1
|
Geirnaert F, Kerkhove L, Rifi A, Everaert T, Sanders J, Coppens J, Vandenplas H, Corbet C, Gevaert T, Dufait I, De Ridder M. Revisiting hydrogen peroxide as radiosensitizer for solid tumor cells. Radiother Oncol 2024; 203:110692. [PMID: 39716590 DOI: 10.1016/j.radonc.2024.110692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/01/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND AND PURPOSE Tumor hypoxia is the principal cause of clinical radioresistance. Despite its established role as radiosensitizer, hydrogen peroxide (H2O2) encounters clinical limitations due to stability and toxicity concerns. Recent advancements in drug delivery combine H2O2 with sodium hyaluronate (SH), enabling intratumoral administration of H2O2. This study investigates the radiomodulatory pathways of Kochi Oxydol-Radiation for Unresectable Carcinomas (KORTUC) (H2O2 + SH) under hypoxia. MATERIALS AND METHODS CT26 and 4T1 tumor cells were exposed to H2O2, SH and KORTUC under hypoxic conditions. Toxicity levels were determined using MTT and live-cell analysis. KORTUC's radiomodulatory properties were evaluated by colony formation assay and in spheroids. Reactive oxygen species (ROS) levels, DNA damage, apoptosis and ferroptosis were analyzed using flow cytometry. Oxygen consumption rate (OCR) and mitochondrial complex activity were assessed by Seahorse Analyzer. Oxygen levels were investigated using fiber-optic sensors. The in vitro findings were validated in CT26-bearing mice. RESULTS KORTUC demonstrated less cytotoxicity than H2O2-alone. KORTUC radiosensitized hypoxic tumor cells in a dose-dependent manner with enhancement ratios of 3.1 (CT26) and 2.7 (4T1). Dose-dependent OCR reduction following KORTUC exposure correlated with complex I and II inhibition, accompanied by mitochondrial ROS elevation. KORTUC injection into a 2D hypoxic tumor model surged O2 levels. KORTUC radiosensitized CT26-tumors, delaying growth by 14 days. CONCLUSIONS SH in KORTUC mitigates H2O2 cytotoxicity. We demonstrate that KORTUC overcomes hypoxia-induced radioresistance through inhibition of OCR, via complex I- and II-blockade, leading to tumor reoxygenation. Understanding KORTUC's pathways is essential for developing effective cancer combination therapies.
Collapse
Affiliation(s)
- F Geirnaert
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - L Kerkhove
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - A Rifi
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - T Everaert
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - J Sanders
- Department of Chemical and Physical Health Risks, Sciensano, 1050 Brussels, Belgium; Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - J Coppens
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - H Vandenplas
- Department of Medical Oncology, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - C Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, 1200 Brussels, Belgium
| | - T Gevaert
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - I Dufait
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - M De Ridder
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| |
Collapse
|
2
|
Abou-Foul AK, Dretzke J, Albon E, Kristunas C, Moore DJ, Karwath A, Gkoutos G, Mehanna H, Nankivell P. Clinical predictive models for recurrence and survival in treated laryngeal and hypopharyngeal cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1478385. [PMID: 39711957 PMCID: PMC11659268 DOI: 10.3389/fonc.2024.1478385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/06/2024] [Indexed: 12/24/2024] Open
Abstract
Background The limitations of the traditional TNM system have spurred interest in multivariable models for personalized prognostication in laryngeal and hypopharyngeal cancers (LSCC/HPSCC). However, the performance of these models depends on the quality of data and modelling methodology, affecting their potential for clinical adoption. This systematic review and meta-analysis (SR-MA) evaluated clinical predictive models (CPMs) for recurrence and survival in treated LSCC/HPSCC. We assessed models' characteristics and methodologies, as well as performance, risk of bias (RoB), and applicability. Methods Literature searches were conducted in MEDLINE (OVID), Embase (OVID) and IEEE databases from January 2005 to November 2023. The search algorithm used comprehensive text word and index term combinations without language or publication type restrictions. Independent reviewers screened titles and abstracts using a predefined Population, Index, Comparator, Outcomes, Timing and Setting (PICOTS) framework. We included externally validated (EV) multivariable models, with at least one clinical predictor, that provided recurrence or survival predictions. The SR-MA followed PRISMA reporting guidelines, and PROBAST framework for RoB assessment. Model discrimination was assessed using C-index/AUC, and was presented for all models using forest plots. MA was only performed for models that were externally validated in two or more cohorts, using random-effects model. The main outcomes were model discrimination and calibration measures for survival (OS) and/or local recurrence (LR) prediction. All measures and assessments were preplanned prior to data collection. Results The SR-MA identified 11 models, reported in 16 studies. Seven models for OS showed good discrimination on development, with only one excelling (C-index >0.9), and three had weak or poor discrimination. Inclusion of a radiomics score as a model parameter achieved relatively better performance. Most models had poor generalisability, demonstrated by worse discrimination performance on EV, but they still outperformed the TNM system. Only two models met the criteria for MA, with pooled EV AUCs 0.73 (95% CI 0.71-0.76) and 0.67 (95% CI 0.6-0.74). RoB was high for all models, particularly in the analysis domain. Conclusions This review highlighted the shortcomings of currently available models, while emphasizing the need for rigorous independent evaluations. Despite the proliferation of models, most exhibited methodological limitations and bias. Currently, no models can confidently be recommended for routine clinical use. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021248762, identifier CRD42021248762.
Collapse
Affiliation(s)
- Ahmad K. Abou-Foul
- Institute for Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
- Department of Cancer and Genomic Sciences & Centre for Health Data Science, University of Birmingham, Birmingham, United Kingdom
| | - Janine Dretzke
- Department of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Esther Albon
- Department of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Caroline Kristunas
- Institute for Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
| | - David J. Moore
- Department of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Andreas Karwath
- Department of Cancer and Genomic Sciences & Centre for Health Data Science, University of Birmingham, Birmingham, United Kingdom
| | - Georgios Gkoutos
- Department of Cancer and Genomic Sciences & Centre for Health Data Science, University of Birmingham, Birmingham, United Kingdom
| | - Hisham Mehanna
- Institute for Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
| | - Paul Nankivell
- Institute for Head and Neck Studies and Education, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
3
|
Duan H, Ren J, Wei S, Yang Z, Li C, Wang Z, Li M, Wei Z, Liu Y, Wang X, Lan H, Zeng Z, Xie M, Xie Y, Wu S, Hu W, Guo C, Zhang X, Liang L, Yu C, Mou Y, Jiang Y, Li H, Sugarman E, Deek RA, Chen Z, Li T, Chen Y, Yao M, Chen L, Liu L, Zhang G, Mou Y. Integrated analyses of multi-omic data derived from paired primary lung cancer and brain metastasis reveal the metabolic vulnerability as a novel therapeutic target. Genome Med 2024; 16:138. [PMID: 39593114 PMCID: PMC11590298 DOI: 10.1186/s13073-024-01410-8] [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: 09/17/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Lung cancer brain metastases (LC-BrMs) are frequently associated with dismal mortality rates in patients with lung cancer; however, standard of care therapies for LC-BrMs are still limited in their efficacy. A deep understanding of molecular mechanisms and tumor microenvironment of LC-BrMs will provide us with new insights into developing novel therapeutics for treating patients with LC-BrMs. METHODS Here, we performed integrated analyses of genomic, transcriptomic, proteomic, metabolomic, and single-cell RNA sequencing data which were derived from a total number of 154 patients with paired and unpaired primary lung cancer and LC-BrM, spanning four published and two newly generated patient cohorts on both bulk and single cell levels. RESULTS We uncovered that LC-BrMs exhibited a significantly greater intra-tumor heterogeneity. We also observed that mutations in a subset of genes were almost always shared by both primary lung cancers and LC-BrM lesions, including TTN, TP53, MUC16, LRP1B, RYR2, and EGFR. In addition, the genome-wide landscape of somatic copy number alterations was similar between primary lung cancers and LC-BrM lesions. Nevertheless, several regions of focal amplification were significantly enriched in LC-BrMs, including 5p15.33 and 20q13.33. Intriguingly, integrated analyses of transcriptomic, proteomic, and metabolomic data revealed mitochondrial-specific metabolism was activated but tumor immune microenvironment was suppressed in LC-BrMs. Subsequently, we validated our results by conducting real-time quantitative reverse transcription PCR experiments, immunohistochemistry, and multiplexed immunofluorescence staining of patients' paired tumor specimens. Therapeutically, targeting oxidative phosphorylation with gamitrinib in patient-derived organoids of LC-BrMs induced apoptosis and inhibited cell proliferation. The combination of gamitrinib plus anti-PD-1 immunotherapy significantly improved survival of mice bearing LC-BrMs. Patients with a higher expression of mitochondrial metabolism genes but a lower expression of immune genes in their LC-BrM lesions tended to have a worse survival outcome. CONCLUSIONS In conclusion, our findings not only provide comprehensive and integrated perspectives of molecular underpinnings of LC-BrMs but also contribute to the development of a potential, rationale-based combinatorial therapeutic strategy with the goal of translating it into clinical trials for patients with LC-BrMs.
Collapse
Affiliation(s)
- Hao Duan
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jianlan Ren
- Department of Computer Science, Ying Wu College of Computing, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Shiyou Wei
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Yang
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Li
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenning Wang
- Department of Neurosurgery, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Meichen Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zhi Wei
- Department of Computer Science, Ying Wu College of Computing, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
| | - Yu Liu
- Faculty of Dentistry, Prince Philip Dental Hospital, the University of Hong Kong, Sai Ying Pun, Hong Kong, China
| | - Xiuqi Wang
- Faculty of Dentistry, Prince Philip Dental Hospital, the University of Hong Kong, Sai Ying Pun, Hong Kong, China
| | - Hongbin Lan
- Faculty of Dentistry, Prince Philip Dental Hospital, the University of Hong Kong, Sai Ying Pun, Hong Kong, China
| | - Zhen Zeng
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Maodi Xie
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuan Xie
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Suwen Wu
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Wanming Hu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Chengcheng Guo
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiangheng Zhang
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Lun Liang
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Chengwei Yu
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou, 510530, China
| | - Yanhao Mou
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yu Jiang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Houde Li
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Eric Sugarman
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, 19131, USA
| | - Rebecca A Deek
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zexin Chen
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510535, China
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaohui Chen
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Maojin Yao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, China.
| | - Likun Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Lunxu Liu
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Gao Zhang
- Faculty of Dentistry, Prince Philip Dental Hospital, the University of Hong Kong, Sai Ying Pun, Hong Kong, China.
| | - Yonggao Mou
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
4
|
Rey-Keim S, Schito L. Origins and molecular effects of hypoxia in cancer. Semin Cancer Biol 2024; 106-107:166-178. [PMID: 39427969 DOI: 10.1016/j.semcancer.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Hypoxia (insufficient O2) is a pivotal factor in cancer progression, triggering genetic, transcriptional, translational and epigenetic adaptations associated to therapy resistance, metastasis and patient mortality. In this review, we outline the microenvironmental origins and molecular mechanisms responsible for hypoxic cancer cell adaptations in situ and in vitro, whilst outlining current approaches to stratify, quantify and therapeutically target hypoxia in the context of precision oncology.
Collapse
Affiliation(s)
- Sergio Rey-Keim
- UCD School of Medicine, University College Dublin, Belfield, Dublin D04 C7X2, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin D04 C7X2, Ireland.
| | - Luana Schito
- UCD School of Medicine, University College Dublin, Belfield, Dublin D04 C7X2, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin D04 C7X2, Ireland.
| |
Collapse
|
5
|
Elmali A, Guler OC, Demirhan B, Yavuz M, Onal C. Long-term analysis of hematological parameters as predictors of recurrence patterns and treatment outcomes in cervical cancer patients undergoing definitive chemoradiotherapy. Strahlenther Onkol 2024; 200:949-957. [PMID: 39102040 PMCID: PMC11527943 DOI: 10.1007/s00066-024-02278-8] [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: 03/29/2024] [Accepted: 07/07/2024] [Indexed: 08/06/2024]
Abstract
PURPOSE This study sought to determine the predictive and prognostic value of clinicopathological parameters and neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and hemoglobin (Hgb) level in predicting recurrence patterns and locoregional relapse-free survival (LRFS) and distant metastasis-free survival (DMFS) in cervical cancer patients receiving definitive chemoradiotherapy (ChRT). METHODS This study included 261 cervical cancer patients treated with ChRT. The primary endpoints were the predictors of local recurrence (LR) and distant metastasis (DM), whereas the secondary endpoints were LRFS and DMFS. The association of survival with potential prognostic factors was analyzed using Cox regression analysis, and the predictors of LR and DM were identified using logistic regression analysis. RESULTS The median follow-up time was 10.9 years. Recurrences occurred in 132 patients (50.6%) within a median of 11.2 months after definitive ChRT. NLR and PLR values were significantly higher in patients with LR and DM than in those without, with no significant differences in Hgb levels in patients with or without LR and DM. In the multivariable logistic regression analysis, lymph node metastasis, elevated NLR, and low Hgb level were significantly correlated with LR and DM. In the multivariable analysis, large tumor size, presence of lymph node metastasis, and elevated NLR were the independent predictors for poor LRFS and DMFS, and Hgb level was an additional prognostic factor for DMFS. CONCLUSION Hematological markers, particularly NLR and Hgb, may serve as cost-effective and readily accessible indicators for predicting recurrence and survival in cervical cancer patients, contributing to their practical use in routine assessments.
