1
|
McClurg DP, Sanghera C, Mukherjee S, Fitzgerald RC, Jones CM. A systematic review of circulating predictive and prognostic biomarkers to aid the personalised use of radiotherapy in the radical treatment of patients with oesophageal cancer. Radiother Oncol 2024; 195:110224. [PMID: 38479442 DOI: 10.1016/j.radonc.2024.110224] [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: 01/12/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The availability of circulating biomarkers that are predictive of treatment response or prognostic of overall outcome could enable the personalised and adaptive use of radiotherapy (RT) in patients with oesophageal adenocarcinoma (OAC) and squamous cell carcinoma (OSCC). METHODS A systematic review was carried out following Preferred Reporting Items for Systematic Reviews guidance. Medline, EMBASE, PubMed, Cochrane Library, CINAHL, Scopus and the Web of Science databases were searched for studies published between January 2005-February 2023 relating to circulating biomarkers evaluated in the context of neoadjuvant or definitive RT delivered for OAC/OSCC. Study quality was assessed using predefined criteria. RESULTS A total of 3012 studies were screened and 57 subsequently included, across which 61 biomarkers were reported. A majority (43/57,75.4%) of studies were of Asian origin and retrospective (40/57, 70.2%), with most (52/57, 91.2%) biomarkers reported in the context of patients with OSCC. There was marked inter-study heterogeneity in patient populations, treatment characteristics, biomarker measurement and the cut points used to define biomarker positivity. Nevertheless, there is evidence for the prognostic and predictive value of circulating tumour DNA and numerous miRNAs in OAC and OSCC, as well as for the prognostic and predictive value of circulating levels of CYFRA21.1 in OSCC. CONCLUSIONS There is consistent evidence for the potential predictive and prognostic value of a small number of biomarkers in OSCC and OAC, though these data are insufficient for translation to current clinical practice. Well-designed prospective studies are now required to validate their role in stratified and personalised RT treatment approaches.
Collapse
Affiliation(s)
- Dylan P McClurg
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Chandan Sanghera
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Somnath Mukherjee
- Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Christopher M Jones
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
2
|
Bleaney CW, Abdelaal H, Reardon M, Anandadas C, Hoskin P, Choudhury A, Forker L. Clinical Biomarkers of Tumour Radiosensitivity and Predicting Benefit from Radiotherapy: A Systematic Review. Cancers (Basel) 2024; 16:1942. [PMID: 38792019 PMCID: PMC11119069 DOI: 10.3390/cancers16101942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/18/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Modern advanced radiotherapy techniques have improved the precision and accuracy of radiotherapy delivery, with resulting plans being highly personalised based on individual anatomy. Adaptation for individual tumour biology remains elusive. There is an unmet need for biomarkers of intrinsic radiosensitivity that can predict tumour response to radiation to facilitate individualised decision-making, dosing and treatment planning. Over the last few decades, the use of high throughput molecular biology technologies has led to an explosion of newly discovered cancer biomarkers. Gene expression signatures are now used routinely in clinic to aid decision-making regarding adjuvant systemic therapy. They have great potential as radiotherapy biomarkers. A previous systematic review published in 2015 reported only five studies of signatures evaluated for their ability to predict radiotherapy benefits in clinical cohorts. This updated systematic review encompasses the expanded number of studies reported in the last decade. An additional 27 studies were identified. In total, 22 distinct signatures were recognised (5 pre-2015, 17 post-2015). Seventeen signatures were 'radiosensitivity' signatures and five were breast cancer prognostic signatures aiming to identify patients at an increased risk of local recurrence and therefore were more likely to benefit from adjuvant radiation. Most signatures (15/22) had not progressed beyond the discovery phase of development, with no suitable validated clinical-grade assay for application. Very few signatures (4/17 'radiosensitivity' signatures) had undergone any laboratory-based biological validation of their ability to predict tumour radiosensitivity. No signatures have been assessed prospectively in a phase III biomarker-led trial to date and none are recommended for routine use in clinical guidelines. A phase III prospective evaluation is ongoing for two breast cancer prognostic signatures. The most promising radiosensitivity signature remains the radiosensitivity index (RSI), which is used to calculate a genomic adjusted radiation dose (GARD). There is an ongoing phase II prospective biomarker-led study of RSI/GARD in triple negative breast cancer. The results of these trials are eagerly anticipated over the coming years. Future work in this area should focus on (1) robust biological validation; (2) building biobanks alongside large radiotherapy randomised controlled trials with dose variance (to demonstrate an interaction between radiosensitivity signature and dose); (3) a validation of clinical-grade cost-effective assays that are deliverable within current healthcare infrastructure; and (4) an integration with biomarkers of other determinants of radiation response.
Collapse
Affiliation(s)
- Christopher W. Bleaney
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Hebatalla Abdelaal
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Mark Reardon
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
| | - Carmel Anandadas
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Peter Hoskin
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Ananya Choudhury
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Laura Forker
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
3
|
Liew H, Tessonnier T, Mein S, Magro G, Glimelius L, Coniavitis E, Held T, Haberer T, Abdollahi A, Debus J, Dokic I, Mairani A. Robustness of carbon-ion radiotherapy against DNA damage repair associated radiosensitivity variation based on a biophysical model. Med Phys 2024; 51:3782-3795. [PMID: 38569067 DOI: 10.1002/mp.17045] [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: 12/22/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Interpatient variation of tumor radiosensitivity is rarely considered during the treatment planning process despite its known significance for the therapeutic outcome. PURPOSE To apply our mechanistic biophysical model to investigate the biological robustness of carbon ion radiotherapy (CIRT) against DNA damage repair interference (DDRi) associated patient-to-patient variability in radiosensitivity and its potential clinical advantages against conventional radiotherapy approaches. METHODS AND MATERIALS The "UNIfied and VERSatile bio response Engine" (UNIVERSE) was extended by carbon ions and its predictions were compared to a panel of in vitro and in vivo data including various endpoints and DDRi settings within clinically relevant dose and linear energy transfer (LET) ranges. The implications of UNIVERSE predictions were then assessed in a clinical patient scenario considering DDRi variance. RESULTS UNIVERSE tests well against the applied benchmarks. While in vitro survival curves were predicted with an R2 > 0.92, deviations from in vivo RBE data were less than 5.6% The conducted paradigmatic patient plan study implies a markedly reduced significance of DDRi based radiosensitivity variability in CIRT (13% change ofD 50 ${{D}_{50}}$ in target) compared to conventional radiotherapy (62%) and that boosting the LET within the target further amplifies this robustness of CIRT (8%). In the case of heightened tumor radiosensitivity, a dose de-escalation strategy for photons allows a reduction of the maximum effective dose within the normal tissue (NT) from aD 2 ${{D}_2}$ of 2.65 to 1.64 Gy, which lies below the level found for CIRT (D 2 ${{D}_2}$ = 2.41 Gy) for the analyzed plan and parameters. However, even after de-escalation, the integral effective dose in the NT is found to be substantially higher for conventional radiotherapy in comparison to CIRT (D m e a n ${{D}_{mean}}$ of 0.75, 0.46, and 0.24 Gy for the conventional plan, its de-escalation and CIRT, respectively). CONCLUSIONS The framework offers adequate predictions of in vitro and in vivo radiation effects of CIRT while allowing the consideration of DRRi based solely on parameters derived from photon data. The results of the patient planning study underline the potential of CIRT to minimize important sources of interpatient divergence in therapy outcome, especially when combined with techniques that allow to maximize the LET within the tumor. Despite the potential of de-escalation strategies for conventional radiotherapy to reduce the maximum effective dose in the NT, CIRT appears to remain a more favorable option due to its ability to reduce the integral effective dose within the NT.
