotherapy response. This review overviews the current preclinical as well as clinical evidence supporting exercise modulation of therapeutic response and postulated biological mechanisms underpinning such effects. We also examine the implications for tumor response to radiation, chemotherapy, and immunotherapy.
Collapse
Affiliation(s)
- Kathleen A Ashcraft
- Departments of Radiation Oncology, Duke University School of Medicine, Durham, NC
| | | | - Lee W Jones
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.; Weill Cornell Medical College, New York, NY
| | - Mark W Dewhirst
- Departments of Radiation Oncology, Duke University School of Medicine, Durham, NC..
| |
Collapse
|
33
|
Elming PB, Sørensen BS, Oei AL, Franken NAP, Crezee J, Overgaard J, Horsman MR. Hyperthermia: The Optimal Treatment to Overcome Radiation Resistant Hypoxia. Cancers (Basel) 2019; 11:E60. [PMID: 30634444 PMCID: PMC6356970 DOI: 10.3390/cancers11010060] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/14/2018] [Accepted: 12/29/2018] [Indexed: 12/23/2022] Open
Abstract
Regions of low oxygenation (hypoxia) are a characteristic feature of solid tumors, and cells existing in these regions are a major factor influencing radiation resistance as well as playing a significant role in malignant progression. Consequently, numerous pre-clinical and clinical attempts have been made to try and overcome this hypoxia. These approaches involve improving oxygen availability, radio-sensitizing or killing the hypoxic cells, or utilizing high LET (linear energy transfer) radiation leading to a lower OER (oxygen enhancement ratio). Interestingly, hyperthermia (heat treatments of 39⁻45 °C) induces many of these effects. Specifically, it increases blood flow thereby improving tissue oxygenation, radio-sensitizes via DNA repair inhibition, and can kill cells either directly or indirectly by causing vascular damage. Combining hyperthermia with low LET radiation can even result in anti-tumor effects equivalent to those seen with high LET. The various mechanisms depend on the time and sequence between radiation and hyperthermia, the heating temperature, and the time of heating. We will discuss the role these factors play in influencing the interaction between hyperthermia and radiation, and summarize the randomized clinical trials showing a benefit of such a combination as well as suggest the potential future clinical application of this combination.
Collapse
Affiliation(s)
- Pernille B Elming
- Department of Experimental Clinical Oncology, Aarhus University Hospital, DK-8000 Aarhus C, Denmark.
| | - Brita S Sørensen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, DK-8000 Aarhus C, Denmark.
| | - Arlene L Oei
- Department of Radiation Oncology, Academic University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands.
| | - Nicolaas A P Franken
- Department of Radiation Oncology, Academic University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands.
| | - Johannes Crezee
- Department of Radiation Oncology, Academic University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands.
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, DK-8000 Aarhus C, Denmark.
| | - Michael R Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
34
|
Dewhirst MW, Mowery YM, Mitchell JB, Cherukuri MK, Secomb TW. Rationale for hypoxia assessment and amelioration for precision therapy and immunotherapy studies. J Clin Invest 2019; 129:489-491. [PMID: 30614815 DOI: 10.1172/jci126044] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Mark W Dewhirst
- Radiation Oncology Department, Duke University Medical School, Durham, North Carolina, USA
| | - Yvonne M Mowery
- Radiation Oncology Department, Duke University Medical School, Durham, North Carolina, USA
| | - James B Mitchell
- Radiation Biology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Murali K Cherukuri
- Radiation Biology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Timothy W Secomb
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona, USA
| |
Collapse
|
35
|
Clinical and Pre-clinical Methods for Quantifying Tumor Hypoxia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:19-41. [PMID: 31201714 DOI: 10.1007/978-3-030-12734-3_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, a prevalent characteristic of most solid malignant tumors, contributes to diminished therapeutic responses and more aggressive phenotypes. The term hypoxia has two definitions. One definition would be a physiologic state where the oxygen partial pressure is below the normal physiologic range. For most normal tissues, the normal physiologic range is between 10 and 20 mmHg. Hypoxic regions develop when there is an imbalance between oxygen supply and demand. The impact of hypoxia on cancer therapeutics is significant: hypoxic tissue is 3× less radiosensitive than normoxic tissue, the impaired blood flow found in hypoxic tumor regions influences chemotherapy delivery, and the immune system is dependent on oxygen for functionality. Despite the clinical implications of hypoxia, there is not a universal, ideal method for quantifying hypoxia, particularly cycling hypoxia because of its complexity and heterogeneity across tumor types and individuals. Most standard imaging techniques can be modified and applied to measuring hypoxia and quantifying its effects; however, the benefits and challenges of each imaging modality makes imaging hypoxia case-dependent. In this chapter, a comprehensive overview of the preclinical and clinical methods for quantifying hypoxia is presented along with the advantages and disadvantages of each.