Collapse
Affiliation(s)
- Aysenur Elmali
- Faculty of Medicine, Department of Radiation Oncology, Baskent University, Ankara, Turkey
| | - Ozan Cem Guler
- Faculty of Medicine, Adana Dr. Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Baskent University, 01120, Adana, Turkey
| | - Birhan Demirhan
- Division of Radiation Oncology, Iskenderun Gelisim Hospital, Hatay, Turkey
| | - Melek Yavuz
- Faculty of Medicine, Adana Dr. Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Baskent University, 01120, Adana, Turkey
| | - Cem Onal
- Faculty of Medicine, Department of Radiation Oncology, Baskent University, Ankara, Turkey.
- Faculty of Medicine, Adana Dr. Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Baskent University, 01120, Adana, Turkey.
- Division of Radiation Oncology, Iskenderun Gelisim Hospital, Hatay, Turkey.
| |
Collapse
|
6
|
Ripoll-Viladomiu I, Prina-Mello A, Movia D, Marignol L. Extracellular vesicles and the "six Rs" in radiotherapy. Cancer Treat Rev 2024; 129:102799. [PMID: 38970839 DOI: 10.1016/j.ctrv.2024.102799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Over half of patients with cancer receive radiation therapy during the course of their disease. Decades of radiobiological research have identified 6 parameters affecting the biological response to radiation referred to as the 6 "Rs": Repair, Radiosensitivity, Repopulation, Redistribution, Reoxygenation, and Reactivation of the anti-tumour immune response. Extracellular Vesicles (EVs) are small membrane-bound particles whose multiple biological functions are increasingly documented. Here we discuss the evidence for a role of EVs in the orchestration of the response of cancer cells to radiotherapy. We highlight that EVs are involved in DNA repair mechanisms, modulation of cellular sensitivity to radiation, and facilitation of tumour repopulation. Moreover, EVs influence tumour reoxygenation dynamics, and play a pivotal role in fostering radioresistance. Last, we examine how EV-related strategies could be translated into novel strategies aimed at enhancing the efficacy of radiation therapy against cancer.
Collapse
Affiliation(s)
- Isabel Ripoll-Viladomiu
- Trinity St. James's Cancer Institute, Radiobiology and Molecular Oncology Research Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Ireland; Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Dania Movia
- Trinity St. James's Cancer Institute, Radiobiology and Molecular Oncology Research Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Ireland; Department of Biology and Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Callan Building, Maynooth, Ireland
| | - Laure Marignol
- Trinity St. James's Cancer Institute, Radiobiology and Molecular Oncology Research Group, Applied Radiation Therapy Trinity, Discipline of Radiation Therapy, Trinity College Dublin, Ireland.
| |
Collapse
|
7
|
Petusseau AF, Ochoa M, Reed M, Doyley MM, Hasan T, Bruza P, Pogue BW. Pressure-enhanced sensing of tissue oxygenation via endogenous porphyrin: Implications for dynamic visualization of cancer in surgery. Proc Natl Acad Sci U S A 2024; 121:e2405628121. [PMID: 39141355 PMCID: PMC11348300 DOI: 10.1073/pnas.2405628121] [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: 03/18/2024] [Accepted: 06/21/2024] [Indexed: 08/15/2024] Open
Abstract
Fluorescence guidance is routinely used in surgery to enhance perfusion contrast in multiple types of diseases. Pressure-enhanced sensing of tissue oxygenation (PRESTO) via fluorescence is a technique extensively analyzed here, that uses an FDA-approved human precursor molecule, 5-aminolevulinic acid (ALA), to stimulate a unique delayed fluorescence signal that is representative of tissue hypoxia. The ALA precontrast agent is metabolized in most tissues into a red fluorescent molecule, protoporphyrin IX (PpIX), which has both prompt fluorescence, indicative of the concentration, and a delayed fluorescence, that is amplified in low tissue oxygen situations. Applied pressure from palpation induces transient capillary stasis and a resulting transient PRESTO contrast, dominant when there is near hypoxia. This study examined the kinetics and behavior of this effect in both normal and tumor tissues, with a prolonged high PRESTO contrast (contrast to background of 7.3) across 5 tumor models, due to sluggish capillaries and inhibited vasodynamics. This tissue function imaging approach is a fundamentally unique tool for real-time palpation-induced tissue response in vivo, relevant for chronic hypoxia, such as vascular diseases or oncologic surgery.
Collapse
Affiliation(s)
| | - Marien Ochoa
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI53705
| | - Matthew Reed
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI53705
| | - Marvin M. Doyley
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY14627
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Petr Bruza
- Thayer School of Engineering, Dartmouth College, Hanover, NH03755
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH03755
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI53705
| |
Collapse
|
8
|
Sambasivan K, Tyrrell WE, Farooq R, Mynerich J, Edwards RS, Tanc M, Urbano TG, Witney TH. [ 18F]FSPG-PET provides an early marker of radiotherapy response in head and neck squamous cell cancer. NPJ IMAGING 2024; 2:28. [PMID: 39132311 PMCID: PMC11315666 DOI: 10.1038/s44303-024-00038-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/31/2024] [Indexed: 08/13/2024]
Abstract
The ability to image early treatment response to radiotherapy in head and neck squamous cell carcinoma (HNSCC) will enable the identification of radioresistant tumor volumes suitable for treatment intensification. Here, we propose the system xc - radiotracer (4S)-4-(3-[18F]fluoropropyl)-L-glutamate ([18F]FSPG) as a non-invasive method to monitor radiation response in HNSCC. We assessed temporal changes in cell death, antioxidant status, and [18F]FSPG retention following a single dose of 10 Gy irradiation in FaDU HNSCC cells. Next, using a fractionated course of radiotherapy, we assessed tumor volume changes and performed [18F]FSPG-PET imaging in FaDU-bearing mouse xenografts, followed by ex vivo response assessment. In cells, 10 Gy irradiation reduced [18F]FSPG retention, coinciding with the induction of apoptosis and the production of reactive oxygen species. In vivo, [18F]FSPG tumor retention was halved seven days after the start of treatment, which preceded radiotherapy-induced tumor shrinkage, thereby confirming [18F]FSPG-PET as an early and sensitive marker of radiation response.
Collapse
Affiliation(s)
- Khrishanthne Sambasivan
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
- Department of Clinical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Will E. Tyrrell
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Rizwan Farooq
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Jenasee Mynerich
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Richard S. Edwards
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Muhammet Tanc
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Teresa Guerrero Urbano
- Department of Clinical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Faculty of Dentistry, Oral & Craniofacial Sciences and School of Cancer & Pharmaceutical Sciences, King’s College London, London, UK
| | - Timothy H. Witney
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| |
Collapse
|
9
|
Morse RT, Mell LK. Hypoxic Cell Radiosensitization in Head and Neck Squamous Cell Carcinoma: Running Out of Air. Int J Radiat Oncol Biol Phys 2024; 119:783-785. [PMID: 38851276 DOI: 10.1016/j.ijrobp.2024.02.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 06/10/2024]
Affiliation(s)
- Ryan T Morse
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Loren K Mell
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California.
| |
Collapse
|
10
|
Thomson DJ, Slevin NJ, Baines H, Betts G, Bolton S, Evans M, Garcez K, Irlam J, Lee L, Melillo N, Mistry H, More E, Nutting C, Price JM, Schipani S, Sen M, Yang H, West CM. Randomized Phase 3 Trial of the Hypoxia Modifier Nimorazole Added to Radiation Therapy With Benefit Assessed in Hypoxic Head and Neck Cancers Determined Using a Gene Signature (NIMRAD). Int J Radiat Oncol Biol Phys 2024; 119:771-782. [PMID: 38072326 DOI: 10.1016/j.ijrobp.2023.11.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 01/27/2024]
Abstract
PURPOSE Tumor hypoxia is an adverse prognostic factor in head and neck squamous cell carcinoma (HNSCC). We assessed whether patients with hypoxic HNSCC benefited from the addition of nimorazole to definitive intensity modulated radiation therapy (IMRT). METHODS AND MATERIALS NIMRAD was a phase 3, multicenter, placebo-controlled, double-anonymized trial of patients with HNSCC unsuitable for concurrent platinum chemotherapy or cetuximab with definitive IMRT (NCT01950689). Patients were randomized 1:1 to receive IMRT (65 Gy in 30 fractions over 6 weeks) plus nimorazole (1.2 g/m2 daily, before IMRT) or placebo. The primary endpoint was freedom from locoregional progression (FFLRP) in patients with hypoxic tumors, defined as greater than or equal to the median tumor hypoxia score of the first 50 patients analyzed (≥0.079), using a validated 26-gene signature. The planned sample size was 340 patients, allowing for signature generation in 85% and an assumed hazard ratio (HR) of 0.50 for nimorazole effectiveness in the hypoxic group and requiring 66 locoregional failures to have 80% power in a 2-tail log-rank test at the 5% significance level. RESULTS Three hundred thirty-eight patients were randomized by 19 centers in the United Kingdom from May 2014 to May 2019, with a median follow-up of 3.1 years (95% CI, 2.9-3.4). Hypoxia scores were available for 286 (85%). The median patient age was 73 years (range, 44-88; IQR, 70-76). There were 36 (25.9%) locoregional failures in the hypoxic group, in which nimorazole + IMRT did not improve FFLRP (adjusted HR, 0.72; 95% CI, 0.36-1.44; P = .35) or overall survival (adjusted HR, 0.96; 95% CI, 0.53-1.72; P = .88) compared with placebo + IMRT. Similarly, nimorazole + IMRT did not improve FFLRP or overall survival in the whole population. In total (N = 338), 73% of patients allocated nimorazole adhered to the drug for ≥50% of IMRT fractions. Nimorazole + IMRT caused more acute nausea compared with placebo + IMRT (Common Terminology Criteria for Adverse Events version 4.0 G1+2: 56.6% vs 42.4%, G3: 10.1% vs 5.3%, respectively; P < .05). CONCLUSIONS Addition of the hypoxia modifier nimorazole to IMRT for locally advanced HNSCC in older and less fit patients did not improve locoregional control or survival.
Collapse
Affiliation(s)
- David J Thomson
- The Christie NHS Foundation Trust, Manchester, United Kingdom; University of Liverpool, Liverpool, United Kingdom; Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Nick J Slevin
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Helen Baines
- National Radiotherapy Trials Quality Assurance (RTTQA) Group, Northwood, United Kingdom; Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Guy Betts
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Steve Bolton
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Mererid Evans
- Cardiff University and Velindre Cancer Centre, Cardiff, United Kingdom
| | - Kate Garcez
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Joely Irlam
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Lip Lee
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | - Hitesh Mistry
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom; SystemsForecastingUK Ltd, Lancaster, United Kingdom
| | - Elisabet More
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | | | - James M Price
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Stefano Schipani
- Beatson West of Scotland Cancer Centre and University of Glasgow, Glasgow, United Kingdom
| | - Mehmet Sen
- Leeds Teaching Hospital NHS Trust, Leeds, United Kingdom
| | - Huiqi Yang
- National Radiotherapy Trials Quality Assurance (RTTQA) Group, Northwood, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Catharine M West
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom.
| |
Collapse
|
11
|
Li CX, Gong ZC, Zhang WN, Zhang Y, Zhao HR. Radioresistance or/and radiosensitivity of head and neck squamous cell carcinoma: biological angle. Oral Maxillofac Surg 2024; 28:547-555. [PMID: 37935817 DOI: 10.1007/s10006-023-01189-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/29/2023] [Indexed: 11/09/2023]
Abstract
OBJECTIVE This narrative review aimed to compile and summarize clinically relevant literature in radiation therapy and to discuss the potential in radioresistant and radiosensitive head and neck squamous cell carcinoma (HNSCC). METHODS AND MATERIALS Google Scholar, PubMed, and the Cochrane Library were retrieved using combined key words such as "radiotherapy" and "head and neck cancer." Search strings additionally queried were "radioresistant," "radiosensitive," "head and neck region," "squamous cell carcinoma," in combination with Boolean operators 'AND' and 'OR.' Subsequently, the resulting publications were included for review of the full text. RESULTS Radiotherapeutic responses currently in clinical observation referred to HNSCC scoping were selected into this review. The compiled mechanisms were then detailed concerning on the clinical significance, biological characteristics, and molecular function. CONCLUSIONS Brachytherapy or/and external-beam radiotherapy are crucial for treating HNSCC especially the early stage patients, but in some patients with locally advanced tumors, their outcome with radiation therapy is poor due to obvious radioresistance. The curative effects mainly depend on the response to radiation therapy so an updated review is needed to optimize further applications in HNSCC radiotherapy.
Collapse
Affiliation(s)
- Chen-Xi Li
- Department of Oral and Maxillofacial Oncology & Surgery, School / Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, Urumqi, 830054, China.
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhong-Cheng Gong
- Department of Oral and Maxillofacial Oncology & Surgery, School / Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, Urumqi, 830054, China.