Collapse
Affiliation(s)
- Hans Liew
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Consortium (DKTK) Core-Center Heidelberg, 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, 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, Heidelberg, Germany
| | - Thomas Tessonnier
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stewart Mein
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Consortium (DKTK) Core-Center Heidelberg, 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, 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, Heidelberg, Germany
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Giuseppe Magro
- National Center for Oncological Hadrontherapy (CNAO), Medical Physics, Pavia, Italy
| | | | | | - Thomas Held
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Haberer
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
| | - Amir Abdollahi
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Consortium (DKTK) Core-Center Heidelberg, 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, 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, Heidelberg, Germany
| | - Jürgen Debus
- Division of Molecular and Translational 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, Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Ivana Dokic
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Consortium (DKTK) Core-Center Heidelberg, 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, 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, Heidelberg, Germany
| | - Andrea Mairani
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, Heidelberg, Germany
- National Center for Oncological Hadrontherapy (CNAO), Medical Physics, Pavia, Italy
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Tumor Diseases (NCT), University Hospital Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
4
|
Shannon AH, Manne A, Diaz Pardo DA, Pawlik TM. Combined radiotherapy and immune checkpoint inhibition for the treatment of advanced hepatocellular carcinoma. Front Oncol 2023; 13:1193762. [PMID: 37554167 PMCID: PMC10405730 DOI: 10.3389/fonc.2023.1193762] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/03/2023] [Indexed: 08/10/2023] Open
Abstract
Hepatocellular Carcinoma (HCC) is one of the most common cancers and a leading cause of cancer related death worldwide. Until recently, systemic therapy for advanced HCC, defined as Barcelona Clinic Liver Cancer (BCLC) stage B or C, was limited and ineffective in terms of long-term survival. However, over the past decade, immune check point inhibitors (ICI) combinations have emerged as a potential therapeutic option for patients with nonresectable disease. ICI modulate the tumor microenvironment to prevent progression of the tumor. Radiotherapy is a crucial tool in treating unresectable HCC and may enhance the efficacy of ICI by manipulating the tumor microenvironment and decreasing tumor resistance to certain therapies. We herein review developments in the field of ICI combined with radiotherapy for the treatment of HCC, as well as look at challenges associated with these treatment modalities, and review future directions of combination therapy.
Collapse
Affiliation(s)
- Alexander H. Shannon
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Ashish Manne
- Department of Internal Medicine, Division of Medical Oncology at the Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Dayssy A. Diaz Pardo
- Department of Radiation Oncology, The Ohio State University, Comprehensive Cancer Center-James Hospital and Solove Research Institute, Columbus, OH, United States
| | - Timothy M. Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
5
|
Gao Z, Zhao Q, Xu Y, Wang L. Improving the efficacy of combined radiotherapy and immunotherapy: focusing on the effects of radiosensitivity. Radiat Oncol 2023; 18:89. [PMID: 37226275 DOI: 10.1186/s13014-023-02278-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
Cancer treatment is gradually entering an era of precision, with multitude studies in gene testing and immunotherapy. Tumor cells can be recognized and eliminated by the immune system through the expression of tumor-associated antigens, but when the cancer escapes or otherwise suppresses immunity, the balance between cancer cell proliferation and immune-induced cancer cell killing may be interrupted, resulting in tumor proliferation and progression. There has been significant attention to combining conventional cancer therapies (i.e., radiotherapy) with immunotherapy as opposed to treatment alone. The combination of radio-immunotherapy has been demonstrated in both basic research and clinical trials to provide more effective anti-tumor responses. However, the absolute benefits of radio-immunotherapy are dependent on individual characteristics and not all patients can benefit from radio-immunotherapy. At present, there are numerous articles about exploring the optimal models for combination radio-immunotherapy, but the factors affecting the efficacy of the combination, especially with regard to radiosensitivity remain inconclusive. Radiosensitivity is a measure of the response of cells, tissues, or individuals to ionizing radiation, and various studies have shown that the radiosensitivity index (RSI) will be a potential biomarker for predicting the efficacy of combination radio-immunotherapy. The purpose of this review is to focus on the factors that influence and predict the radiosensitivity of tumor cells, and to evaluate the impact and predictive significance of radiosensitivity on the efficacy of radio-immunotherapy combination.