Collapse
|
36
|
Yanazume S, Karakida N, Higashi R, Fukuda M, Togami S, Kamio M, Ota S, Kobayashi H. Tumor bleeding requiring intervention and the correlation with anemia in uterine cervical cancer for definitive radiotherapy. Jpn J Clin Oncol 2018; 48:892-899. [PMID: 30165631 DOI: 10.1093/jjco/hyy113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 07/17/2018] [Indexed: 01/31/2023] Open
Abstract
Background The prognostic impact of tumor bleeding requiring intervention and the correlation with anemia on the survival outcome of cervical cancer radiotherapy is unclear. Methods One hundred and ninety-six patients requiring hemostatic intervention between January 2006 and March 2014 were retrospectively investigated. The correlation between anemia and bleeding during radiotherapy, the prognostic impact of genital bleeding during radiotherapy and the influence of blood transfusion were estimated. Results None of the patients had incomplete or prolonged treatment exceeding 1 week due to bleeding. All tumor bleeding could be controlled by gauze packing, and no patients suffered from fatal genital bleeding. Bleeding significantly correlated with progression-free survival (P = 0.015) and overall survival (P = 0.048). Regarding the risk factors of anemia: age (P = 0.043), FIGO stage (P < 0.001), tumor diameter (P < 0.001), and bleeding (P = 0.002) were significant. Multivariate analysis revealed FIGO stage (Odds Ratio: 2.360; 95% CI = 1.202-4.633; P = 0.013), tumor diameter (Odds Ratio: 2.089; 95% CI = 1.048-4.162; P = 0.036) and Bleeding (Odds Ratio: 2.226; 95% CI = 1.052-4.709; P = 0.036) were independent to anemia. Anemia (Hazard Ratio = 1.894; 95% CI = 1.082-3.318; P = 0.025) was only independently correlated with progression free survival, while bleeding (Hazard Ratio = 1.156; 95% CI = 0.556-2.406; P = 0.698) had no independent correlation. Blood transfusion did not improve progression-free survival in patients with anemia or genital bleeding (P = 0.742). Conclusion We have proved that genital bleeding requiring intervention during cervical cancer radiotherapy is a negligible prognostic factor and is the independent factor for causing anemia. Easily bleeding tumors are potential prognostic markers, which are not effectively treated using existing radiotherapy.
Collapse
Affiliation(s)
- Shintaro Yanazume
- Department of Obstetrics & Gynecology, Faculty of Medicine, Kagoshima University
| | - Noriko Karakida
- Department of Gynecology, National Hospital Organization Kagoshima Medical Center
| | - Ryutaro Higashi
- Department of Radiology, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Mika Fukuda
- Department of Obstetrics & Gynecology, Faculty of Medicine, Kagoshima University
| | - Shinichi Togami
- Department of Obstetrics & Gynecology, Faculty of Medicine, Kagoshima University
| | - Masaki Kamio
- Department of Obstetrics & Gynecology, Faculty of Medicine, Kagoshima University
| | - Shunichiro Ota
- Department of Gynecology, National Hospital Organization Kagoshima Medical Center
| | - Hiroaki Kobayashi
- Department of Obstetrics & Gynecology, Faculty of Medicine, Kagoshima University
| |
Collapse
|
37
|
A potential solution for eliminating hypoxia as a cause for radioresistance. Proc Natl Acad Sci U S A 2018; 115:10548-10550. [PMID: 30301796 DOI: 10.1073/pnas.1814212115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
38
|
DNA Repair Deficient Chinese Hamster Ovary Cells Exhibiting Differential Sensitivity to Charged Particle Radiation under Aerobic and Hypoxic Conditions. Int J Mol Sci 2018; 19:ijms19082228. [PMID: 30061540 PMCID: PMC6121575 DOI: 10.3390/ijms19082228] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 12/31/2022] Open
Abstract
It has been well established that hypoxia significantly increases both cellular and tumor resistance to ionizing radiation. Hypoxia associated radiation resistance has been known for some time but there has been limited success in sensitizing cells to radiation under hypoxic conditions. These studies show that, when irradiated with low linear energy transfer (LET) gamma-rays, poly (ADP-ribose), polymerase (PARP), Fanconi Anemia (FANC), and mutant Chinese Hamster Ovary (CHO) cells respond similarly to the non-homologous end joining (NHEJ) and the homologous recombination (HR) repair mutant CHO cells. Comparable results were observed in cells exposed to 13 keV/μm carbon