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China.
| | - Wei-Na Zhang
- Ear, Nose & Throat Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yang Zhang
- The First Ward of Oncological Department, Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Hua-Rong Zhao
- The First Ward of Oncological Department, Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| |
Collapse
|
12
|
Epel B, Viswakarma N, Sundramoorthy SV, Pawar NJ, Kotecha M. Oxygen Imaging of a Rabbit Tumor Using a Human-Sized Pulse Electron Paramagnetic Resonance Imager. Mol Imaging Biol 2024; 26:403-410. [PMID: 37715089 DOI: 10.1007/s11307-023-01852-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE Spatial heterogeneity in tumor hypoxia is one of the most important factors regulating tumor growth, development, aggressiveness, metastasis, and affecting treatment outcome. Most solid tumors are known to have hypoxia or low oxygen levels (pO2 ≤10 torr). Electron paramagnetic resonance oxygen imaging (EPROI) is an emerging oxygen mapping technology. EPROI utilizes the linear relationship between the relaxation rates of the injectable OX071 trityl spin probe and the partial oxygen pressure (pO2). However, most of the EPROI studies have been limited to mouse models of solid tumors because of the instrument-size limitations. The purpose of this work was to develop a human-sized 9-mT (250 MHz resonance frequency, 60 cm bore size) pulse EPROI instrument and evaluate its performance with rabbit VX-2 tumor oxygen imaging. METHODS A New Zealand white rabbit with a 3.2-cm VX-2 tumor in the calf muscle was imaged using the human-sized EPROI instrument and a 2.25-in. ID volume coil. The animal received a ~8-min intravenous injection of OX071 (5.2 mL total volume at 72 mM concentration) and, after 75 min, an intratumoral injection (120 μL total at 5 mM OX071 concentration) and underwent EPROI. At the end of the experiments, MRI was performed using a preclinical 9.4-T MRI system to outline the tumor boundaries. RESULTS For the first time, a human-sized pulse EPROI instrument with a 60-cm bore size/250-MHz frequency was built and evaluated using rabbit tumor oxygen imaging. For the first time, the systemic IV injection of the oxygen-sensitive trityl OX071 spin probe was used for an animal of this size. The resulting EPROI image from the IV injection showed complete tumor coverage. The image obtained after intratumoral injection showed localized coverage in the upper lobe of the tumor, demonstrating the need for improved intratumoral injection protocol. CONCLUSIONS This study demonstrates the performance of the world's first human-sized pulse EPROI instrument. It also demonstrates that the EPROI of larger animals can be performed using the systemic injection of a manageable amount of the spin probe. This brings EPROI one step closer to clinical applications in cancer therapies. Oxygen imaging is a platform technology, and the instrument and techniques developed here will also be useful for other clinical applications.
Collapse
Affiliation(s)
- Boris Epel
- O2M Technologies, LLC, Chicago, IL, 60612, USA.
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, 60637, USA.
| | | | | | | | | |
Collapse
|
13
|
Kristensen MH, Sørensen MK, Tramm T, Alsner J, Sørensen BS, Maare C, Johansen J, Primdahl H, Bratland Å, Kristensen CA, Andersen M, Lilja-Fischer JK, Holm AIS, Samsøe E, Hansen CR, Zukauskaite R, Overgaard J, Eriksen JG. Tumor volume and cancer stem cell expression as prognostic markers for high-dose loco-regional failure in head and neck squamous cell carcinoma - A DAHANCA 19 study. Radiother Oncol 2024; 193:110149. [PMID: 38341096 DOI: 10.1016/j.radonc.2024.110149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND AND PURPOSE Reliable and accessible biomarkers for patients with Head and Neck Squamous Cell Carcinoma (HNSCC) are warranted for biologically driven radiotherapy (RT). This study aimed to investigate the prognostic value of putative cancer stem cell (CSC) markers, hypoxia, and tumor volume using loco-regional high-dose failure (HDF) as endpoint. MATERIALS AND METHODS Tumor tissue was retrieved from patients treated with primary chemo-(C-)RT and nimorazole for HNSCC in the Danish Head and Neck Cancer Study Group (DAHANCA) 19 study. Tumor volume, hypoxic classification, and expression of CSC markers CD44, SLC3A2, and MET were analyzed. For patients with eligible data on all parameters (n = 340), the risk of HDF following primary chemo-(C-)RT were analyzed by these biomarkers as a whole and stratified for p16-positive oropharynx (p16 + OPSCC) vs p16-negative (p16-) tumors (oral cavity, p16- oropharynx, hypopharynx and larynx). RESULTS Higher risk of HDF was seen for patients with larger primary and nodal volume (>25 cm3, Hazard Ratio (HR): 3.00 [95 % CI: 1.73-5.18]), high SLC3A2 (HR: 2.99 [1.28-6.99]), CD44 (>30 % positive, HR: 2.29 [1.05-5.00]), and p16- tumors (HR: 2.53 [1.05-6.11]). p16- tumors had a higher CSC marker expression than p16 + OPSCC. The factors associated with the highest risk of HDF were larger volume (HR: 3.29 [1.79-6.04]) for p16- tumors (n = 178) and high SLC3A2 (HR: 6.19 [1.58-24.23]) for p16 + OPSCC (n = 162). CONCLUSION Tumor volume, p16, and CSC markers are potential biomarkers for HDF for patients with HNSCC treated with (C-)RT. Lower expression of CSC in p16 + OPSCC may contribute to better tumor control.
Collapse
Affiliation(s)
| | - Mia Kristina Sørensen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Trine Tramm
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 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; Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jørgen Johansen
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Hanne Primdahl
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Åse Bratland
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | | | - Maria Andersen
- Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | - Jacob Kinggaard Lilja-Fischer
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Otolaryngology - Head & Neck Surgery, Aarhus University Hospital, Denmark
| | | | - Eva Samsøe
- Zealand University Hospital, Department of Oncology, Næstved, Denmark
| | - Christian Rønn Hansen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Odense University Hospital, Laboratory of Radiation Physics, Odense, Denmark; University of Southern Denmark, Department of Clinical Research, Odense, Denmark
| | - Ruta Zukauskaite
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper Grau Eriksen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
14
|
McCabe A, Martin S, Rowe S, Shah J, Morgan PS, Borys D, Panek R. Oxygen-enhanced MRI assessment of tumour hypoxia in head and neck cancer is feasible and well tolerated in the clinical setting. Eur Radiol Exp 2024; 8:27. [PMID: 38443722 PMCID: PMC10914657 DOI: 10.1186/s41747-024-00429-1] [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: 09/01/2023] [Accepted: 01/08/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Tumour hypoxia is a recognised cause of radiotherapy treatment resistance in head and neck squamous cell carcinoma (HNSCC). Current positron emission tomography-based hypoxia imaging techniques are not routinely available in many centres. We investigated if an alternative technique called oxygen-enhanced magnetic resonance imaging (OE-MRI) could be performed in HNSCC. METHODS A volumetric OE-MRI protocol for dynamic T1 relaxation time mapping was implemented on 1.5-T clinical scanners. Participants were scanned breathing room air and during high-flow oxygen administration. Oxygen-induced changes in T1 times (ΔT1) and R2* rates (ΔR2*) were measured in malignant tissue and healthy organs. Unequal variance t-test was used. Patients were surveyed on their experience of the OE-MRI protocol. RESULTS Fifteen patients with HNSCC (median age 59 years, range 38 to 76) and 10 non-HNSCC subjects (median age 46.5 years, range 32 to 62) were scanned; the OE-MRI acquisition took less than 10 min and was well tolerated. Fifteen histologically confirmed primary tumours and 41 malignant nodal masses were identified. Median (range) of ΔT1 times and hypoxic fraction estimates for primary tumours were -3.5% (-7.0 to -0.3%) and 30.7% (6.5 to 78.6%) respectively. Radiotherapy-responsive and radiotherapy-resistant primary tumours had mean estimated hypoxic fractions of 36.8% (95% confidence interval [CI] 17.4 to 56.2%) and 59.0% (95% CI 44.6 to 73.3%), respectively (p = 0.111). CONCLUSIONS We present a well-tolerated implementation of dynamic, volumetric OE-MRI of the head and neck region allowing discernment of differing oxygen responses within biopsy-confirmed HNSCC. TRIAL REGISTRATION ClinicalTrials.gov, NCT04724096 . Registered on 26 January 2021. RELEVANCE STATEMENT MRI of tumour hypoxia in head and neck cancer using routine clinical equipment is feasible and well tolerated and allows estimates of tumour hypoxic fractions in less than ten minutes. KEY POINTS • Oxygen-enhanced MRI (OE-MRI) can estimate tumour hypoxic fractions in ten-minute scanning. • OE-MRI may be incorporable into routine clinical tumour imaging. • OE-MRI has the potential to predict outcomes after radiotherapy treatment.
Collapse
Affiliation(s)
- Alastair McCabe
- Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK.
- Department of Clinical Oncology, Nottingham University Hospitals NHS Trust, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK.
| | - Stewart Martin
- Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Selene Rowe
- Department of Radiology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Jagrit Shah
- Department of Radiology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Paul S Morgan
- Mental Health & Clinical Neurosciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Medical Physics & Clinical Engineering, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Damian Borys
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Rafal Panek
- Mental Health & Clinical Neurosciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Medical Physics & Clinical Engineering, Nottingham University Hospitals NHS Trust, Nottingham, UK
| |
Collapse
|
15
|
Smith TAD, West CML, Joseph N, Lane B, Irlam-Jones J, More E, Mistry H, Reeves KJ, Song YP, Reardon M, Hoskin PJ, Hussain SA, Denley H, Hall E, Porta N, Huddart RA, James ND, Choudhury A. A hypoxia biomarker does not predict benefit from giving chemotherapy with radiotherapy in the BC2001 randomised controlled trial. EBioMedicine 2024; 101:105032. [PMID: 38387404 PMCID: PMC10897900 DOI: 10.1016/j.ebiom.2024.105032] [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: 06/19/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND BC2001 showed combining chemotherapy (5-FU + mitomycin-C) with radiotherapy improves loco-regional disease-free survival in patients with muscle-invasive bladder cancer (MIBC). We previously showed a 24-gene hypoxia-associated signature predicted benefit from hypoxia-modifying radiosensitisation in BCON and hypothesised that only patients with low hypoxia scores (HSs) would benefit from chemotherapy in BC2001. BC2001 allowed conventional (64Gy/32 fractions) or hypofractionated (55Gy/20 fractions) radiotherapy. An exploratory analysis tested an additional hypothesis that hypofractionation reduces reoxygenation and would be detrimental for patients with hypoxic tumours. METHODS RNA was extracted from pre-treatment biopsies (298 BC2001 patients), transcriptomic data generated (Affymetrix Clariom-S arrays), HSs calculated (median expression of 24-signature genes) and patients stratified as hypoxia-high or -low (cut-off: cohort median). PRIMARY ENDPOINT invasive loco-regional control (ILRC); secondary overall survival. FINDINGS Hypoxia affected overall survival (HR = 1.30; 95% CI 0.99-1.70; p = 0.062): more uncertainty for ILRC (HR = 1.29; 95% CI 0.82-2.03; p = 0.264). Benefit from chemotherapy was similar for patients with high or low HSs, with no interaction between HS and treatment arm. High HS associated with poor ILRC following hypofractionated (n = 90, HR 1.69; 95% CI 0.99-2.89 p = 0.057) but not conventional (n = 207, HR 0.70; 95% CI 0.28-1.80, p = 0.461) radiotherapy. The finding was confirmed in an independent cohort (BCON) where hypoxia associated with a poor prognosis for patients receiving hypofractionated (n = 51; HR 14.2; 95% CI 1.7-119; p = 0.015) but not conventional (n = 24, HR 1.04; 95% CI 0.07-15.5, p = 0.978) radiotherapy. INTERPRETATION Tumour hypoxia status does not affect benefit from BC2001 chemotherapy. Hypoxia appears to affect fractionation sensitivity. Use of HSs to personalise treatment needs testing in a biomarker-stratified trial. FUNDING Cancer Research UK, NIHR, MRC.