Collapse
Affiliation(s)
- Zhiru Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Qian Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430064, China
| | - Yiyue Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
6
|
Liu D, Wei M, Yan W, Xie H, Sun Y, Yuan B, Jin Y. Potential applications of drug delivery technologies against radiation enteritis. Expert Opin Drug Deliv 2023; 20:435-455. [PMID: 36809906 DOI: 10.1080/17425247.2023.2183948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
INTRODUCTION The incidence of abdominal tumors, such as colorectal and prostate cancers, continually increases. Radiation therapy is widely applied in the clinical treatment of patients with abdominal/pelvic cancers, but it often unfortunately causes radiation enteritis (RE) involving the intestine, colon, and rectum. However, there is a lack of suitable treatment options for effective prevention and treatment of RE. AREAS COVERED Conventional clinical drugs for preventing and treating RE are usually applied by enemas and oral administration. Innovative gut-targeted drug delivery systems including hydrogels, microspheres, and nanoparticles are proposed to improve the prevention and curation of RE. EXPERT OPINION The prevention and treatment of RE have not attracted sufficient attention in the clinical practice, especially compared to the treatment of tumors, although RE takes patients great pains. Drug delivery to the pathological sites of RE is a huge challenge. The short retention and weak targeting of conventional drug delivery systems affect the therapeutic efficiency of anti-RE drugs. Novel drug delivery systems including hydrogels, microspheres, and nanoparticles can allow drugs long-term retention in the gut and targeting the inflammation sites to alleviate radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Meng Wei
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wenrui Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hua Xie
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yingbao Sun
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Bochuan Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
7
|
Sigurdson S, Thibodeau S, Korzeniowski M, Moraes FY. A Precise Approach for Radiotherapy of Breast Cancer. Cancer Treat Res 2023; 188:175-198. [PMID: 38175346 DOI: 10.1007/978-3-031-33602-7_7] [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] [Indexed: 01/05/2024]
Abstract
Radiotherapy is an integral part of the multidisciplinary management of breast cancer (BC). There have been multiple recent advances in the delivery of radiotherapy, reviewed with a critical discussion of the evidence from trials investigating adjuvant ultra-hypofractionation and partial breast irradiation for early-stage BC, and the locoregional management of lymph nodes in locally advanced BC. Multiple precision medicine-based approaches have been developed as prognostic and/or predictive for BC patients and identifying biomarkers of radioresistance could help identify patients that may benefit from dose-escalated radiotherapy or radiosensitizers. Radiotherapy after breast reconstruction is an area of current controversy in the field, and we evaluated the decision-making considerations in this situation. The oligometastatic state is an emerging field for many cancer sites based on recent trials investigating ablative radiotherapy for oligometastatic BC. This chapter is an overview of radiotherapy for BC, with a focus on recent advances in early-stage, locally advanced, and oligometastatic disease.
Collapse
Affiliation(s)
- Samantha Sigurdson
- Department of Oncology - Division of Radiation Oncology, Kingston Health Sciences Centre and Queen's University, Kingston, Canada
| | - Stephane Thibodeau
- Department of Oncology - Division of Radiation Oncology, Kingston Health Sciences Centre and Queen's University, Kingston, Canada
| | - Martin Korzeniowski
- Department of Oncology - Division of Radiation Oncology, Kingston Health Sciences Centre and Queen's University, Kingston, Canada
| | - Fabio Ynoe Moraes
- Department of Oncology - Division of Radiation Oncology, Kingston Health Sciences Centre and Queen's University, Kingston, Canada.
| |
Collapse
|
8
|
Polygenic risk score for prediction of radiotherapy efficacy and radiosensitivity in patients with non-metastatic breast cancer. RADIATION MEDICINE AND PROTECTION 2023. [DOI: 10.1016/j.radmp.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
9
|
Montori A, Germani A, Ferri M, Milano A, Ranalli TV, Piane M, Pilozzi E. Somatic NGS Analysis of DNA Damage Response (DDR) Genes ATM, MRE11A, RAD50, NBN, and ATR in Locally Advanced Rectal Cancer Treated with Neoadjuvant Chemo-Radiotherapy. Biomedicines 2022; 10:biomedicines10123247. [PMID: 36552003 PMCID: PMC9775018 DOI: 10.3390/biomedicines10123247] [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: 10/25/2022] [Revised: 11/25/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neoadjuvant chemo-radiotherapy (nCRT) represents the standard of care for locally advanced rectal cancer (LARC); however, there exists no biomarker that can predict the cancer's response to treatment as less than 20% of patients experience pathological complete response (pCR). Ionizing radiations induce double strand breaks (DSBs) and trigger a DNA damage response (DDR) involving ATM, ATR, and the MRN complex (MRE11, Rad50, and NBS1). In this study, we performed an extensive mutational analysis of the genes involved in the DDR pathway in LARC patients who have undergone nCRT. METHODS 13 LARC patients with pCR and 11 LARC patients with partial response (pPR) were investigated using a NGS dedicated panel, designed for formalin-fixed paraffin-embedded (FFPE) samples, containing ATR, ATM, and MRE11-RAD50-NBN genes. The identified variants were classified according to guidelines' recommendations. RESULTS Eight non-benign variants, six of which were observed in 3 (23%) out of 13 pCR patients, were identified. In particular, a pCR patient carried out a pathogenetic frameshift mutation in exon 21 of the RAD50 gene. The two remaining non-benign missense variants were found in 2 (18%) out of 11 patients in the pPR group. CONCLUSIONS Our data show that the genes involved in the Homologous Recombination (HR) pathway are rarely mutated in LARC; however, given the identification of a missense mutation in RAD 50 in one case of pCR, it could be worth exploring its potential role as a biomarker in larger series.
Collapse
Affiliation(s)
- Andrea Montori
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Unit of Pathologic Morphological and Molecular Anatomy, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Mario Ferri
- Department of Medical-Surgical Sciences and Translational Medicine, “Sapienza” University of Rome, Piazzale Aldo Moro 5, 00189 Rome, Italy
- Unit of Gastrointestinal Surgery, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Annalisa Milano
- Unit of Oncology, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
| | | | - Maria Piane
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Unit of Medical Genetics and Advanced Cellular Diagnostic, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Unit of Pathologic Morphological and Molecular Anatomy, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
- Correspondence:
| |
Collapse
|
10
|
Rodrigues-Ferreira S, Nahmias C. Predictive biomarkers for personalized medicine in breast cancer. Cancer Lett 2022; 545:215828. [PMID: 35853538 DOI: 10.1016/j.canlet.2022.215828] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/04/2022] [Accepted: 07/10/2022] [Indexed: 12/14/2022]
Abstract
Breast cancer is one of the most frequent malignancies among women worldwide. Based on clinical and molecular features of breast tumors, patients are treated with chemotherapy, hormonal therapy and/or radiotherapy and more recently with immunotherapy or targeted therapy. These different therapeutic options have markedly improved patient outcomes. However, further improvement is needed to fight against resistance to treatment. In the rapidly growing area of research for personalized medicine, predictive biomarkers - which predict patient response to therapy - are essential tools to select the patients who are most likely to benefit from the treatment, with the aim to give the right therapy to the right patient and avoid unnecessary overtreatment. The search for predictive biomarkers is an active field of research that includes genomic, proteomic and/or machine learning approaches. In this review, we describe current strategies and innovative tools to identify, evaluate and validate new biomarkers. We also summarize current predictive biomarkers in breast cancer and discuss companion biomarkers of targeted therapy in the context of precision medicine.