ions. However, when irradiated with higher LET spread out Bragg peak (SOBP) carbon ions, we observed a decrease in the oxygen enhancement ratio (OER) in all the DNA of repair mutant cell lines. Interestingly, PARP mutant cells were observed as having the largest decrease in OER. Finally, these studies show a significant increase in the relative biological effectiveness (RBE) of high LET SOBP carbon and iron ions in HR and PARP mutants. There was also an increase in the RBE of NHEJ mutants when irradiated to SOBP carbon and iron ions. However, this increase was lower than in other mutant cell lines. These findings indicate that high LET radiation produces unique types of DNA damage under hypoxic conditions and PARP and HR repair pathways play a role in repairing this damage.
Collapse
|
39
|
Bennett MH, Feldmeier J, Smee R, Milross C. Hyperbaric oxygenation for tumour sensitisation to radiotherapy. Cochrane Database Syst Rev 2018; 4:CD005007. [PMID: 29637538 PMCID: PMC6494427 DOI: 10.1002/14651858.cd005007.pub4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cancer is a common disease and radiotherapy is one well-established treatment for some solid tumours. Hyperbaric oxygenation therapy (HBOT) may improve the ability of radiotherapy to kill hypoxic cancer cells, so the administration of radiotherapy while breathing hyperbaric oxygen may result in a reduction in mortality and recurrence. OBJECTIVES To assess the benefits and harms of administering radiotherapy for the treatment of malignant tumours while breathing HBO. SEARCH METHODS In September 2017 we searched the Cochrane Central Register of Controlled Trials (CENTRAL), the Cochrane Library Issue 8, 2017, MEDLINE, Embase, and the Database of Randomised Trials in Hyperbaric Medicine using the same strategies used in 2011 and 2015, and examined the reference lists of included articles. SELECTION CRITERIA Randomised and quasi-randomised studies comparing the outcome of malignant tumours following radiation therapy while breathing HBO versus air or an alternative sensitising agent. DATA COLLECTION AND ANALYSIS Three review authors independently evaluated the quality of and extracted data from the included trials. MAIN RESULTS We included 19 trials in this review (2286 participants: 1103 allocated to HBOT and 1153 to control).For head and neck cancer, there was an overall reduction in the risk of dying at both one year and five years after therapy (risk ratio (RR) 0.83, 95% confidence interval (CI) 0.70 to 0.98, number needed to treat for an additional beneficial outcome (NNTB) = 11 and RR 0.82, 95% CI 0.69 to 0.98, high-quality evidence), and some evidence of improved local tumour control immediately following irradiation (RR with HBOT 0.58, 95% CI 0.39 to 0.85, moderate-quality evidence due to imprecision). There was a lower incidence of local recurrence of tumour when using HBOT at both one and five years (RR at one year 0.66, 95% CI 0.56 to 0.78, high-quality evidence; RR at five years 0.77, 95% CI 0.62 to 0.95, moderate-quality evidence due to inconsistency between trials). There was also some evidence with regard to the chance of metastasis at five years (RR with HBOT 0.45 95% CI 0.09 to 2.30, single trial moderate quality evidence imprecision). No trials reported a quality of life assessment. Any benefits come at the cost of an increased risk of severe local radiation reactions with HBOT (severe radiation reaction RR 2.64, 95% CI 1.65 to 4.23, high-quality evidence). However, the available evidence failed to clearly demonstrate an increased risk of seizures from acute oxygen toxicity (RR 4.3, 95% CI 0.47 to 39.6, moderate-quality evidence).For carcinoma of the uterine cervix, there was no clear benefit in terms of mortality at either one year or five years (RR with HBOT at one year 0.88, 95% CI 0.69 to 1.11, high-quality evidence; RR at five years 0.95, 95% CI 0.80 to 1.14, moderate-quality evidence due to inconsistency between trials). Similarly, there was no clear evidence of a benefit of HBOT in the reported rate of local recurrence (RR with HBOT at one year 0.82, 95% CI 0.63 to 1.06, high-quality evidence; RR at five years 0.85, 95% CI 0.65 to 1.13, moderate-quality evidence due to inconsistency between trials). We also found no clear evidence for any effect of HBOT on the rate of development of metastases at both two years and five years (two years RR with HBOT 1.05, 95% CI 