Collapse
Affiliation(s)
- Tim A D Smith
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK; Nuclear Futures Institute, School of Computer Science and Electronic Engineering, Bangor University, Bangor, UK
| | - Catharine M L West
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK.
| | - Nuradh Joseph
- Sri Lanka Cancer Research Group, Maharagama, Sri Lanka
| | - Brian Lane
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Joely Irlam-Jones
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Elisabet More
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Hitesh Mistry
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Kimberley J Reeves
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Yee Pei Song
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Mark Reardon
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Peter J Hoskin
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK; Mount Vernon Cancer Centre, Northwood, London, UK
| | - Syed A Hussain
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Helen Denley
- Pathology Centre, Shrewsbury and Telford NHS Trust, Royal Shrewsbury Hospital, Shrewsbury, UK
| | - Emma Hall
- Institute of Cancer Research, Clinical Trials & Statistics Unit, London, UK
| | - Nuria Porta
- Institute of Cancer Research, Clinical Trials & Statistics Unit, London, UK
| | - Robert A Huddart
- Royal Marsden NHS Trust, Department of Oncology, Downs Road, Sutton, Surrey, England, UK
| | - Nick D James
- Royal Marsden NHS Trust, Department of Oncology, Downs Road, Sutton, Surrey, England, UK
| | - Ananya Choudhury
- Translational Radiobiology Group, Division of Cancer Sciences, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
16
|
Li CX, Tan XR, Wei W, Li MQ, Zhang WN, Gong ZC, Zhang Y, Zhao HR. A radiobiological perspective on radioresistance or/and radiosensitivity of head and neck squamous cell carcinoma. Rep Pract Oncol Radiother 2024; 28:809-822. [PMID: 38515813 PMCID: PMC10954264 DOI: 10.5603/rpor.99355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/11/2023] [Indexed: 03/23/2024] Open
Abstract
Background This article aimed to compile and summarize clinically relevant literature in radiation therapy, and to discuss the potential in radioresistant and radiosensitive head and neck cancer. Study Design Narrative review. Materials and methods Google Scholar, PubMed and the Cochrane Library were retrieved using combined key words such as "radiotherapy" and "head and neck cancer". Search strings additionally queried were "radioresistant", "radiosensitive", "head and neck region", "squamous cell carcinoma", in combination with Boolean Operators 'AND' and 'OR'. Subsequently, the resulting publications were included for review of the full text. Results Radiotherapeutic response currently in clinical observation referred to HNSCC scoping were selected into this review. The compiled mechanisms were then detailed concerning on the clinical significance, biological characteristics, and molecular function. Conclusions Brachytherapy or/and external-beam radiotherapy are crucial for treating HNSCC, especially the early stage patients, but in patients with locally advanced tumors, their outcome with radiation therapy is poor due to obvious radioresistance. The curative effects mainly depend on the response of radiation therapy, so an updated review is needed to optimize further applications in HNSCC radiotherapy.
Collapse
Affiliation(s)
- Chen-xi Li
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-rong Tan
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wei Wei
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Mu-qiu Li
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wei-na Zhang
- Ear, Nose & Throat Department, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhong-cheng Gong
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yang Zhang
- The First Ward of Oncological Department, Cancer Center, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hua-rong Zhao
- The First Ward of Oncological Department, Cancer Center, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
17
|
Morozas A, Malyško-Ptašinskė V, Kulbacka J, Ivaška J, Ivaškienė T, Novickij V. Electrochemotherapy for head and neck cancers: possibilities and limitations. Front Oncol 2024; 14:1353800. [PMID: 38434679 PMCID: PMC10905418 DOI: 10.3389/fonc.2024.1353800] [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: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024] Open
Abstract
Head and neck cancer continues to be among the most prevalent types of cancer globally, yet it can be managed with appropriate treatment approaches. Presently, chemotherapy and radiotherapy stand as the primary treatment modalities for various groups and regions affected by head and neck cancer. Nonetheless, these treatments are linked to adverse side effects in patients. Moreover, due to tumor resistance to multiple drugs (both intrinsic and extrinsic) and radiotherapy, along with numerous other factors, recurrences or metastases often occur. Electrochemotherapy (ECT) emerges as a clinically proven alternative that offers high efficacy, localized effect, and diminished negative factors. Electrochemotherapy involves the treatment of solid tumors by combining a non-permeable cytotoxic drug, such as bleomycin, with a locally administered pulsed electric field (PEF). It is crucial to employ this method effectively by utilizing optimal PEF protocols and drugs at concentrations that do not possess inherent cytotoxic properties. This review emphasizes an examination of diverse clinical practices of ECT concerning head and neck cancer. It specifically delves into the treatment procedure, the choice of anti-cancer drugs, pre-treatment planning, PEF protocols, and electroporation electrodes as well as the efficacy of tumor response to the treatment and encountered obstacles. We have also highlighted the significance of assessing the spatial electric field distribution in both tumor and adjacent tissues prior to treatment as it plays a pivotal role in determining treatment success. Finally, we compare the ECT methodology to conventional treatments to highlight the potential for improvement and to facilitate popularization of the technique in the area of head and neck cancers where it is not widespread yet while it is not the case with other cancer types.
Collapse
Affiliation(s)
- Arnoldas Morozas
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre of Innovative Medicine, Vilnius, Lithuania
| | | | - Julita Kulbacka
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre of Innovative Medicine, Vilnius, Lithuania
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Wroclaw, Poland
| | - Justinas Ivaška
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre of Innovative Medicine, Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Tatjana Ivaškienė
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre of Innovative Medicine, Vilnius, Lithuania
| | - Vitalij Novickij
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre of Innovative Medicine, Vilnius, Lithuania
- Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| |
Collapse
|
18
|
Panahi Meymandi AR, Akbari B, Soltantoyeh T, Shahosseini Z, Hosseini M, Hadjati J, Mirzaei HR. PX-478, an HIF-1α inhibitor, impairs mesoCAR T cell antitumor function in cervical cancer. Front Oncol 2024; 14:1357801. [PMID: 38425341 PMCID: PMC10903365 DOI: 10.3389/fonc.2024.1357801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Chimeric Antigen Receptor (CAR) T cell therapy has demonstrated remarkable success in treating hematological malignancies. However, its efficacy against solid tumors, including cervical cancer, remains a challenge. Hypoxia, a common feature of the tumor microenvironment, profoundly impacts CAR T cell function, emphasizing the need to explore strategies targeting hypoxia-inducible factor-1α (HIF-1α). Methods In this study, we evaluated the effects of the HIF-1α inhibitor PX-478 on mesoCAR T cell function through in-silico and in vitro experiments. We conducted comprehensive analyses of HIF-1α expression in cervical cancer patients and examined the impact of PX-478 on T cell proliferation, cytokine production, cytotoxicity, and exhaustion markers. Results Our in-silico analyses revealed high expression of HIF-1α in cervical cancer patients, correlating with poor prognosis. PX-478 effectively reduced HIF-1α levels in T and HeLa cells. While PX-478 exhibited dose-dependent inhibition of antigen-nonspecific T and mesoCAR T cell proliferation, it had minimal impact on antigen-specific mesoCAR T cell proliferation. Notably, PX-478 significantly impaired the cytotoxic function of mesoCAR T cells and induced terminally exhausted T cells. Discussion Our results underscore the significant potential and physiological relevance of the HIF-1α pathway in determining the fate and function of both T and CAR T cells. However, we recognize the imperative for further molecular investigations aimed at unraveling the intricate downstream targets associated with HIF-1α and its influence on antitumor immunity, particularly within the context of hypoxic tumors. These insights serve as a foundation for the careful development of combination therapies tailored to counter immunosuppressive pathways within hypoxic environments and fine-tune CAR T cell performance in the intricate tumor microenvironment.
Collapse
Affiliation(s)
- Ahmad Reza Panahi Meymandi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnia Akbari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahereh Soltantoyeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Shahosseini
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
- Virology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mina Hosseini
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Chang B, Chen J, Bao J, Sun T, Cheng Z. Molecularly Engineered Room-Temperature Phosphorescence for Biomedical Application: From the Visible toward Second Near-Infrared Window. Chem Rev 2023; 123:13966-14037. [PMID: 37991875 DOI: 10.1021/acs.chemrev.3c00401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Phosphorescence, characterized by luminescent lifetimes significantly longer than that of biological autofluorescence under ambient environment, is of great value for biomedical applications. Academic evidence of fluorescence imaging indicates that virtually all imaging metrics (sensitivity, resolution, and penetration depths) are improved when progressing into longer wavelength regions, especially the recently reported second near-infrared (NIR-II, 1000-1700 nm) window. Although the emission wavelength of probes does matter, it is not clear whether the guideline of "the longer the wavelength, the better the imaging effect" is still suitable for developing phosphorescent probes. For tissue-specific bioimaging, long-lived probes, even if they emit visible phosphorescence, enable accurate visualization of large deep tissues. For studies dealing with bioimaging of tiny biological architectures or dynamic physiopathological activities, the prerequisite is rigorous planning of long-wavelength phosphorescence, being aware of the cooperative contribution of long wavelengths and long lifetimes for improving the spatiotemporal resolution, penetration depth, and sensitivity of bioimaging. In this Review, emerging molecular engineering methods of room-temperature phosphorescence are discussed through the lens of photophysical mechanisms. We highlight the roles of phosphorescence with emission from visible to NIR-II windows toward bioapplications. To appreciate such advances, challenges and prospects in rapidly growing studies of room-temperature phosphorescence are described.
Collapse
Affiliation(s)
- Baisong Chang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Jie Chen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Jiasheng Bao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Taolei Sun
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264000, China
| |
Collapse
|
20
|
Viktorsson K, Rieckmann T, Fleischmann M, Diefenhardt M, Hehlgans S, Rödel F. Advances in molecular targeted therapies to increase efficacy of (chemo)radiation therapy. Strahlenther Onkol 2023; 199:1091-1109. [PMID: 37041372 PMCID: PMC10673805 DOI: 10.1007/s00066-023-02064-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/19/2023] [Indexed: 04/13/2023]
Abstract
Recent advances in understanding the tumor's biology in line with a constantly growing number of innovative technologies have prompted characterization of patients' individual malignancies and may display a prerequisite to treat cancer at its patient individual tumor vulnerability. In recent decades, radiation- induced signaling and tumor promoting local events for radiation sensitization were explored in detail, resulting the development of novel molecular targets. A multitude of pharmacological, genetic, and immunological principles, including small molecule- and antibody-based targeted strategies, have been developed that are suitable for combined concepts with radiation (RT) or chemoradiation therapy (CRT). Despite a plethora of promising experimental and preclinical findings, however, so far, only a very limited number of clinical trials have demonstrated a better outcome and/or patient benefit when RT or CRT are combined with targeted agents. The current review aims to summarize recent progress in molecular therapies targeting oncogenic drivers, DNA damage and cell cycle response, apoptosis signaling pathways, cell adhesion molecules, hypoxia, and the tumor microenvironment to impact therapy refractoriness and to boost radiation response. In addition, we will discuss recent advances in nanotechnology, e.g., RNA technologies and protein-degrading proteolysis-targeting chimeras (PROTACs) that may open new and innovative ways to benefit from molecular-targeted therapy approaches with improved efficacy.
Collapse
Affiliation(s)
- Kristina Viktorsson
- Department of Oncology/Pathology, Karolinska Institutet, Visionsgatan 4, 17164, Solna, Sweden
| | - Thorsten Rieckmann
- Department of Radiation Oncology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of Otolaryngology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Maximilian Fleischmann
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Markus Diefenhardt
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Stephanie Hehlgans
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute (FCI), University of Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
- German Cancer Consortium (DKTK) partner site: Frankfurt, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
21
|
Gertsenshteyn I, Epel B, Giurcanu M, Barth E, Lukens J, Hall K, Martinez JF, Grana M, Maggio M, Miller RC, Sundramoorthy SV, Krzykawska-Serda M, Pearson E, Aydogan B, Weichselbaum RR, Tormyshev VM, Kotecha M, Halpern HJ. Absolute oxygen-guided radiation therapy improves tumor control in three preclinical tumor models. Front Med (Lausanne) 2023; 10:1269689. [PMID: 37904839 PMCID: PMC10613495 DOI: 10.3389/fmed.2023.1269689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 09/28/2023] [Indexed: 11/01/2023] Open
Abstract
Background Clinical attempts to find benefit from specifically targeting and boosting resistant hypoxic tumor subvolumes have been promising but inconclusive. While a first preclinical murine tumor type showed significant improved control with hypoxic tumor boosts, a more thorough investigation of efficacy from boosting hypoxic subvolumes defined by electron paramagnetic resonance oxygen imaging (EPROI) is necessary. The present study confirms improved hypoxic tumor control results in three different tumor types using a clonogenic assay and explores potential confounding experimental conditions. Materials and methods Three murine tumor models were used for multi-modal imaging and radiotherapy: MCa-4 mammary adenocarcinomas, SCC7 squamous cell carcinomas, and FSa fibrosarcomas. Registered T2-weighted MRI tumor boundaries, hypoxia defined by EPROI as pO2 ≤ 10 mmHg, and X-RAD 225Cx CT boost boundaries were obtained for all animals. 13 Gy boosts were directed to hypoxic or equal-integral-volume oxygenated tumor regions and monitored for regrowth. Kaplan-Meier survival analysis was used to assess local tumor control probability (LTCP). The Cox proportional hazards model was used to assess the hazard ratio of tumor progression of Hypoxic Boost vs. Oxygenated Boost for each tumor type controlling for experimental confounding variables such as EPROI radiofrequency, tumor volume, hypoxic fraction, and delay between imaging and radiation treatment. Results An overall significant increase in LTCP from Hypoxia Boost vs. Oxygenated Boost treatments was observed in the full group of three tumor types (p < 0.0001). The effects of tumor volume and hypoxic fraction on LTCP were dependent on tumor type. The delay between imaging and boost treatments did not have a significant effect on LTCP for all tumor types. Conclusion This study confirms that EPROI locates resistant tumor hypoxic regions for radiation boost, increasing clonogenic LTCP, with potential enhanced therapeutic index in three tumor types. Preclinical absolute EPROI may provide correction for clinical hypoxia images using additional clinical physiologic MRI.