Collapse
Affiliation(s)
- Sylvie Rodrigues-Ferreira
- Gustave Roussy Institute, INSERM U981, Prédicteurs moléculaires et nouvelles cibles en oncologie, Villejuif, France; LabEx LERMIT, Université Paris-Saclay, 92296 Châtenay-Malabry, France; Inovarion, 75005, Paris, France
| | - Clara Nahmias
- Gustave Roussy Institute, INSERM U981, Prédicteurs moléculaires et nouvelles cibles en oncologie, Villejuif, France; LabEx LERMIT, Université Paris-Saclay, 92296 Châtenay-Malabry, France.
| |
Collapse
|
11
|
Inalegwu A, Cuypers B, Claesen J, Janssen A, Coolkens A, Baatout S, Laukens K, De Vos WH, Quintens R. Fractionated irradiation of MCF7 breast cancer cells rewires a gene regulatory circuit towards a treatment-resistant stemness phenotype. Mol Oncol 2022; 16:3410-3435. [PMID: 35579852 PMCID: PMC9533694 DOI: 10.1002/1878-0261.13226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/07/2022] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
Radiotherapy is the standard of care for breast cancer. However, surviving radioresistant cells can repopulate following treatment and provoke relapse. Better understanding of the molecular mechanisms of radiation resistance may help improve treatment of radioresistant tumors. To emulate radiation therapy at the cellular level, we exposed MCF7 breast cancer cells to daily radiation doses of 2 Gy up to an accumulated dose of 20 Gy. Fractionally irradiated cells (FIR20) displayed increased clonogenic survival and population doubling time as compared to age-matched sham-irradiated cells and untreated parental MCF7 cells. RNA-sequencing revealed a core signature of 229 mRNAs and 7 circular RNAs of which the expression was significantly altered in FIR20 cells. Dysregulation of several top genes was mirrored at the protein level. The FIR20 cell transcriptome overlapped significantly with canonical radiation response signatures and demonstrated a remarkable commonality with radiation and endocrine therapy resistance expression profiles, suggesting crosstalk between both acquired resistance pathways, as indicated by reduced sensitivity to tamoxifen cytotoxicity of FIR20 cells. Using predictive analyses and functional enrichment, we identified a gene-regulatory network that promotes stemness and inflammatory signaling in FIR20 cells. We propose that these phenotypic traits render breast cancer cells more radioresistant but may at the same time serve as potential targets for combination therapies.
Collapse
Affiliation(s)
- Auchi Inalegwu
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium.,Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610, Antwerp, Belgium.,Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium.,Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610, Antwerp, Belgium
| | - Bart Cuypers
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | - Jürgen Claesen
- Department of Epidemiology and Data Science, Amsterdam UMC, VU, Amsterdam, Netherlands
| | - Ann Janssen
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| | - Amelie Coolkens
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | - Winnok H De Vos
- Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610, Antwerp, Belgium.,Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Antwerp, Belgium.,µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Roel Quintens
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| |
Collapse
|
12
|
Portner R, Bajaj A, Elumalai T, Huddart R, Murthy V, Nightingale H, Patel K, Sargos P, Song Y, Hoskin P, Choudhury A. A practical approach to bladder preservation with hypofractionated radiotherapy for localised muscle-invasive bladder cancer. Clin Transl Radiat Oncol 2021; 31:1-7. [PMID: 34466667 PMCID: PMC8385113 DOI: 10.1016/j.ctro.2021.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Bladder preservation with trimodality treatment (TMT) is an alternative strategy to radical cystectomy (RC) for the management of localised muscle invasive bladder cancer (MIBC). TMT comprises of transurethral resection of the bladder tumour (TURBT) followed by radiotherapy with concurrent radiosensitisation. TMT studies have shown neo-adjuvant chemotherapy with cisplatin-based regimens is often given to further improve survival outcomes. A hypofractionated radiotherapy regimen is preferable due to its non-inferiority in local control and late toxicities. Radiosensitisation can comprise concurrent chemotherapy (with gemcitabine, cisplatin or combination fluorouracil and mitomycin), CON (carbogen and nicotinomide) or hyperthermic treatment. Radiotherapy techniques are continuously improving and becoming more personalised. As the bladder is a mobile structure subject to volumetric changes from filling, an adaptive approach can optimise bladder coverage and reduce dose to normal tissue. Adaptive radiotherapy (ART) is an evolving field that aims to overcome this. Improved knowledge of tumour biology and advances in imaging techniques aims to further optimise and personalise treatment.
Collapse
Affiliation(s)
- R. Portner
- The Christie NHS Foundation Trust, Manchester, UK
| | - A. Bajaj
- Department of Radiation Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - T. Elumalai
- The Christie NHS Foundation Trust, Manchester, UK
| | - R. Huddart
- Royal Marsden NHS Foundation Trust, London, UK
- Institute of Cancer Research, UK
| | - V. Murthy
- Department of Radiation Oncology, ACTREC and Tata Memorial Hospital, Homi Bhabha National University, Mumbai, India
| | | | - K. Patel
- The Christie NHS Foundation Trust, Manchester, UK
| | - P. Sargos
- Department of Radiation Oncology, Institut Bergonié, F-33076 Bordeaux Cedex, France
| | - Y. Song
- The Christie NHS Foundation Trust, Manchester, UK
| | - P. Hoskin
- Mount Vernon Cancer Centre, Northwood, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - A. Choudhury
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Yan A, Hanna A, Wilson TG, Deraniyagala R, Krauss DJ, Grzywacz VP, Yan D, Wilson GD. Correlation between tumor voxel dose response matrix and tumor biomarker profile in patients with head and neck squamous cell carcinoma. Radiother Oncol 2021; 164:196-201. [PMID: 34619238 DOI: 10.1016/j.radonc.2021.09.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/13/2021] [Accepted: 09/24/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND We have developed a novel imaging analysis procedure that is highly predictive of local failure after chemoradiation in head and neck cancer. In this study we investigated whether any pretreatment biomarkers correlated with key imaging parameters. METHODS Pretreatment biopsy material was available for 28 patients entered into an institutional trial of adaptive radiotherapy in which FDG-PET images were collected weekly during treatment. The biopsies were immunohistochemically stained for CD44, EGFR, GLUT1, ALDH1, Ki-67 and p53 and quantified using image analysis. Expression levels were correlated with previously derived imaging parameters, the pretreatment SUVmax and the dose response matrix (DRM). RESULTS The different parameters of the SUVmax and DRM did not correlate with each other. We observed a positive and highly significant (p = 0.0088) correlation between CD44 expression and volume of tumor with a DRM greater than 0.8. We found no correlation between any DRM parameter and GLUT1, p53, Ki-67 and EGFR or ALDH1. GLUT1 expression did correlate with the maximum SUV0 and the volume of tumor with an SUV0 greater than 20. CONCLUSIONS The pretreatment SUVmax and DRM are independent imaging parameters that combine to predict local recurrence. The significant correlation between CD44 expression, a known cancer stem cell (CSC) marker, and volume of tumor with a DRM greater than 0.8 is consistent with concept that specific foci of cells are responsible for tumor recurrence and that CSCs may be randomly distributed in tumors in specific niches. Dose painting these small areas may lead to improved tumor control.