0.84 to 1.31, high quality evidence; five years RR 0.79, 95% CI 0.50 to 1.26, moderate-quality evidence due to inconsistency). There were, however, increased adverse effects with HBOT. The risk of a severe radiation injury at the time of treatment with HBOT was 2.05, 95% CI 1.22 to 3.46, high-quality evidence. No trials reported any failure of local tumour control, quality of life assessments, or the risk of seizures during treatment.With regard to the treatment of urinary bladder cancer, there was no clear evidence of a benefit in terms of mortality from HBOT at one year (RR 0.97, 95% CI 0.74 to 1.27, high-quality evidence), nor any benefit in the risk of developing metastases at two years (RR 2.0, 95% CI 0.58 to 6.91, moderate-quality evidence due to imprecision). No trial reported on failure of local control, local recurrence, quality of life, or adverse effects.When all cancer types were combined, there was evidence for an increased risk of severe radiation tissue injury during the course of radiotherapy with HBOT (RR 2.35, 95% CI 1.66 to 3.33, high-quality evidence) and of oxygen toxic seizures during treatment (RR with HBOT 6.76, 96% CI 1.16 to 39.31, moderate-quality evidence due to imprecision). AUTHORS' CONCLUSIONS We found evidence that HBOT improves local tumour control, mortality, and local tumour recurrence for cancers of the head and neck. These benefits may only occur with unusual fractionation schemes. Hyperbaric oxygenation therapy is associated with severe tissue radiation injury. Given the methodological and reporting inadequacies of the included studies, our results demand a cautious interpretation. More research is needed for head and neck cancer, but is probably not justified for uterine cervical or bladder cancer. There is little evidence available concerning malignancies at other anatomical sites.
Collapse
Affiliation(s)
- Michael H Bennett
- Prince of Wales Clinical School, University of NSWDepartment of AnaesthesiaSydneyNSWAustralia
| | - John Feldmeier
- Medical College of OhioDepartment of Radiation OncologyToledoOhioUSA
| | - Robert Smee
- Prince of Wales HospitalDepartment of Radiation OncologyBarker StreetRandwickNSWAustralia2031
| | - Christopher Milross
- Chris O'Brien LifehouseRadiation Oncology and Medical Services119‐143 Missenden RoadPO Box M33 Missenden RoadCamperdownNSWAustralia2050
| | | |
Collapse
|
40
|
Verma A, Rich LJ, Vincent-Chong VK, Seshadri M. Visualizing the effects of metformin on tumor growth, vascularity, and metabolism in head and neck cancer. J Oral Pathol Med 2018; 47:484-491. [PMID: 29573032 DOI: 10.1111/jop.12705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND The antidiabetic drug metformin (Met) is believed to inhibit tumor proliferation by altering the metabolism of cancer cells. In this study, we examined the effects of Met on tumor oxygenation, metabolism, and growth in head and neck squamous cell carcinoma (HNSCC) using non-invasive multimodal imaging. MATERIALS AND METHODS Severe combined immunodeficient (SCID) mice bearing orthotopic FaDu HNSCC xenografts were treated with Met (200 mg/kg, ip) once daily for 5 days. Tumor oxygen saturation (%sO2 ) and hemoglobin concentration (HbT) were measured using photoacoustic imaging (PAI). Fluorescence imaging was employed to measure intratumoral uptake of 2-deoxyglucosone (2-DG) following Met treatment while magnetic resonance imaging (MRI) was utilized to measure tumor volume. Correlative immunostaining of tumor sections for markers of proliferation (Ki67) and vascularity (CD31) was also performed. RESULTS At 5 days post-Met treatment, PAI revealed a significant increase (P < .05) in %sO2 and HbT levels in treated tumors compared to untreated controls. Fluorescence imaging at this time point revealed a 46% decrease in mean 2-DG uptake compared to controls. No changes in hemodynamic parameters were observed in mouse salivary gland tissue. A significant decrease in Ki-67 staining (P < .001) and MR-based tumor volume was also observed in Met-treated tumors compared to controls with no change in CD31 + vessel count following Met therapy. CONCLUSION Our results provide, for the first time, direct in vivo evidence of Met-induced changes in tumor microenvironmental parameters in HNSCC xenografts. Our findings highlight the utility of multimodal functional imaging for non-invasive mapping of the effects of Met in HNSCC.