Collapse
Affiliation(s)
- Inna Gertsenshteyn
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Department of Radiology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Boris Epel
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
- O2M Technologies, Chicago, IL, United States
| | - Mihai Giurcanu
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, United States
| | - Eugene Barth
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - John Lukens
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Kayla Hall
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Jenipher Flores Martinez
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Mellissa Grana
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Matthew Maggio
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Richard C. Miller
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Subramanian V. Sundramoorthy
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Martyna Krzykawska-Serda
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
- Department of Biophysics and Cancer Biology, Jagiellonian University, Kraków, Poland
| | - Erik Pearson
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| | - Bulent Aydogan
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
| | | | | | - Howard J. Halpern
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, United States
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
22
|
Mittal S, Mallia MB. Molecular imaging of tumor hypoxia: Evolution of nitroimidazole radiopharmaceuticals and insights for future development. Bioorg Chem 2023; 139:106687. [PMID: 37406518 DOI: 10.1016/j.bioorg.2023.106687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
Though growing evidence has been collected in support of the concept of dose escalation based on the molecular level images indicating hypoxic tumor sub-volumes that could be radio-resistant, validation of the concept is still a work in progress. Molecular imaging of tumor hypoxia using radiopharmaceuticals is expected to provide the required input to plan dose escalation through Image Guided Radiation Therapy (IGRT) to kill/control the radio-resistant hypoxic tumor cells. The success of the IGRT, therefore, is heavily dependent on the quality of images obtained using the radiopharmaceutical and the extent to which the image represents the true hypoxic status of the tumor in spite of the heterogeneous nature of tumor hypoxia. Available literature on radiopharmaceuticals for imaging hypoxia is highly skewed in favor of nitroimidazole as the pharmacophore given their ability to undergo oxygen dependent reduction in hypoxic cells. In this context, present review on nitroimidazole radiopharmaceuticals would be immensely helpful to the researchers to obtain a birds-eye view on what has been achieved so far and what can be tried differently to obtain a better hypoxia imaging agent. The review also covers various methods of radiolabeling that could be utilized for developing radiotracers for hypoxia targeting applications.
Collapse
Affiliation(s)
- Sweety Mittal
- Radiopharmaceuticals Division, Bhabha Atomic Research Center, Mumbai 400085, India.
| | - Madhava B Mallia
- Radiopharmaceuticals Division, Bhabha Atomic Research Center, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
23
|
Gul SK, Tepetam H, Yildiz F, Er I, Oksuz DC, Parvizi M, Ozden AS, Alicikus ZA, Sari SY, Alomari O, Gorken IB. Revisiting the Radical Radiotherapy-Radiochemotherapy Results in Anal Canal Cancers: (TROD Gastrointestinal Group Study 02-005). Clin Colorectal Cancer 2023; 22:318-326. [PMID: 37336706 DOI: 10.1016/j.clcc.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND AND AIM This study aimed to determine treatment outcomes and factors affecting prognosis in patients diagnosed with anal canal cancer who received radical radiotherapy (RT) or radiotherapy combined with chemotherapy (CT-RT) in radiation oncology centers in Turkey and compare the results with literature. MATERIAL AND METHOD The study included 193 patients with anal canal cancer reported between 1995 and 2019, of which 162 had complete data. The study was conducted in 11 radiation oncology centers, and a joint database was shared among them. Patients received radiotherapy doses of 45 Gy to 60 Gy. Data analysis was done using SPSS for Windows version 20. RESULTS Median follow-up was 48.51 months (2-214). All patients received radiotherapy, and 140 (86.4%) received concurrent chemotherapy. Radiotherapy doses of 50.4 Gy to 60 Gy were administered to 74 patients (45.7%) using 2-dimensional-3-dimensional (2D-3D) conformal therapy and 70 patients (43.2%) using intensity modulated radiotherapy technique (IMRT). Acute phase hematologic toxicity was observed in 62 patients (38.3%), and nonhematologic toxicity in 123 patients (75.9%). The 5-year overall survival (OS) rate was 75.1% and disease-specific survival (DSS) rate was 76.4%. OS without colostomy was achieved in 79,8 % at 5 years, and complete response in 112 patients (69.1%). OS rate was significantly higher in 142 patients with positive response (P < .000) and 112 with complete response (P < .000). Anemia (P < .002), local progression, and systemic progression (P < .000) resulted in lower OS (P < .002). In univariate analysis, factors affecting OS rate were: gender, age, stage, lymph node status, T stage, RT treatment duration, and treatment planning with PET fusion, which were found to be statistically significant. Completing radiotherapy in less than 45 days, concurrent chemotherapy, and continued administration of mitomycin and 5 FU as chemotherapy had a significant positive effect on overall survival. OS rate was higher in patients receiving RT dose of 58 Gy or less and undergoing IMRT planning in radiotherapy. IMRT was associated with lower acute and late side effects. CONCLUSION Radiochemotherapy is the primary treatment for anal canal cancer and advanced radiotherapy techniques may increase survival by reducing side effects and improving treatment continuation. Higher treatment doses require further investigation. The efficacy of treatment can be improved by including patients treated with modern radiotherapy techniques in multicenter prospective studies using new and more effective chemotherapy and immunotherapy agents.
Collapse
Affiliation(s)
- Sule Karabulut Gul
- Department of Radiation Oncology, University of Health Sciences, Dr. Lutfi Kirdar Kartal Training and Research Hospital, Istanbul, Turkey.
| | - Huseyin Tepetam
- Department of Radiation Oncology, University of Health Sciences, Dr. Lutfi Kirdar Kartal Training and Research Hospital, Istanbul, Turkey
| | - Ferah Yildiz
- Department of Radiation Oncology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ilhami Er
- Department of Radiation Oncology, Izmir Katip Celebi University, Ataturk Training and Research Hospital, Izmir, Turkey
| | - Didem Colpan Oksuz
- Istanbul University Department of Radiation Oncology, Hospital of Cerrahpasa school of Medicine, Istanbul, Turkey
| | - Murtaza Parvizi
- Department of Radiation Oncology, Manisa State Hospital, Manisa, Turkey
| | - Ayse Sevgi Ozden
- Department of Radiation Oncology, University of Health Sciences, Dr. Lutfi Kirdar Kartal Training and Research Hospital, Istanbul, Turkey
| | | | - Sezin Yuce Sari
- Department of Radiation Oncology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Omar Alomari
- Hamidiye International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Ilknur Bilkay Gorken
- Department of Radiation Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
24
|
Yaromina A, Koi L, Schuitmaker L, van der Wiel AMMA, Dubois LJ, Krause M, Lambin P. Overcoming radioresistance with the hypoxia-activated prodrug CP-506: A pre-clinical study of local tumour control probability. Radiother Oncol 2023; 186:109738. [PMID: 37315579 DOI: 10.1016/j.radonc.2023.109738] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND PURPOSE Tumour hypoxia is an established radioresistance factor. A novel hypoxia-activated prodrug CP-506 has been proven to selectively target hypoxic tumour cells and to cause anti-tumour activity. The current study investigates whether CP-506 improves outcome of radiotherapy in vivo. MATERIALS AND METHODS Mice bearing FaDu and UT-SCC-5 xenografts were randomized to receive 5 daily injections of CP-506/vehicle followed by single dose (SD) irradiation. In addition, CP-506 was combined once per week with fractionated irradiation (30 fractions/6 weeks). Animals were followed-up to score all recurrences. In parallel, tumours were harvested to evaluate pimonidazole hypoxia, DNA damage (γH2AX), expression of oxidoreductases. RESULTS CP-506 treatment significantly increased local control rate after SD in FaDu, 62% vs. 27% (p = 0.024). In UT-SCC-5, this effect was not curative and only marginally significant. CP-506 induced significant DNA damage in FaDu (p = 0.009) but not in UT- SCC-5. Hypoxic volume (HV) was significantly smaller (p = 0.038) after pretreatment with CP-506 as compared to vehicle in FaDu but not in less responsive UT-SCC-5. Adding CP-506 to fractionated radiotherapy in FaDu did not result in significant benefit. CONCLUSION The results support the use of CP-506 in combination with radiation in particular using hypofractionation schedules in hypoxic tumours. The magnitude of effect depends on the tumour model, therefore it is expected that applying appropriate patient stratification strategy will further enhance the benefit of CP-506 treatment for cancer patients. A phase I-IIA clinical trial of CP-506 in monotherapy or in combination with carboplatin or a checkpoint inhibitor has been approved (NCT04954599).
Collapse
Affiliation(s)
- Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands.
| | - Lydia Koi
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Lesley Schuitmaker
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | | | - Ludwig Jerome Dubois
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology-OncoRay, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, German Cancer Research Center, Heidelberg, National Center for Tumour Diseases (NCT), partner site Dresden, German Cancer Consortium (DKTK), core center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
25
|
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: 2.0] [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
|
26
|
Pontoriero A, Critelli P, Chillari F, Ferrantelli G, Sciacca M, Brogna A, Parisi S, Pergolizzi S. Modulation of Radiation Doses and Chimeric Antigen Receptor T Cells: A Promising New Weapon in Solid Tumors-A Narrative Review. J Pers Med 2023; 13:1261. [PMID: 37623511 PMCID: PMC10455986 DOI: 10.3390/jpm13081261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Tumor behavior is determined by its interaction with the tumor microenvironment (TME). Chimeric antigen receptor (CART) cell therapy represents a new form of cellular immunotherapy (IT). Immune cells present a different sensitivity to radiation therapy (RT). RT can affect tumor cells both modifying the TME and inducing DNA damage, with different effects depending on the low and high doses delivered, and can favor the expression of CART cells. CART cells are patients' T cells genetically engineered to recognize surface structure and to eradicate cancer cells. High-dose radiation therapy (HDRT, >10-20 Gy/fractions) converts immunologically "cold" tumors into "hot" ones by inducing necrosis and massive inflammation and death. LDRT (low-dose radiation therapy, >5-10 Gy/fractions) increases the expansion of CART cells and leads to non-immunogenetic death. An innovative approach, defined as the LATTICE technique, combines a high dose in higher FDG- uptake areas and a low dose to the tumor periphery. The association of RT and immune checkpoint inhibitors increases tumor immunogenicity and immune response both in irradiated and non-irradiated sites. The aim of this narrative review is to clarify the knowledge, to date, on CART cell therapy and its possible association with radiation therapy in solid tumors.
Collapse
Affiliation(s)
- Antonio Pontoriero
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Paola Critelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Federico Chillari
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Giacomo Ferrantelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Miriam Sciacca
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Anna Brogna
- Radiotherapy Unit, Medical Physics Unit, A.O.U. “G. Martino”, 98125 Messina, Italy;
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| |
Collapse
|
27
|
Abstract
Hypoxia (oxygen deprivation) occurs in most solid malignancies, albeit with considerable heterogeneity. Hypoxia is associated with an aggressive cancer phenotype by promotion of genomic instability, evasion of anti-cancer therapies including radiotherapy and enhancement of metastatic risk. Therefore, hypoxia results in poor cancer outcomes. Targeting hypoxia to improve cancer outcomes is an attractive therapeutic strategy. Hypoxia-targeted dose painting escalates radiotherapy dose to hypoxic sub-volumes, as quantified and spatially mapped using hypoxia imaging. This therapeutic approach could overcome hypoxia-induced radioresistance and improve patient outcomes without the need for hypoxia-targeted drugs. This article will review the premise and underpinning evidence for personalized hypoxia-targeted dose painting. It will present data on relevant hypoxia imaging biomarkers, highlight the challenges and potential benefit of this approach and provide recommendations for future research priorities in this field. Personalized hypoxia-based radiotherapy de-escalation strategies will also be addressed.
Collapse
Affiliation(s)
- Ahmed Salem
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, Hashemite University, Zarqa, Jordan; Division of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
28
|
Rahimy E, Gensheimer MF, Beadle B, Le QT. Lessons and Opportunities for Biomarker-Driven Radiation Personalization in Head and Neck Cancer. Semin Radiat Oncol 2023; 33:336-347. [PMID: 37331788 DOI: 10.1016/j.semradonc.2023.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Head and neck cancer is notoriously challenging to treat in part because it constitutes an anatomically and biologically diverse group of cancers with heterogeneous prognoses. While treatment can be associated with significant late toxicities, recurrence is often difficult to salvage with poor survival rates and functional morbidity.1,2 Thus, achieving tumor control and cure at the initial diagnosis is the highest priority. Given the differing outcome expectations (even within a specific sub-site like oropharyngeal carcinoma), there has been growing interest in personalizing treatment: de-escalation in selected cancers to decrease the risk of late toxicity without compromising oncologic outcomes, and intensification for more aggressive cancers to improve oncologic outcomes without causing undue toxicity. This risk stratification is increasingly accomplished using biomarkers, which can represent molecular, clinicopathologic, and/or radiologic data. In this review, we will focus on biomarker-driven radiotherapy dose personalization with emphasis on oropharyngeal and nasopharyngeal carcinoma. This radiation personalization is largely performed on the population level by identifying patients with good prognosis via traditional clinicopathologic factors, although there are emerging studies supporting inter-tumor and intra-tumor level personalization via imaging and molecular biomarkers.