Collapse
Affiliation(s)
- Arthur Yan
- Department of Radiation Oncology, Beaumont Health, USA
| | - Alaa Hanna
- Department of Radiation Oncology, Beaumont Health, USA
| | | | | | | | | | - Di Yan
- Department of Radiation Oncology, Beaumont Health, USA
| | | |
Collapse
|
14
|
Batis N, Brooks JM, Payne K, Sharma N, Nankivell P, Mehanna H. Lack of predictive tools for conventional and targeted cancer therapy: Barriers to biomarker development and clinical translation. Adv Drug Deliv Rev 2021; 176:113854. [PMID: 34192550 PMCID: PMC8448142 DOI: 10.1016/j.addr.2021.113854] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/30/2022]
Abstract
Predictive tools, utilising biomarkers, aim to objectively assessthe potentialresponse toa particular clinical intervention in order to direct treatment.Conventional cancer therapy remains poorly served by predictive biomarkers, despite being the mainstay of treatment for most patients. In contrast, targeted therapy benefits from a clearly defined protein target for potential biomarker assessment. We discuss potential data sources of predictive biomarkers for conventional and targeted therapy, including patient clinical data andmulti-omicbiomarkers (genomic, transcriptomic and protein expression).Key examples, either clinically adopted or demonstrating promise for clinical translation, are highlighted. Following this, we provide an outline of potential barriers to predictive biomarker development; broadly discussing themes of approaches to translational research and study/trial design, and the impact of cellular and molecular tumor heterogeneity. Future avenues of research are also highlighted.
Collapse
Affiliation(s)
- Nikolaos Batis
- Institute of Head and Neck Studies and Education (InHANSE), Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.
| | - Jill M Brooks
- Institute of Head and Neck Studies and Education (InHANSE), Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Karl Payne
- Institute of Head and Neck Studies and Education (InHANSE), Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neil Sharma
- Institute of Head and Neck Studies and Education (InHANSE), Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Head and Neck Surgery, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Paul Nankivell
- Institute of Head and Neck Studies and Education (InHANSE), Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Head and Neck Surgery, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Hisham Mehanna
- Institute of Head and Neck Studies and Education (InHANSE), Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Head and Neck Surgery, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom.
| |
Collapse
|
15
|
Jang BS, Chang JH, Jeon SH, Song MG, Lee KH, Im SA, Kim JI, Kim TY, Chie EK. Radiation Response Prediction Model based on Integrated Clinical and Genomic Data Analysis. Cancer Res Treat 2021; 54:383-395. [PMID: 34425668 PMCID: PMC9016297 DOI: 10.4143/crt.2021.759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/23/2021] [Indexed: 11/21/2022] Open
Abstract
Purpose The value of the genomic profiling by targeted gene-sequencing on radiation therapy response prediction was evaluated through integrated analysis including clinical information. Radiation response prediction model was constructed based on the analyzed findings. Materials and Methods Patients who had the tumor sequenced using institutional cancer panel after informed consent and received radiotherapy for the measurable disease served as the target cohort. Patients with irradiated tumor locally controlled for more than 6 months after radiotherapy were defined as the durable local control (DLC) group, otherwise, non-durable local control (NDLC) group. Significant genomic factors and domain knowledge were used to develop the Bayesian Network model to predict radiotherapy response. Results Altogether, 88 patients were collected for analysis. Of those, 41 (43.6%) and 47 (54.4%) patients were classified as the NDLC and DLC group, respectively. Somatic mutations of NOTCH2 and BCL were enriched in the NDLC group, whereas, mutations of CHEK2, MSH2, and NOTCH1 were more frequently found in the DLC group. Altered DNA repair pathway was associated with better local failure-free survival (HR 0.40, 95%CI 0.19-0.86, p=0.014). Smoking somatic signature was found more frequently in the DLC group. AUC of the Bayesian Network model predicting probability of 6-month local control was 0.83. Conclusion Durable radiation response was associated with alterations of DNA repair pathway and smoking somatic signature. Bayesian network model could provide helpful insights for high precision radiotherapy. However, these findings should be verified in prospective cohort for further individualization.
Collapse
Affiliation(s)
- Bum-Sup Jang
- Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji-Hyun Chang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Hyuck Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Myung Geun Song
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Tae-You Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Eui Kyu Chie
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
16
|
Manem VSK. Development and validation of genomic predictors of radiation sensitivity using preclinical data. BMC Cancer 2021; 21:937. [PMID: 34416855 PMCID: PMC8377977 DOI: 10.1186/s12885-021-08652-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/25/2021] [Indexed: 11/19/2022] Open
Abstract
Background Radiation therapy is among the most effective and commonly used therapeutic modalities of cancer treatments in current clinical practice. The fundamental paradigm that has guided radiotherapeutic regimens are ‘one-size-fits-all’, which are not in line with the dogma of precision medicine. While there were efforts to build radioresponse signatures using OMICS data, their ability to accurately predict in patients is still limited. Methods We proposed to integrate two large-scale radiogenomics datasets consisting of 511 with 23 tissues and 60 cancer cell lines with 9 tissues to build and validate radiation response biomarkers. We used intrinsic radiation sensitivity, i.e., surviving fraction of cells (SF2) as the radiation response indicator. Gene set enrichment analysis was used to examine the biological determinants driving SF2. Using SF2 as a continuous variable, we used five different approaches, univariate, rank gene ensemble, rank gene multivariate, mRMR and elasticNet to build genomic predictors of radiation response through a cross-validation framework. Results Through the pathway analysis, we found 159 pathways to be statistically significant, out of which 54 and 105 were positively and negatively enriched with SF2. More importantly, we found cell cycle and repair pathways to be enriched with SF2, which are inline with the fundamental aspects of radiation biology. With regards to the radiation response gene signature, we found that all multivariate models outperformed the univariate model with a ranking based approach performing well compared to other models, indicating complex biological processes underpinning radiation response. Conclusion To summarize, we found biological processes underpinning SF2 and systematically compared different machine learning approaches to develop and validate predictors of radiation response. With more patient data available in the future, the clinical value of these biomarkers can be assessed that would allow for personalization of radiotherapy.