Collapse
Affiliation(s)
- Aparajita Verma
- Department of Oral Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Laurie J Rich
- Department of Oral Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Mukund Seshadri
- Department of Oral Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Molecular and Cellular Biophysics and Biochemistry, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Head and Neck Surgery, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
41
|
Apeke S, Gaubert L, Boussion N, Lambin P, Visvikis D, Rodin V, Redou P. Multi-Scale Modeling and Oxygen Impact on Tumor Temporal Evolution: Application on Rectal Cancer During Radiotherapy. IEEE TRANSACTIONS ON MEDICAL IMAGING 2018; 37:871-880. [PMID: 29610067 DOI: 10.1109/tmi.2017.2771379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
We present a multi-scale approach of tumor modeling in order to predict its evolution during radiotherapy. Within this context we focus on three different scales of tumor modeling: microscopic (individual cells in a voxel), mesoscopic (population of cells in a voxel) and macroscopic (whole tumor), with transition interfaces between these three scales. At the cellular level, the description is based on phase transfer probabilities in the cellular cycle. At the mesoscopic scale we represent populations of cells according to different stages in a cell cycle. Finally, at the macroscopic scale, the tumor description is based on the use of FDG PET image voxels. These three scales exist naturally: biological data are collected at the macroscopic scale, but the pathological behavior of the tumor is based on an abnormal cell-cycle at the microscopic scale. On the other hand, the introduction of a mesoscopic scale is essential in order to reduce the gap between the two extreme, in terms of resolution, description levels. It also reduces the computational burden of simulating a large number of individual cells. As an application of the proposed multi-scale model, we simulate the effect of oxygen on tumor evolution during radiotherapy. Two consecutive FDG PET images of 17 rectal cancer patients undergoing radiotherapy are used to simulate the tumor evolution during treatment. The simulated results are compared with those obtained on a third FDG PET image acquired two weeks after the beginning of the treatment.
Collapse
|
42
|
Effectiveness of radiotherapy + ozone on tumoral tissue and survival in tongue cancer rat model. Auris Nasus Larynx 2018; 45:128-134. [DOI: 10.1016/j.anl.2017.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/27/2017] [Accepted: 03/14/2017] [Indexed: 01/22/2023]
|
43
|
Zhou H, Arias-Ramos N, López-Larrubia P, Mason RP, Cerdán S, Pacheco-Torres J. Oxygenation Imaging by Nuclear Magnetic Resonance Methods. Methods Mol Biol 2018; 1718:297-313. [PMID: 29341016 DOI: 10.1007/978-1-4939-7531-0_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxygen monitoring is a topic of exhaustive research due to its central role in many biological processes, from energy metabolism to gene regulation. The ability to monitor in vivo the physiological distribution and the dynamics of oxygen from subcellular to macroscopic levels is a prerequisite to better understand the mechanisms associated with both normal and disease states (cancer, neurodegeneration, stroke, etc.). This chapter focuses on magnetic resonance imaging (MRI) based techniques to assess oxygenation in vivo. The first methodology uses injected fluorinated agents to provide quantitative pO2 measurements with high precision and suitable spatial and temporal resolution for many applications. The second method exploits changes in endogenous contrasts, i.e., deoxyhemoglobin and oxygen molecules through measurements of T 2* and T 1, in response to an intervention to qualitatively evaluate hypoxia and its potential modulation.