Collapse
Affiliation(s)
- Elham Rahimy
- Department of Radiation Oncology, Stanford University, Stanford, CA.
| | | | - Beth Beadle
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University, Stanford, CA
| |
Collapse
|
29
|
Dubec MJ, Buckley DL, Berks M, Clough A, Gaffney J, Datta A, McHugh DJ, Porta N, Little RA, Cheung S, Hague C, Eccles CL, Hoskin PJ, Bristow RG, Matthews JC, van Herk M, Choudhury A, Parker GJM, McPartlin A, O'Connor JPB. First-in-human technique translation of oxygen-enhanced MRI to an MR Linac system in patients with head and neck cancer. Radiother Oncol 2023; 183:109592. [PMID: 36870608 DOI: 10.1016/j.radonc.2023.109592] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
BACKGROUND AND PURPOSE Tumour hypoxia is prognostic in head and neck cancer (HNC), associated with poor loco-regional control, poor survival and treatment resistance. The advent of hybrid MRI - radiotherapy linear accelerator or 'MR Linac' systems - could permit imaging for treatment adaptation based on hypoxic status. We sought to develop oxygen-enhanced MRI (OE-MRI) in HNC and translate the technique onto an MR Linac system. MATERIALS AND METHODS MRI sequences were developed in phantoms and 15 healthy participants. Next, 14 HNC patients (with 21 primary or local nodal tumours) were evaluated. Baseline tissue longitudinal relaxation time (T1) was measured alongside the change in 1/T1 (termed ΔR1) between air and oxygen gas breathing phases. We compared results from 1.5 T diagnostic MR and MR Linac systems. RESULTS Baseline T1 had excellent repeatability in phantoms, healthy participants and patients on both systems. Cohort nasal concha oxygen-induced ΔR1 significantly increased (p < 0.0001) in healthy participants demonstrating OE-MRI feasibility. ΔR1 repeatability coefficients (RC) were 0.023-0.040 s-1 across both MR systems. The tumour ΔR1 RC was 0.013 s-1 and the within-subject coefficient of variation (wCV) was 25% on the diagnostic MR. Tumour ΔR1 RC was 0.020 s-1 and wCV was 33% on the MR Linac. ΔR1 magnitude and time-course trends were similar on both systems. CONCLUSION We demonstrate first-in-human translation of volumetric, dynamic OE-MRI onto an MR Linac system, yielding repeatable hypoxia biomarkers. Data were equivalent on the diagnostic MR and MR Linac systems. OE-MRI has potential to guide future clinical trials of biology guided adaptive radiotherapy.
Collapse
Affiliation(s)
- Michael J Dubec
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK.
| | - David L Buckley
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK; Biomedical Imaging, University of Leeds, Leeds, UK
| | - Michael Berks
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Abigael Clough
- Radiotherapy, The Christie NHS Foundation Trust, Manchester, UK
| | - John Gaffney
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Anubhav Datta
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Radiology, The Christie NHS Foundation Trust, Manchester, UK
| | - Damien J McHugh
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Nuria Porta
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Ross A Little
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Susan Cheung
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Christina Hague
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Cynthia L Eccles
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Radiotherapy, The Christie NHS Foundation Trust, Manchester, UK
| | - Peter J Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Department of Clinical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | - Robert G Bristow
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Julian C Matthews
- Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Marcel van Herk
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Geoff J M Parker
- Bioxydyn Ltd, Manchester, UK; Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Andrew McPartlin
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Radiation Oncology, Princess Margaret Cancer Center, Toronto, Canada
| | - James P B O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Radiology, The Christie NHS Foundation Trust, Manchester, UK; Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| |
Collapse
|
30
|
Cadassou O, Petter Jordheim L. OXPHOS inhibitors, metabolism and targeted therapies in cancer. Biochem Pharmacol 2023; 211:115531. [PMID: 37019188 DOI: 10.1016/j.bcp.2023.115531] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
More and more studies highlight the complex metabolic characteristics and plasticity of cancer cells. To address these specificities and explore the associated vulnerabilities, new metabolism-targeting therapeutic strategies are being developed. It is more and more accepted that cancer cells do not produce their energy only from aerobic glycolysis, as some subtypes strongly rely on mitochondrial respiration (OXPHOS). This review focuses on classical and promising OXPHOS inhibitors (OXPHOSi), unravelling their interest and modes of actions in cancer, particularly in combination with other strategies. Indeed, in monotherapy, OXPHOSi display limited efficiency as they mostly trigger cell death in cancer cell subtypes that strongly depend on mitochondrial respiration and are not able to shift to other metabolic pathways to produce energy. Nevertheless, they remain very interesting in combination with conventional therapeutic strategies such as chemotherapy and radiotherapy, increasing their anti-tumoral actions. In addition, OXPHOSi can be included in even more innovative strategies such as combinations with other metabolic drugs or immunotherapies.
Collapse
|
31
|
Lee JJW, Kunaratnam V, Kim CJH, Pienkowski M, Hueniken K, Sahovaler A, Lam ACL, Davies JC, Brown CM, De Almeida JR, Huang SH, Waldron JN, Spreafico A, Hung RJ, Xu W, Goldstein DP, Liu G. Cigarette smoking cessation, duration of smoking abstinence, and head and neck squamous cell carcinoma prognosis. Cancer 2023; 129:867-877. [PMID: 36653915 DOI: 10.1002/cncr.34620] [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: 05/25/2022] [Revised: 09/20/2022] [Accepted: 11/07/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND Tobacco use is a major risk factor for developing head and neck squamous cell carcinoma (HNSCC). However, the prognostic associations with smoking cessation are limited. The authors assessed whether smoking cessation and increased duration of abstinence were associated with improved overall (OS) and HNSCC-specific survival. METHODS Clinicodemographic and smoking data from patients with HNSCC at Princess Margaret Cancer Center (2006-2019) were prospectively collected. Multivariable Cox and Fine and Gray competing-risk models were used to assess the impact of smoking cessation and duration of abstinence on overall mortality and HNSCC-specific/noncancer mortality, respectively. RESULTS Among 2482 patients who had HNSCC, former smokers (adjusted hazard ratio [aHR], 0.71; 95% CI, 0.58-0.87; p = .001; N = 841) had a reduced risk of overall mortality compared with current smokers (N = 931). Compared with current smokers, former smokers who quit >10 years before diagnosis (long-term abstinence; n = 615) had the most improved OS (aHR, 0.72; 95% CI, 0.56-0.93; p = .001). The 5-year actuarial rates of HNSCC-specific and noncancer deaths were 16.8% and 9.4%, respectively. Former smokers (aHR, 0.71; 95% CI, 0.54-0.95; p = .019) had reduced HNSCC-specific mortality compared with current smokers, but there was no difference in noncancer mortality. Abstinence for >10 years was associated with decreased HNSCC-specific death compared with current smoking (aHR, 0.64; 95% CI, 0.46-0.91; p = .012). Smoking cessation with a longer duration of quitting was significantly associated with reduced overall and HNSCC-specific mortality in patients who received primary radiation. CONCLUSIONS Smoking cessation before the time of diagnosis reduced overall mortality and cancer-specific mortality among patients with HNSCC, but no difference was observed in noncancer mortality. Long-term abstinence (>10 pack-years) had a significant OS and HNSCC-specific survival benefit.
Collapse
Affiliation(s)
- John J W Lee
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Vijay Kunaratnam
- Department of Biostatistics, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Christina J H Kim
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Martha Pienkowski
- Department of Biostatistics, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Katrina Hueniken
- Department of Biostatistics, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Axel Sahovaler
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada.,Head and Neck Surgery, University College London Hospitals, London, UK
| | - Andrew C L Lam
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Joel C Davies
- Department of Otolaryngology-Head and Neck Surgery, Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Catherine M Brown
- Department of Biostatistics, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - John R De Almeida
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Shao Hui Huang
- Department of Radiation Oncology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - John N Waldron
- Department of Radiation Oncology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - David P Goldstein
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Department of Biostatistics, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada.,Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
32
|
How the histological structure of some lung cancers shaped almost 70 years of radiobiology. Br J Cancer 2023; 128:407-412. [PMID: 36344595 PMCID: PMC9938174 DOI: 10.1038/s41416-022-02041-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
Pivotal research led by Louis Harold Gray in the 1950s suggested that oxygen plays a vital role during radiotherapy. By proving that tumours have large necrotic cores due to hypoxia and that hypoxic cells require significantly larger doses of ionising radiation to achieve the same cell kill, Thomlinson and Gray inspired the subsequent decades of research into better defining the mechanistic role of molecular oxygen at the time of radiation. Ultimately, the work pioneered by Thomlinson and Gray led to numerous elegant studies which demonstrated that tumour hypoxia predicts for poor patient outcomes. Furthermore, this subsequently resulted in investigations into markers and measurement of hypoxia, as well as modification strategies. However, despite an abundance of pre-clinical data supporting hypoxia-targeted treatments, there is limited widespread application of hypoxia-targeted therapies routinely used in clinical practice. Significant contributing factors underpinning disappointing clinical trial results include the use of model systems which are more hypoxic than human tumours and a failure to stratify patients based on levels of hypoxia. However, translating the original findings of Thomlinson and Gray remains a research priority with the potential to significantly improve patient outcomes and specifically those receiving radiotherapy.
Collapse
|
33
|
Xu YY, Chen YH, Jin J, Yuan Y, Li JM, Cai XJ, Zhang RY. Modulating tumour vascular normalisation using triptolide-loaded NGR-functionalized liposomes for enhanced cancer radiotherapy. J Liposome Res 2023:1-7. [PMID: 36601687 DOI: 10.1080/08982104.2022.2161095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Radiotherapy is an effective therapy in tumour treatment. However, the characteristics of the tumour microenvironment, including hypoxia, low pH, and interstitial fluid pressure bring about radioresistance. To improve the anti-tumour effect of radiotherapy, it has been demonstrated that antiangiogenic therapy can be employed to repair the structural and functional defects of tumour angiogenic vessels, thereby preventing radioresistance or poor therapeutic drug delivery. In this study, we prepared triptolide (TP)-loaded Asn-Gly-Arg (NGR) peptide conjugated mPEG2000-DSPE-targeted liposomes (NGR-PEG-TP-LPs) to induce tumour blood vessel normalisation, to the end of increasing the sensitivity of tumour cells to radiotherapy. Further, to quantify the tumour vessel normalisation window, the structure and functionality of tumour blood vessels post NGR-PEG-TP-LPs treatment were evaluated. Thereafter, the anti-tumour effect of radiotherapy following these treatments was evaluated using HCT116 xenograft-bearing mouse models based on the tumour vessel normalisation period window. The results obtained showed that NGR-PEG-TP-LPs could modulate tumour vascular normalisation to increase the oxygen content of the tumour microenvironment and enhance the efficacy of radiotherapy. Further, liver and kidney toxicity tests indicated that NGR-PEG-TP-LPs are safe for application in cancer treatment.
Collapse
Affiliation(s)
- Ying-Ying Xu
- Department of Pharmacy, Hangzhou Red Cross Hospital (Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College), Hangzhou, People's Republic of China
| | - Yan-Hong Chen
- Laboratory Animal Center of Zhejiang University, Hangzhou, People's Republic of China
| | - Jie Jin
- Department of Pharmacy, Hangzhou Red Cross Hospital (Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College), Hangzhou, People's Republic of China
| | - Yuan Yuan
- Department of Pharmacy, Hangzhou Red Cross Hospital (Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College), Hangzhou, People's Republic of China
| | - Jin-Meng Li
- Department of Pharmacy, Hangzhou Red Cross Hospital (Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College), Hangzhou, People's Republic of China
| | - Xin-Jun Cai
- Department of Pharmacy, Hangzhou Red Cross Hospital (Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College), Hangzhou, People's Republic of China
| | - Ruo-Ying Zhang
- Department of Pharmacy, Hangzhou Red Cross Hospital (Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College), Hangzhou, People's Republic of China
| |
Collapse
|
34
|
Siqueira JM, Heguedusch D, Rodini CO, Nunes FD, Rodrigues MFSD. Mechanisms involved in cancer stem cell resistance in head and neck squamous cell carcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:116-137. [PMID: 37065869 PMCID: PMC10099599 DOI: 10.20517/cdr.2022.107] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/04/2023] [Accepted: 02/08/2023] [Indexed: 04/18/2023]
Abstract
Despite scientific advances in the Oncology field, cancer remains a leading cause of death worldwide. Molecular and cellular heterogeneity of head and neck squamous cell carcinoma (HNSCC) is a significant contributor to the unpredictability of the clinical response and failure in cancer treatment. Cancer stem cells (CSCs) are recognized as a subpopulation of tumor cells that can drive and maintain tumorigenesis and metastasis, leading to poor prognosis in different types of cancer. CSCs exhibit a high level of plasticity, quickly adapting to the tumor microenvironment changes, and are intrinsically resistant to current chemo and radiotherapies. The mechanisms of CSC-mediated therapy resistance are not fully understood. However, they include different strategies used by CSCs to overcome challenges imposed by treatment, such as activation of DNA repair system, anti-apoptotic mechanisms, acquisition of quiescent state and Epithelial-mesenchymal transition, increased drug efflux capacity, hypoxic environment, protection by the CSC niche, overexpression of stemness related genes, and immune surveillance. Complete elimination of CSCs seems to be the main target for achieving tumor control and improving overall survival for cancer patients. This review will focus on the multi-factorial mechanisms by which CSCs are resistant to radiotherapy and chemotherapy in HNSCC, supporting the use of possible strategies to overcome therapy failure.