Collapse
Affiliation(s)
- Venkata S K Manem
- Quebec Heart & Lung Institute Research Center, Quebec City, Quebec, G1V 4G5, Canada. .,Faculty of Pharmacy, Laval University, Quebec City, Quebec, G1V 0A6, Canada.
| |
Collapse
|
17
|
A Novel Approach for the Discovery of Biomarkers of Radiotherapy Response in Breast Cancer. J Pers Med 2021; 11:jpm11080796. [PMID: 34442440 PMCID: PMC8399231 DOI: 10.3390/jpm11080796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 01/08/2023] Open
Abstract
Radiotherapy (RT) is an important treatment modality for the local control of breast cancer (BC). Unfortunately, not all patients that receive RT will obtain a therapeutic benefit, as cancer cells that either possess intrinsic radioresistance or develop resistance during treatment can reduce its efficacy. For RT treatment regimens to become personalised, there is a need to identify biomarkers that can predict and/or monitor a tumour's response to radiation. Here we describe a novel method to identify such biomarkers. Liquid chromatography-mass spectrometry (LC-MS) was used on conditioned media (CM) samples from a radiosensitive oestrogen receptor positive (ER+) BC cell line (MCF-7) to identify cancer-secreted biomarkers which reflected a response to radiation. A total of 33 radiation-induced secreted proteins that had higher (up to 12-fold) secretion levels at 24 h post-2 Gy radiation were identified. Secretomic results were combined with whole-transcriptome gene expression experiments, using both radiosensitive and radioresistant cells, to identify a signature related to intrinsic radiosensitivity. Gene expression analysis assessing the levels of the 33 proteins showed that 5 (YBX3, EIF4EBP2, DKK1, GNPNAT1 and TK1) had higher expression levels in the radiosensitive cells compared to their radioresistant derivatives; 3 of these proteins (DKK1, GNPNAT1 and TK1) underwent in-lab and initial clinical validation. Western blot analysis using CM samples from cell lines confirmed a significant increase in the release of each candidate biomarker from radiosensitive cells 24 h after treatment with a 2 Gy dose of radiation; no significant increase in secretion was observed in the radioresistant cells after radiation. Immunohistochemistry showed that higher intracellular protein levels of the biomarkers were associated with greater radiosensitivity. Intracellular levels were further assessed in pre-treatment biopsy tissues from patients diagnosed with ER+ BC that were subsequently treated with breast-conserving surgery and RT. High DKK1 and GNPNAT1 intracellular levels were associated with significantly increased recurrence-free survival times, indicating that these two candidate biomarkers have the potential to predict sensitivity to RT. We suggest that the methods highlighted in this study could be utilised for the identification of biomarkers that may have a potential clinical role in personalising and optimising RT dosing regimens, whilst limiting the administration of RT to patients who are unlikely to benefit.
Collapse
|
18
|
Angiogenesis and immune checkpoint dual blockade in combination with radiotherapy for treatment of solid cancers: opportunities and challenges. Oncogenesis 2021; 10:47. [PMID: 34247198 PMCID: PMC8272720 DOI: 10.1038/s41389-021-00335-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/02/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Several immune checkpoint blockades (ICBs) capable of overcoming the immunosuppressive roles of the tumor immune microenvironment have been approved by the US Food and Drug Administration as front-line treatments of various tumor types. However, due to the considerable heterogeneity of solid tumor cells, inhibiting one target will only influence a portion of the tumor cells. One way to enhance the tumor-killing efficiency is to develop a multiagent therapeutic strategy targeting different aspects of tumor biology and the microenvironment to provide the maximal clinical benefit for patients with late-stage disease. One such strategy is the administration of anti-PD1, an ICB, in combination with the humanized monoclonal antibody bevacizumab, an anti-angiogenic therapy, to patients with recurrent/metastatic malignancies, including hepatocellular carcinoma, metastatic renal cell carcinoma, non-small cell lung cancer, and uterine cancer. Radiotherapy (RT), a critical component of solid cancer management, has the capacity to prime the immune system for an adaptive antitumor response. Here, we present an overview of the most recent published data in preclinical and clinical studies elucidating that RT could further potentiate the antitumor effects of immune checkpoint and angiogenesis dual blockade. In addition, we explore opportunities of triple combinational treatment, as well as discuss the challenges of validating biomarkers and the management of associated toxicity.
Collapse
|
19
|
MRE11 as a molecular signature and therapeutic target for cancer treatment with radiotherapy. Cancer Lett 2021; 514:1-11. [PMID: 34022282 DOI: 10.1016/j.canlet.2021.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 01/02/2023]
Abstract
MRE11, the core of the MRE11/RAD50/NBS1 complex, is one of key DNA damage response proteins. Increasing evidence suggests that its expression in cancer cells is critical to developing radioresistance; as such, MRE11 is an emerging marker for targeted radiosensitization strategies. Elevated MRE11 in tumor tissues has been associated with poor survival in patients undergoing radiotherapy, although in some cancer types, the opposite has been noted. The recent discovery of ionizing radiation-induced truncation of MRE11, which decreases its efficacy, may explain some of these paradoxical findings. The progress of research on the biological modulation of MRE11 expression is also discussed, with the potential application of small molecule or large molecule inhibitors of MRE11 for enhancing radiosensitivity. Current research has further highlighted both nuclease and non-nuclease activities of MRE11 in cancer cells treated with ionizing radiation, and differentiation between these is essential to verify the targeting effects of radiosensitizing agents. These updates clarify our understanding of how MRE11 expression may be utilized in future stratification of cancer patients for radiotherapy, and how it may be leveraged in shaping novel radiosensitization strategies.