Collapse
Affiliation(s)
- Heling Zhou
- Prognostic Imaging Research Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Nuria Arias-Ramos
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas 'Alberto Sols' C.S.I.C./U.A.M., Madrid, Spain
| | - Ralph P Mason
- Prognostic Imaging Research Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas 'Alberto Sols' C.S.I.C./U.A.M., Madrid, Spain
| | - Jesús Pacheco-Torres
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, San Juan de Alicante, Alicante, Spain.
| |
Collapse
|
44
|
Szili EJ, Oh JS, Fukuhara H, Bhatia R, Gaur N, Nguyen CK, Hong SH, Ito S, Ogawa K, Kawada C, Shuin T, Tsuda M, Furihata M, Kurabayashi A, Furuta H, Ito M, Inoue K, Hatta A, Short RD. Modelling the helium plasma jet delivery of reactive species into a 3D cancer tumour. ACTA ACUST UNITED AC 2017. [DOI: 10.1088/1361-6595/aa9b3b] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
45
|
Wang H, Mu X, He H, Zhang XD. Cancer Radiosensitizers. Trends Pharmacol Sci 2017; 39:24-48. [PMID: 29224916 DOI: 10.1016/j.tips.2017.11.003] [Citation(s) in RCA: 339] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023]
Abstract
Radiotherapy (RT) is a mainstay treatment for many types of cancer, although it is still a large challenge to enhance radiation damage to tumor tissue and reduce side effects to healthy tissue. Radiosensitizers are promising agents that enhance injury to tumor tissue by accelerating DNA damage and producing free radicals. Several strategies have been exploited to develop highly effective and low-toxicity radiosensitizers. In this review, we highlight recent progress on radiosensitizers, including small molecules, macromolecules, and nanomaterials. First, small molecules are reviewed based on free radicals, pseudosubstrates, and other mechanisms. Second, nanomaterials, such as nanometallic materials, especially gold-based materials that have flexible surface engineering and favorable kinetic properties, have emerged as promising radiosensitizers. Finally, emerging macromolecules have shown significant advantages in RT because these molecules can be combined with biological therapy as well as drug delivery. Further research on the mechanisms of radioresistance and multidisciplinary approaches will accelerate the development of radiosensitizers.
Collapse
Affiliation(s)
- Hao Wang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Number 238, Baidi Road, Tianjin 300192, China; These authors have contributed equally
| | - Xiaoyu Mu
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China; These authors have contributed equally
| | - Hua He
- State Key Laboratory of Heavy Oil Processing and Center for Bioengineering and Biotechnology, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiao-Dong Zhang
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China; Tianjin Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, China.
| |
Collapse
|
46
|
Mistry IN, Thomas M, Calder EDD, Conway SJ, Hammond EM. Clinical Advances of Hypoxia-Activated Prodrugs in Combination With Radiation Therapy. Int J Radiat Oncol Biol Phys 2017; 98:1183-1196. [PMID: 28721903 DOI: 10.1016/j.ijrobp.2017.03.024] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/24/2017] [Accepted: 03/14/2017] [Indexed: 12/29/2022]
Abstract
With the increasing incidence of cancer worldwide, the need for specific, effective therapies is ever more urgent. One example of targeted cancer therapeutics is hypoxia-activated prodrugs (HAPs), also known as bioreductive prodrugs. These prodrugs are inactive in cells with normal oxygen levels but in hypoxic cells (with low oxygen levels) undergo chemical reduction to the active compound. Hypoxia is a common feature of solid tumors and is associated with a more aggressive phenotype and resistance to all modes of therapy. Therefore, the combination of radiation therapy and bioreductive drugs presents an attractive opportunity for synergistic effects, because the HAP targets the radiation-resistant hypoxic cells. Hypoxia-activated prodrugs have typically been precursors of DNA-damaging agents, but a new generation of molecularly targeted HAPs is emerging. By targeting proteins associated with tumorigenesis and survival, these compounds may result in greater selectivity over healthy tissue. We review the clinical progress of HAPs as adjuncts to radiation therapy and conclude that the use of HAPs alongside radiation is vastly underexplored at the clinical level.