Collapse
Affiliation(s)
- Juliana Mota Siqueira
- Department of Stomatology, Discipline of Oral and Maxillofacial Pathology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Daniele Heguedusch
- Department of Stomatology, Discipline of Oral and Maxillofacial Pathology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Camila Oliveira Rodini
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, São Paulo 17012-230, Brazil
| | - Fabio Daumas Nunes
- Department of Stomatology, Discipline of Oral and Maxillofacial Pathology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Maria Fernanda Setúbal Destro Rodrigues
- Biophotonics Applied to Health Sciences, Nove de Julho University, UNINOVE, São Paulo 01504-001, Brazil
- Correspondence to: PhD. Maria Fernanda Setúbal Destro Rodrigues. Biophotonics Applied to Health Sciences, Nove de Julho University, UNINOVE, Rua Vergueiro, 235/249 - Liberdade, São Paulo 01504-001, Brazil. E-mail:
| |
Collapse
|
35
|
O'Cathail SM, Chalmers AJ. Integrating Novel Cancer Therapies with Radiation - Illuminating the Tunnel. Clin Oncol (R Coll Radiol) 2023; 35:38-41. [PMID: 36333159 DOI: 10.1016/j.clon.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/15/2022] [Accepted: 10/14/2022] [Indexed: 12/31/2022]
Affiliation(s)
- S M O'Cathail
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | - A J Chalmers
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
36
|
Abstract
ABSTRACT Head and neck squamous cell carcinomas are rising in incidence worldwide, and despite the advent of improved surgical and radiation techniques, a substantial proportion of patients have disease recurrence, where systemic therapies are the mainstay of management. Recent advances in systemic therapy include the development of epidermal growth factor receptor- and programmed death 1-targeting drugs, which have produced incremental improvements in disease outcomes. However, for most patients, responses to treatment remain elusive because of primary or acquired resistance. Novel drugs and rational drug combinations need to be tested based on biomarker identification and preclinical science that will ultimately advance outcomes for our patients. This review focuses on efforts untaken for epidermal growth factor receptor targeting in head and neck squamous cell carcinoma to date.
Collapse
|
37
|
Fletcher T, Thompson AJ, Ashrafian H, Darzi A. The measurement and modification of hypoxia in colorectal cancer: overlooked but not forgotten. Gastroenterol Rep (Oxf) 2022; 10:goac042. [PMID: 36032656 PMCID: PMC9406947 DOI: 10.1093/gastro/goac042] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/18/2022] [Accepted: 07/21/2022] [Indexed: 11/14/2022] Open
Abstract
Tumour hypoxia is the inevitable consequence of a tumour's rapid growth and disorganized, inefficient vasculature. The compensatory mechanisms employed by tumours, and indeed the absence of oxygen itself, hinder the ability of all treatment modalities. The clinical consequence is poorer overall survival, disease-free survival, and locoregional control. Recognizing this, clinicians have been attenuating the effect of hypoxia, primarily with hypoxic modification or with hypoxia-activated pro-drugs, and notable success has been demonstrated. However, in the case of colorectal cancer (CRC), there is a general paucity of knowledge and evidence surrounding the measurement and modification of hypoxia, and this is possibly due to the comparative inaccessibility of such tumours. We specifically review the role of hypoxia in CRC and focus on the current evidence for the existence of hypoxia in CRC, the majority of which originates from indirect positron emission topography imaging with hypoxia selective radiotracers; the evidence correlating CRC hypoxia with poorer oncological outcome, which is largely based on the measurement of hypoxia inducible factor in correlation with clinical outcome; the evidence of hypoxic modification in CRC, of which no direct evidence exists, but is reflected in a number of indirect markers; the prognostic and monitoring implications of accurate CRC hypoxia quantification and its potential in the field of precision oncology; and the present and future imaging tools and technologies being developed for the measurement of CRC hypoxia, including the use of blood-oxygen-level-dependent magnetic resonance imaging and diffuse reflectance spectroscopy.
Collapse
Affiliation(s)
- Teddy Fletcher
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| | - Alex J Thompson
- The Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK
| | - Hutan Ashrafian
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| | - Ara Darzi
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| |
Collapse
|
38
|
Xiang J, He Y, Li Y, Wu K, Cheng M, Wang Y, Chen R. A hypoxia-related lncRNA model for prediction of head and neck squamous cell carcinoma prognosis. Cancer Med 2022; 12:3773-3785. [PMID: 35920349 PMCID: PMC9939198 DOI: 10.1002/cam4.5102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/19/2022] [Accepted: 07/19/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is one of the most common and highly heterogeneous malignancies worldwide. Increasing studies have proven that hypoxia and related long non-coding RNA (lncRNA) are involved in the occurrence and prognosis of HNSCC. The goal of this work is to construct a risk assessment model using hypoxia-related lncRNAs (hrlncRNAs) for HNSCC prognosis prediction and personalized treatment. METHODS Transcriptome expression matrix, clinical follow-up data, and somatic mutation data of HNSCC patients were obtained from The Cancer Genome Atlas (TCGA). We used co-expression analysis to identify hrlncRNAs, then screened for differentially expressed lncRNAs (DEhrlncRNAs), and paired these DEhrlncRNAs. The risk model was established through univariate, least absolute shrinkage and selection operator (LASSO), and stepwise multivariate Cox regression. Finally, we assessed the model from multiple perspectives of tumor mutation burden (TMB), tumor immune infiltration, chemotherapeutic sensitivity, immune checkpoint inhibitor (ICI), and functional enrichment. RESULTS The risk assessment model included 14 hrlncRNA pairs. The risk score was observed to be a reliable prognostic factor. The high-risk patients had an unfavorable prognosis and significant differences from the low-risk group in TMB and tumor immune infiltration. In the high-risk patients, the common immune checkpoints were down-regulated, including CTLA4 and PDCD1, and the sensibility to paclitaxel and docetaxel was higher. The functional enrichment analysis suggested that the low-risk group was accompanied by activated immune function. CONCLUSIONS The risk assessment model of 14-hrlncRNA-pairs demonstrated a promising prognostic prediction for HNSCC patients and can guide personalized clinical treatment.
Collapse
Affiliation(s)
- Junwei Xiang
- Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceCollege & Hospital of StomatologyHefeiChina
| | - Yaodong He
- Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceCollege & Hospital of StomatologyHefeiChina
| | - Yunshan Li
- Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceCollege & Hospital of StomatologyHefeiChina
| | - Kexuan Wu
- Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceCollege & Hospital of StomatologyHefeiChina
| | - Mengxiang Cheng
- Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceCollege & Hospital of StomatologyHefeiChina
| | - Yuanyin Wang
- Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceCollege & Hospital of StomatologyHefeiChina
| | - Ran Chen
- Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceCollege & Hospital of StomatologyHefeiChina
| |
Collapse
|
39
|
Yilmaz D, Tuzer M, Unlu MB. Assessing the therapeutic response of tumors to hypoxia-targeted prodrugs with an in silico approach. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:10941-10962. [PMID: 36124576 DOI: 10.3934/mbe.2022511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Tumor hypoxia is commonly recognized as a condition stimulating the progress of the aggressive phenotype of tumor cells. Hypoxic tumor cells inhibit the delivery of cytotoxic drugs, causing hypoxic areas to receive insufficient amounts of anticancer agents, which results in adverse treatment responses. Being such an obstruction to conventional therapies for cancer, hypoxia might be considered a target to facilitate the efficacy of treatments in the resistive environment of tumor sites. In this regard, benefiting from prodrugs that selectively target hypoxic regions remains an effective approach. Additionally, combining hypoxia-activated prodrugs (HAPs) with conventional chemotherapeutic drugs has been used as a promising strategy to eradicate hypoxic cells. However, determining the appropriate sequencing and scheduling of the combination therapy is also of great importance in obtaining favorable results in anticancer therapy. Here, benefiting from a modeling approach, we study the efficacy of HAPs in combination with chemotherapeutic drugs on tumor growth and the treatment response. Different treatment schedules have been investigated to see the importance of determining the optimal schedule in combination therapy. The effectiveness of HAPs in varying hypoxic conditions has also been explored in the study. The model provides qualitative conclusions about the treatment response, as the maximal benefit is obtained from combination therapy with greater cell death for highly hypoxic tumors. It has also been observed that the antitumor effects of HAPs show a hypoxia-dependent profile.
Collapse
Affiliation(s)
- Defne Yilmaz
- Department of Physics, Bogazici University, Istanbul 34342, Turkey
- Center for Life Sciences and Technologies, Bogazici University, Istanbul 34342, Turkey
| | - Mert Tuzer
- Department of Physics, Bogazici University, Istanbul 34342, Turkey
- Center for Life Sciences and Technologies, Bogazici University, Istanbul 34342, Turkey
| | - Mehmet Burcin Unlu
- Department of Physics, Bogazici University, Istanbul 34342, Turkey
- Center for Life Sciences and Technologies, Bogazici University, Istanbul 34342, Turkey
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-8648, Japan
| |
Collapse
|
40
|
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
|
41
|
Dolezel M, Slavik M, Blazek T, Kazda T, Koranda P, Veverkova L, Burkon P, Cvek J. FMISO-Based Adaptive Radiotherapy in Head and Neck Cancer. J Pers Med 2022; 12:jpm12081245. [PMID: 36013194 PMCID: PMC9410424 DOI: 10.3390/jpm12081245] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/12/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Concurrent chemoradiotherapy represents one of the most used strategies in the curative treatment of patients with head and neck (HNC) cancer. Locoregional failure is the predominant recurrence pattern. Tumor hypoxia belongs to the main cause of treatment failure. Positron emission tomography (PET) using hypoxia radiotracers has been studied extensively and has proven its feasibility and reproducibility to detect tumor hypoxia. A number of studies confirmed that the uptake of FMISO in the recurrent region is significantly higher than that in the non-recurrent region. The escalation of dose to hypoxic tumors may improve outcomes. The technical feasibility of optimizing radiotherapeutic plans has been well documented. To define the hypoxic tumour volume, there are two main approaches: dose painting by contour (DPBC) or by number (DPBN) based on PET images. Despite amazing technological advances, precision in target coverage, and surrounding tissue sparring, radiation oncology is still not considered a targeted treatment if the “one dose fits all” approach is used. Using FMISO and other hypoxia tracers may be an important step for individualizing radiation treatment and together with future radiomic principles and a possible genome-based adjusting dose, will move radiation oncology into the precise and personalized era.
Collapse
Affiliation(s)
- Martin Dolezel
- Department of Oncology, Palacky University Medical School & Teaching Hospital, 77900 Olomouc, Czech Republic;
| | - Marek Slavik
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute, 65652 Brno, Czech Republic; (T.K.); (P.B.)
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
- Correspondence:
| | - Tomas Blazek
- Department of Oncology, Faculty of Medicine, University Hospital Ostrava, 70852 Ostrava, Czech Republic; (T.B.); (J.C.)
| | - Tomas Kazda
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute, 65652 Brno, Czech Republic; (T.K.); (P.B.)
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Pavel Koranda
- Department of Nuclear Medicine, Palacky University Medical School & Teaching Hospital, 77900 Olomouc, Czech Republic;
| | - Lucia Veverkova
- Department of Radiology, Palacky University Medical School & Teaching Hospital, 77900 Olomouc, Czech Republic;
| | - Petr Burkon
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute, 65652 Brno, Czech Republic; (T.K.); (P.B.)
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Jakub Cvek
- Department of Oncology, Faculty of Medicine, University Hospital Ostrava, 70852 Ostrava, Czech Republic; (T.B.); (J.C.)
| |
Collapse
|
42
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
43
|
Griazov A, Griazov A, Grydina N, Stuley V. Stereotactic radiosurgery of radioresistant glioblastomas. The ways of overcoming radioresistance of hypoxic tumors. УКРАЇНСЬКИЙ РАДІОЛОГІЧНИЙ ТА ОНКОЛОГІЧНИЙ ЖУРНАЛ 2022. [DOI: 10.46879/ukroj.2.2022.25-40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Background. Taking into account high degree of resistance of glioblastoma to radiation therapy, and also low overall survival rates of patients, it is necessary to develop improved methods of treating this pathology, in particular, complex combined treatment with radiation therapy and radiosensitizers.
Purpose – to assess the effectiveness of radiosensitization of hypoxic tumors in radiosurgical treatment of glioblastomas; to increase non-recurrent and overall survival rate of patients. Materials and methods. Stereotactic radiosurgery (SRS) of glioblastoma was performed in 106 patients (average age – 53 years), 66 males (62,26%) and 40 females (37,73%). The average dose was 18 Gy in a single-fraction SRS, and 32 Gy (7 Gy per fraction) in multi-fraction SRS. The average volume tumor was 29 cm3 . The treatment group consisted of 66 patients who underwent SRS with radiosensitization. 40 patients made up the control group and underwent SRS without radiosensitization.
Results. Median overall survival (MOS) was 20 months in the group with radiosensitization, whereas in the control group it was 12 months. 10-month recurrence-free period after radiosurgery was observed in 95,4% of the patients of the group with radiosensitization and in 70,6% of the patients of the control group. MOS after SRS was similar between the patients with wild-type IDH tumors and patients with tumors with IDH mutation (10,0 months and 11,0 months respectively), and also between the patients with MGMT-methylated tumors and patients with MGMT-nonmethylated tumors (11,2 and 10,2 months respectively). Among all the treated patients, in 20 of them (16,6%) side radiation effects after SRS were observed, and in 9 patients (7,5%) radiation necrosis developed in 3 to 16 months after SRS. The signs of moderate toxicity in the form of vomiting were observed in 6,6% of the patients of the subgroup with metronidazole. There were no signs of toxicity in the subgroup with nimorazole.