Collapse
|
20
|
Vaes RDW, Hendriks LEL, Vooijs M, De Ruysscher D. Biomarkers of Radiotherapy-Induced Immunogenic Cell Death. Cells 2021; 10:cells10040930. [PMID: 33920544 PMCID: PMC8073519 DOI: 10.3390/cells10040930] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 12/20/2022] Open
Abstract
Radiation therapy (RT) can induce an immunogenic variant of regulated cell death that can initiate clinically relevant tumor-targeting immune responses. Immunogenic cell death (ICD) is accompanied by the exposure and release of damage-associated molecular patterns (DAMPs), chemokine release, and stimulation of type I interferon (IFN-I) responses. In recent years, intensive research has unraveled major mechanistic aspects of RT-induced ICD and has resulted in the identification of immunogenic factors that are released by irradiated tumor cells. However, so far, only a limited number of studies have searched for potential biomarkers that can be used to predict if irradiated tumor cells undergo ICD that can elicit an effective immunogenic anti-tumor response. In this article, we summarize the available literature on potential biomarkers of RT-induced ICD that have been evaluated in cancer patients. Additionally, we discuss the clinical relevance of these findings and important aspects that should be considered in future studies.
Collapse
Affiliation(s)
- Rianne D. W. Vaes
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. 616, 6200 MD Maastricht, The Netherlands; (M.V.); (D.D.R.)
- Correspondence: ; Tel.: +31-(0)43-388-1585
| | - Lizza E. L. Hendriks
- Department of Pulmonary Diseases, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. 616, 6200 MD Maastricht, The Netherlands;
| | - Marc Vooijs
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. 616, 6200 MD Maastricht, The Netherlands; (M.V.); (D.D.R.)
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. 616, 6200 MD Maastricht, The Netherlands; (M.V.); (D.D.R.)
| |
Collapse
|
21
|
Silina L, Maksut F, Bernard-Pierrot I, Radvanyi F, Créhange G, Mégnin-Chanet F, Verrelle P. Review of Experimental Studies to Improve Radiotherapy Response in Bladder Cancer: Comments and Perspectives. Cancers (Basel) 2020; 13:E87. [PMID: 33396795 PMCID: PMC7795454 DOI: 10.3390/cancers13010087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 01/04/2023] Open
Abstract
Bladder cancer is among the top ten most common cancer types in the world. Around 25% of all cases are muscle-invasive bladder cancer, for which the gold standard treatment in the absence of metastasis is the cystectomy. In recent years, trimodality treatment associating maximal transurethral resection and radiotherapy combined with concurrent chemotherapy is increasingly used as an organ-preserving alternative. However, the use of this treatment is still limited by the lack of biomarkers predicting tumour response and by a lack of targeted radiosensitising drugs that can improve the therapeutic index, especially by limiting side effects such as bladder fibrosis. In order to improve the bladder-preserving treatment, experimental studies addressing these main issues ought to be considered (both in vitro and in vivo studies). Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for systematic reviews, we conducted a literature search in PubMed on experimental studies investigating how to improve bladder cancer radiotherapy with different radiosensitising agents using a comprehensive search string. We made comments on experimental model selection, experimental design and results, formulating the gaps of knowledge still existing: such as the lack of reliable predictive biomarkers of tumour response to chemoradiation according to the molecular tumour subtype and lack of efficient radiosensitising agents specifically targeting bladder tumour cells. We provided guidance to improve forthcoming studies, such as taking into account molecular characteristics of the preclinical models and highlighted the value of using patient-derived xenografts as well as syngeneic models. Finally, this review could be a useful tool to set up new radiation-based combined treatments with an improved therapeutic index that is needed for bladder preservation.
Collapse
Affiliation(s)
- Linda Silina
- French League Against Cancer Team, CNRS UMR144, Curie Institute and PSL Research University, 75005 Paris, France; (I.B.-P.); (F.R.)
- CNRS UMR 9187, INSERM U1196, Curie Institute, PSL Research University and Paris-Saclay University, Rue H. Becquerel, 91405 Orsay, France; (F.M.); (F.M.-C.)
| | - Fatlinda Maksut
- CNRS UMR 9187, INSERM U1196, Curie Institute, PSL Research University and Paris-Saclay University, Rue H. Becquerel, 91405 Orsay, France; (F.M.); (F.M.-C.)
| | - Isabelle Bernard-Pierrot
- French League Against Cancer Team, CNRS UMR144, Curie Institute and PSL Research University, 75005 Paris, France; (I.B.-P.); (F.R.)
| | - François Radvanyi
- French League Against Cancer Team, CNRS UMR144, Curie Institute and PSL Research University, 75005 Paris, France; (I.B.-P.); (F.R.)
| | - Gilles Créhange
- Radiation Oncology Department, Curie Institute, 75005 Paris, France;
| | - Frédérique Mégnin-Chanet
- CNRS UMR 9187, INSERM U1196, Curie Institute, PSL Research University and Paris-Saclay University, Rue H. Becquerel, 91405 Orsay, France; (F.M.); (F.M.-C.)
| | - Pierre Verrelle
- CNRS UMR 9187, INSERM U1196, Curie Institute, PSL Research University and Paris-Saclay University, Rue H. Becquerel, 91405 Orsay, France; (F.M.); (F.M.-C.)
- Radiation Oncology Department, Curie Institute, 75005 Paris, France;
- Clermont Auvergne University, 63000 Clermont-Ferrand, France
| |
Collapse
|
22
|
Le Compte M, Komen N, Joye I, Peeters M, Prenen H, Smits E, Deben C, de Maat M. Patient-derived organoids as individual patient models for chemoradiation response prediction in gastrointestinal malignancies. Crit Rev Oncol Hematol 2020; 157:103190. [PMID: 33310278 DOI: 10.1016/j.critrevonc.2020.103190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/11/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Chemoradiotherapy (CRT) is an important treatment modality for specific gastrointestinal (GI) cancers, as it has been shown to improve clinical outcomes. Recent developments in the neoadjuvant setting such as wait-and-see strategies for rectal as well as for esophageal cancers have even proven that CRT might be an effective organ-sparing treatment. However, due to molecular heterogeneity, only a subset of patients will show a complete response to CRT, which addresses the need for an individualized treatment approach. In recent years, the demand for more physiologically relevant predictive in vitro models has fostered the development of patient-derived tumor organoids. In this review, we describe the current treatment options for patients with GI cancers who are treated with (neo)adjuvant CRT. Furthermore, we provide an in-depth discussion of the organoid technology in the context of predicting CRT response for GI cancers as well as possible challenges for clinical implementation.