Collapse
Affiliation(s)
- Ishna N Mistry
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Matthew Thomas
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ewen D D Calder
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Stuart J Conway
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Ester M Hammond
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
47
|
Panek R, Welsh L, Baker LCJ, Schmidt MA, Wong KH, Riddell AM, Koh DM, Dunlop A, Mcquaid D, d'Arcy JA, Bhide SA, Harrington KJ, Nutting CM, Hopkinson G, Richardson C, Box C, Eccles SA, Leach MO, Robinson SP, Newbold KL. Noninvasive Imaging of Cycling Hypoxia in Head and Neck Cancer Using Intrinsic Susceptibility MRI. Clin Cancer Res 2017; 23:4233-4241. [PMID: 28314789 PMCID: PMC5516915 DOI: 10.1158/1078-0432.ccr-16-1209] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/19/2016] [Accepted: 03/03/2017] [Indexed: 01/13/2023]
Abstract
Purpose: To evaluate intrinsic susceptibility (IS) MRI for the identification of cycling hypoxia, and the assessment of its extent and spatial distribution, in head and neck squamous cell carcinoma (HNSCC) xenografts and patients.Experimental Design: Quantitation of the transverse relaxation rate, R2*, which is sensitive to paramagnetic deoxyhemoglobin, using serial IS-MRI acquisitions, was used to monitor temporal oscillations in levels of paramagnetic deoxyhemoglobin in human CALR xenografts and patients with HNSCC at 3T. Autocovariance and power spectrum analysis of variations in R2* was performed for each imaged voxel, to assess statistical significance and frequencies of cycling changes in tumor blood oxygenation. Pathologic correlates with tumor perfusion (Hoechst 33342), hypoxia (pimonidazole), and vascular density (CD31) were sought in the xenografts, and dynamic contrast-enhanced (DCE) MRI was used to assess patient tumor vascularization. The prevalence of fluctuations within patient tumors, DCE parameters, and treatment outcome were reported.Results: Spontaneous R2* fluctuations with a median periodicity of 15 minutes were detected in both xenografts and patient tumors. Spatially, these fluctuations were predominantly associated with regions of heterogeneous perfusion and hypoxia in the CALR xenografts. In patients, R2* fluctuations spatially correlated with regions of lymph nodes with low Ktrans values, typically in the vicinity of necrotic cores.Conclusions: IS-MRI can be used to monitor variations in levels of paramagnetic deoxyhemoglobin, associated with cycling hypoxia. The presence of such fluctuations may be linked with impaired tumor vasculature, the presence of which may impact treatment outcome. Clin Cancer Res; 23(15); 4233-41. ©2017 AACR.
Collapse
Affiliation(s)
- Rafal Panek
- CR-UK and EPSRC Cancer Imaging Centre, London, United Kingdom
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Liam Welsh
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Lauren C J Baker
- CR-UK and EPSRC Cancer Imaging Centre, London, United Kingdom
- Institute of Cancer Research, London, United Kingdom
| | - Maria A Schmidt
- CR-UK and EPSRC Cancer Imaging Centre, London, United Kingdom
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Kee H Wong
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Angela M Riddell
- CR-UK and EPSRC Cancer Imaging Centre, London, United Kingdom
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Dow-Mu Koh
- CR-UK and EPSRC Cancer Imaging Centre, London, United Kingdom
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Alex Dunlop
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Dualta Mcquaid
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - James A d'Arcy
- CR-UK and EPSRC Cancer Imaging Centre, London, United Kingdom
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Shreerang A Bhide
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Kevin J Harrington
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | | | | | | | - Carol Box
- Institute of Cancer Research, London, United Kingdom
| | | | - Martin O Leach
- CR-UK and EPSRC Cancer Imaging Centre, London, United Kingdom.