Conclusions. Radiosensitization improves rates of overall survival by 53,3% and recurrence-free survival by 24,8 % in performing SRS of hypoxic radioresistant glioblastomas. Nimorazole and metronidazole are powerful radiosensitizers which increase radiosensitivity of tumor cells through enhancing oxygen saturation of hypoxic cells. In order to determine indications for performing SRS with radiosensitization and periods for performing an SRS session we must take into consideration the result of an oxygen test (level of oxygen saturation of the tumor), the peak of signal intensity in the zone of active tumor growth and the peak of saturation of the whole tumor volume.
Collapse
|
44
|
Steele CD, Abbasi A, Islam SMA, Bowes AL, Khandekar A, Haase K, Hames-Fathi S, Ajayi D, Verfaillie A, Dhami P, McLatchie A, Lechner M, Light N, Shlien A, Malkin D, Feber A, Proszek P, Lesluyes T, Mertens F, Flanagan AM, Tarabichi M, Van Loo P, Alexandrov LB, Pillay N. Signatures of copy number alterations in human cancer. Nature 2022; 606:984-991. [PMID: 35705804 PMCID: PMC9242861 DOI: 10.1038/s41586-022-04738-6] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 04/07/2022] [Indexed: 12/15/2022]
Abstract
Gains and losses of DNA are prevalent in cancer and emerge as a consequence of inter-related processes of replication stress, mitotic errors, spindle multipolarity and breakage-fusion-bridge cycles, among others, which may lead to chromosomal instability and aneuploidy1,2. These copy number alterations contribute to cancer initiation, progression and therapeutic resistance3-5. Here we present a conceptual framework to examine the patterns of copy number alterations in human cancer that is widely applicable to diverse data types, including whole-genome sequencing, whole-exome sequencing, reduced representation bisulfite sequencing, single-cell DNA sequencing and SNP6 microarray data. Deploying this framework to 9,873 cancers representing 33 human cancer types from The Cancer Genome Atlas6 revealed a set of 21 copy number signatures that explain the copy number patterns of 97% of samples. Seventeen copy number signatures were attributed to biological phenomena of whole-genome doubling, aneuploidy, loss of heterozygosity, homologous recombination deficiency, chromothripsis and haploidization. The aetiologies of four copy number signatures remain unexplained. Some cancer types harbour amplicon signatures associated with extrachromosomal DNA, disease-specific survival and proto-oncogene gains such as MDM2. In contrast to base-scale mutational signatures, no copy number signature was associated with many known exogenous cancer risk factors. Our results synthesize the global landscape of copy number alterations in human cancer by revealing a diversity of mutational processes that give rise to these alterations.
Collapse
Affiliation(s)
- Christopher D Steele
- Research Department of Pathology, Cancer Institute, University College London, London, UK
| | - Ammal Abbasi
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - S M Ashiqul Islam
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Amy L Bowes
- Research Department of Pathology, Cancer Institute, University College London, London, UK
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
| | - Azhar Khandekar
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Kerstin Haase
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
| | - Shadi Hames-Fathi
- Research Department of Pathology, Cancer Institute, University College London, London, UK
| | - Dolapo Ajayi
- Research Department of Pathology, Cancer Institute, University College London, London, UK
| | | | - Pawan Dhami
- CRUK-UCL Cancer Institute Translational Technology Platform (Genomics), London, UK
| | - Alex McLatchie
- CRUK-UCL Cancer Institute Translational Technology Platform (Genomics), London, UK
| | - Matt Lechner
- Research Department of Oncology, UCL Cancer Institute, London, UK
| | - Nicholas Light
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Adam Shlien
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - David Malkin
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Feber
- Translational Epigenetics, Division of Molecular Pathology, Institute of Cancer Research, London, UK
- Clinical Genomics, Translational Research Laboratory, Royal Marsden NHS Trust, London, UK
| | - Paula Proszek
- Translational Epigenetics, Division of Molecular Pathology, Institute of Cancer Research, London, UK
- Clinical Genomics, Translational Research Laboratory, Royal Marsden NHS Trust, London, UK
| | - Tom Lesluyes
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
| | - Fredrik Mertens
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Clinical Genetics and Pathology, Division of Laboratory Medicine, Lund, Sweden
| | - Adrienne M Flanagan
- Research Department of Pathology, Cancer Institute, University College London, London, UK
- Department of Cellular and Molecular Pathology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, UK
| | - Maxime Tarabichi
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
- Institute for Interdisciplinary Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Peter Van Loo
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA.
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA.
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA.
| | - Nischalan Pillay
- Research Department of Pathology, Cancer Institute, University College London, London, UK.
- Department of Cellular and Molecular Pathology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, UK.
| |
Collapse
|
45
|
The use of radiosensitizing agents in the therapy of glioblastoma multiforme-a comprehensive review. Strahlenther Onkol 2022; 198:507-526. [PMID: 35503461 PMCID: PMC9165247 DOI: 10.1007/s00066-022-01942-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/30/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Glioblastoma is the most common malignant brain tumor in human adults. Despite several improvements in resective as well as adjuvant therapy over the last decades, its overall prognosis remains poor. As a means of improving patient outcome, the possibility of enhancing radiation response by using radiosensitizing agents has been tested in an array of studies. METHODS A comprehensive review of clinical trials involving radiation therapy in combination with radiosensitizing agents on patients diagnosed with glioblastoma was performed in the National Center for Biotechnology Information's PubMed database. RESULTS A total of 96 papers addressing this matter were published between 1976 and 2021, of which 63 matched the subject of this paper. All papers were reviewed, and their findings discussed in the context of their underlining mechanisms of radiosensitization. CONCLUSION In the history of glioblastoma treatment, several approaches of optimizing radiation-effectiveness using radiosensitizers have been made. Even though several different strategies and agents have been explored, clear evidence of improved patient outcome is still missing. Tissue-selectiveness and penetration of the blood-brain barrier seem to be major roadblocks; nevertheless, modern strategies try to circumvent these obstacles, using novel sensitizers based on preclinical data or alternative ways of delivery.
Collapse
|
46
|
Exploring hypoxic biology to improve radiotherapy outcomes. Expert Rev Mol Med 2022; 24:e21. [DOI: 10.1017/erm.2022.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Swartz JE, Smits HJG, Philippens MEP, de Bree R, H A M Kaanders J, Willems SM. Correlation and colocalization of HIF-1α and pimonidazole staining for hypoxia in laryngeal squamous cell carcinomas: A digital, single-cell-based analysis. Oral Oncol 2022; 128:105862. [PMID: 35447566 DOI: 10.1016/j.oraloncology.2022.105862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/31/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Tumor hypoxia results in worse local control and patient survival. We performed a digital, single-cell-based analysis to compare two biomarkers for hypoxia (hypoxia-inducible factor 1-alpha [HIF-1α] and pimonidazole [PIMO]) and their effect on outcome in laryngeal cancer patients treated with accelerated radiotherapy with or without carbogen breathing and nicotinamide (AR versus ARCON). MATERIALS AND METHODS Immunohistochemical staining was performed for HIF-1α and PIMO in consecutive sections of 44 laryngeal cancer patients randomized between AR and ARCON. HIF-1α expression and PIMO-binding were correlated using digital image analysis in QuPath. High-density areas for each biomarker were automatically annotated and staining overlap was analyzed. Kaplan-Meier survival analyses for local control, regional control and disease-free survival were performed to predict a response benefit of ARCON over AR alone for each biomarker. RESULTS 106 Tissue fragments of 44 patients were analyzed. A weak, significant positive correlation was observed between HIF-1α and PIMO positivity on fragment level, but not on patient level. A moderate strength correlation (r = 0.705, p < 0.001) was observed between the number of high-density staining areas for both biomarkers. Staining overlap was poor. HIF-1α expression, PIMO-binding or a combination could not predict a response benefit of ARCON over AR. CONCLUSION Digital image analysis to compare positive cell fractions and staining overlap between two hypoxia biomarkers using open-source software is feasible. Our results highlight that there are distinct differences between HIF-1α and PIMO as hypoxia biomarkers and therefore suggest co-existence of different forms of hypoxia within a single tumor.
Collapse
Affiliation(s)
- Justin E Swartz
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Hilde J G Smits
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johannes H A M Kaanders
- Department of Radiation Oncology, University Medical Center Nijmegen, Nijmegen, the Netherlands
| | - Stefan M Willems
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
48
|
Schouten D, van Os R, Westermann AM, Crezee H, van Tienhoven G, Kolff MW, Bins AD. A randomized phase-II study of reirradiation and hyperthermia versus reirradiation and hyperthermia plus chemotherapy for locally recurrent breast cancer in previously irradiated area. Acta Oncol 2022; 61:441-448. [PMID: 35139725 DOI: 10.1080/0284186x.2022.2033315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND In patients with inoperable local regional recurrences of breast cancer in previously irradiated areas, local control is difficult to maintain and treatment options are limited. The Dutch standard treatment for such recurrences is reirradiation combined with hyperthermia. Apart from enhancing the effect of reirradiation, hyperthermia is also known to improve local effects of chemotherapy like cisplatin. This randomized phase-II trial compares reirradiation and hyperthermia versus the same treatment combined with cisplatin. PATIENTS AND METHODS From December 2010 up to January 2019, 49 patients were randomized, 27 in the standard arm and 22 in the combined arm. A total of 32 Gy was given in eight fractions of 4 Gy in 4 weeks, at two fractions per week. After January 2015, the radiation schedule was changed to 46 Gy in 23 fractions of 2 Gy, at five fractions per week. Hyperthermia was added once a week after radiotherapy. The combined arm was treated with four cycles of weekly cisplatin 40 mg/m2. RESULTS Complete response rate was 60.9% in the standard arm and 61.1% in the combined arm (p = 0.87). Partial response rate was 30.4% in the standard arm and 33.3% in the combined arm (p = 0.79). One-year overall survival was 63.4% in the standard arm and 57.4% in the combined arm. One-year local progression-free interval was 81.5% in the standard arm and 88.1% in the combined arm (p = 0.95). Twenty-five percentage of patients in the standard arm experienced grade 3 or 4 acute toxicity and 29% of patients in the combined arm (p = 0.79). CONCLUSION No potential benefit could be detected of adding cisplatin to reirradiation and hyperthermia in patients with recurrent breast cancer in a previously irradiated area. With or without cisplatin, most patients had subsequent local control until last follow-up or death.
Collapse
Affiliation(s)
- Daphne Schouten
- Department of Radiotherapy, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Rob van Os
- Department of Radiotherapy, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Anneke M. Westermann
- Medical Oncology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Hans Crezee
- Department of Radiotherapy, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Geertjan van Tienhoven
- Department of Radiotherapy, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - M. Willemijn Kolff
- Department of Radiotherapy, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Adriaan D. Bins
- Medical Oncology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
49
|
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: 4.7] [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
|
50
|
Lefebvre TL, Brown E, Hacker L, Else T, Oraiopoulou ME, Tomaszewski MR, Jena R, Bohndiek SE. The Potential of Photoacoustic Imaging in Radiation Oncology. Front Oncol 2022; 12:803777. [PMID: 35311156 PMCID: PMC8928467 DOI: 10.3389/fonc.2022.803777] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/07/2022] [Indexed: 12/16/2022] Open
Abstract
Radiotherapy is recognized globally as a mainstay of treatment in most solid tumors and is essential in both curative and palliative settings. Ionizing radiation is frequently combined with surgery, either preoperatively or postoperatively, and with systemic chemotherapy. Recent advances in imaging have enabled precise targeting of solid lesions yet substantial intratumoral heterogeneity means that treatment planning and monitoring remains a clinical challenge as therapy response can take weeks to manifest on conventional imaging and early indications of progression can be misleading. Photoacoustic imaging (PAI) is an emerging modality for molecular imaging of cancer, enabling non-invasive assessment of endogenous tissue chromophores with optical contrast at unprecedented spatio-temporal resolution. Preclinical studies in mouse models have shown that PAI could be used to assess response to radiotherapy and chemoradiotherapy based on changes in the tumor vascular architecture and blood oxygen saturation, which are closely linked to tumor hypoxia. Given the strong relationship between hypoxia and radio-resistance, PAI assessment of the tumor microenvironment has the potential to be applied longitudinally during radiotherapy to detect resistance at much earlier time-points than currently achieved by size measurements and tailor treatments based on tumor oxygen availability and vascular heterogeneity. Here, we review the current state-of-the-art in PAI in the context of radiotherapy research. Based on these studies, we identify promising applications of PAI in radiation oncology and discuss the future potential and outstanding challenges in the development of translational PAI biomarkers of early response to radiotherapy.
Collapse
Affiliation(s)
- Thierry L. Lefebvre
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Emma Brown
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Lina Hacker
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Thomas Else
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Mariam-Eleni Oraiopoulou
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Michal R. Tomaszewski
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Rajesh Jena
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Sarah E. Bohndiek
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|