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| | - Simon P Robinson
- CR-UK and EPSRC Cancer Imaging Centre, London, United Kingdom
- Institute of Cancer Research, London, United Kingdom
| | - Kate L Newbold
- Institute of Cancer Research, London, United Kingdom
- Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
48
|
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: 4.4] [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]
|
49
|
Moreno-Acosta P, Vallard A, Carrillo S, Gamboa O, Romero-Rojas A, Molano M, Acosta J, Mayorga D, Rancoule C, Garcia MA, Cotes Mestre M, Magné N. Biomarkers of resistance to radiation therapy: a prospective study in cervical carcinoma. Radiat Oncol 2017; 12:120. [PMID: 28716107 PMCID: PMC5514482 DOI: 10.1186/s13014-017-0856-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 07/12/2017] [Indexed: 12/19/2022] Open
Abstract
Background Clinical parameters and proteins have recently been suggested as possible causes of radiotherapy (RT) resistance in cervical carcinoma (CC). The objective of the present study was to validate prognostic biomarkers of radiation resistance. Methods The present prospective study included patients undergoing RT with curative intent for histologically proven locally advanced squamous cell CC. Tissues and blood samples were systematically collected before RT initiation. Immuno-histochemistry was performed (IGF-IR α and β, GAPDH, HIF-1 alpha, Survivin, GLUT1, CAIX, hTERT and HKII). Response to radiation was assessed through tumour response 3 months after RT completion, through overall survival (OS) and through progression-free survival (PFS). Results One hundred forty nine patients with a mean age of 46 years were included, with FIGO IIB (n = 53) and FIGO IIIB (n = 96) CCs. 61 patients were treated with exclusive RT + brachytherapy and 88 underwent chemo-radiotherapy + brachytherapy. Our findings suggest an association between hemoglobin level (Hb) (>11 g/dL) and 3 months complete response (p = 0.02). Hb level < 11 g/dL was associated with decreased PFS (p = 0.05) and OS (p = 0.08). Overexpression of IGF-1R β was correlated with a decreased OS (p = 0.007). Overexpression of GLUT1 was marginally correlated with reduced OS (p = 0.05). PFS and OS were significantly improved in patients undergoing chemoradiation versus exclusive radiotherapy (PFS: p = 0.04; OS: p = 0.01). Conclusions IGF-1R β overexpression and Hb level (≤11 g/dl) were associated with poor prognosis, and thus appear to be possible interesting biomarkers of radiation resistance. Our results corroborate previous pre-clinical studies suggesting IGF-1R and hypoxia to be part of the biological pathways leading to radio-resistance.
Collapse
Affiliation(s)
- P Moreno-Acosta
- Research Group in Radiobiology Clinical, Molecular and Cellular, National Cancer Institute, Bogotá, Colombia.,Research Group in Cancer Biology, National Cancer Institute, Bogotá, Colombia
| | - A Vallard
- Department of Radiation Oncology, Institut de cancérologie de la Loire-Lucien Neuwirth, 108 bis, Avenue Albert Raimond, BP 60008, 42271, Saint-Priest en Jarez, France.
| | - S Carrillo
- Research Group in Radiobiology Clinical, Molecular and Cellular, National Cancer Institute, Bogotá, Colombia
| | - O Gamboa
- Research Group in Cancer Biology, National Cancer Institute, Bogotá, Colombia.,Unit of Analysis, National Cancer Institute, Bogotá, Colombia
| | - A Romero-Rojas
- Group of Pathology Oncology, National Cancer Institute, Bogotá, Colombia
| | - M Molano
- Microbiology and Infection Diseases, The Royal Women's Hospital, Melbourne, Australia
| | - J Acosta
- Pathology Group, National University of Colombia, Bogotá, Colombia
| | - D Mayorga
- Research Group in Cancer Biology, National Cancer Institute, Bogotá, Colombia
| | - C Rancoule
- Department of Radiation Oncology, Institut de cancérologie de la Loire-Lucien Neuwirth, 108 bis, Avenue Albert Raimond, BP 60008, 42271, Saint-Priest en Jarez, France
| | - M A Garcia
- Department of Radiation Oncology, Institut de cancérologie de la Loire-Lucien Neuwirth, 108 bis, Avenue Albert Raimond, BP 60008, 42271, Saint-Priest en Jarez, France
| | - M Cotes Mestre
- Research Group in Cancer Biology, National Cancer Institute, Bogotá, Colombia.,Group of Radiotherapy and Medical Physical, National Cancer Institute, Bogotá, Colombia
| | - N Magné
- Department of Radiation Oncology, Institut de cancérologie de la Loire-Lucien Neuwirth, 108 bis, Avenue Albert Raimond, BP 60008, 42271, Saint-Priest en Jarez, France
| |
Collapse
|
50
|
|