Collapse
Affiliation(s)
- Maxim Le Compte
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Building T, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Niels Komen
- Department of Abdominal Surgery, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium; Antwerp Surgical Training, Anatomy and Research Centre (ASTRAC), University of Antwerp, Campus Drie Eiken, Building T, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Ines Joye
- Department of Radiation Oncology, Iridium Kankernetwerk, Oosterveldlaan 22, Wilrijk, B-2610, Antwerp, Belgium; Department of Molecular Imaging, Pathology, Radiotherapy and Oncology (MIPRO), Faculty of Medicine and Health Sciences, University of Antwerp, Campus Drie Eiken, Building S, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Building T, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium; Department of Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Hans Prenen
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Building T, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium; Department of Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Building T, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Christophe Deben
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Campus Drie Eiken, Building T, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium.
| | - Michiel de Maat
- Department of Abdominal Surgery, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium; Antwerp Surgical Training, Anatomy and Research Centre (ASTRAC), University of Antwerp, Campus Drie Eiken, Building T, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium.
| |
Collapse
|
23
|
Yan M, Wang W. Radiomic Analysis of CT Predicts Tumor Response in Human Lung Cancer with Radiotherapy. J Digit Imaging 2020; 33:1401-1403. [PMID: 33025167 PMCID: PMC7728862 DOI: 10.1007/s10278-020-00385-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Radiomics features can be positioned to monitor changes throughout treatment. In this study, we evaluated machine learning for predicting tumor response by analyzing CT images of lung cancer patients treated with radiotherapy. EXPERIMENTAL DESIGN For this retrospective study, screening or standard diagnostic CT images were collected for 100 patients (mean age, 67 years; range, 55-82 years; 64 men [mean age, 68 years; range, 55-82 years] and 36 women [mean age, 65 years; range, 60-72 years]) from two institutions between 2013 and 2017. Radiomics analysis was available for each patient. Features were pruned to train machine learning classifiers with 50 patients, then trained in the test dataset. RESULT A support vector machine classifier with 2 radiomic features (flatness and coefficient of variation) achieved an area under the receiver operating characteristic curve (AUC) of 0.91 on the test set. CONCLUSION The 2 radiomic features, flatness, and coefficient of variation, from the volume of interest of lung tumor, can be the biomarkers for predicting tumor response at CT.
Collapse
Affiliation(s)
- Mengmeng Yan
- Urban Vocational College of Sichuan, Chengdu, China.,Sichuan Cancer Hospital & Institute, Chengdu, China
| | - Weidong Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Chengdu, 610041, China. .,Radiation Oncology, Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
24
|
Lee YH, Tai D, Yip C, Choo SP, Chew V. Combinational Immunotherapy for Hepatocellular Carcinoma: Radiotherapy, Immune Checkpoint Blockade and Beyond. Front Immunol 2020; 11:568759. [PMID: 33117354 PMCID: PMC7561368 DOI: 10.3389/fimmu.2020.568759] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
The systemic treatment landscape for advanced hepatocellular carcinoma (HCC) has experienced tremendous paradigm shift towards targeting tumor microenvironment (TME) following recent trials utilizing immune checkpoint blockade (ICB). However, limited success of ICB as monotherapy mandates the evaluation of combination strategies incorporating immunotherapy for improved clinical efficacy. Radiotherapy (RT) is an integral component in treatment of solid cancers, including HCC. Radiation mediates localized tumor killing and TME modification, thereby potentiating the action of ICB. Several preclinical and clinical studies have explored the efficacy of combining RT and ICB in HCC with promising outcomes. Greater efforts are required in discovery and understanding of novel combination strategies to maximize clinical benefit with tolerable adverse effects. This current review provides a comprehensive assessment of RT and ICB in HCC, their respective impact on TME, the rationale for their synergistic combination, as well as the current potential biomarkers available to predict clinical response. We also speculate on novel future strategies to further enhance the efficacy of this combination.
Collapse
Affiliation(s)
- Yun Hua Lee
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, Singapore
| | - David Tai
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Connie Yip
- Division of Radiation Oncology, National Cancer Centre, Singapore, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore.,Curie Oncology, Mount Elizabeth Novena Specialist Centre, Singapore, Singapore
| | - Valerie Chew
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, Singapore
| |
Collapse
|
25
|
Sjöström M, Veenstra C, Holmberg E, Karlsson P, Killander F, Malmström P, Niméus E, Fernö M, Stål O. Expression of HGF, pMet, and pAkt is related to benefit of radiotherapy after breast-conserving surgery: a long-term follow-up of the SweBCG91-RT randomised trial. Mol Oncol 2020; 14:2713-2726. [PMID: 32946618 PMCID: PMC7607179 DOI: 10.1002/1878-0261.12803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/19/2020] [Accepted: 09/11/2020] [Indexed: 01/15/2023] Open
Abstract
Experimental studies suggest that hepatocyte growth factor (HGF) and its transmembrane tyrosine kinase receptor, Met, in part also relying on Akt kinase activity, mediate radioresistance. We investigated the importance of these biomarkers for the risk of ipsilateral breast tumour recurrence (IBTR) after adjuvant radiotherapy (RT) in primary breast cancer. HGF, phosphorylated Met (pMet) and phosphorylated Akt (pAkt) were evaluated immunohistochemically on tissue microarrays from 1004 patients in the SweBCG91‐RT trial, which randomly assigned patients to breast‐conserving therapy, with or without adjuvant RT. HGF was evaluated in the stroma (HGFstr); pMet in the membrane (pMetmem); HGF, pMet and pAkt in the cytoplasm (HGFcyt, pMetcyt, pAktcyt); and pAkt in the nucleus (pAktnuc). The prognostic and treatment predictive effects were evaluated to primary endpoint IBTR as first event during the first 5 years. Patients with tumours expressing low levels of HGFcyt and pMetcyt and high levels of pAktnuc derived a larger benefit from RT [hazard ratio (HR): 0.11 (0.037–0.30), 0.066 (0.016–0.28) and 0.094 (0.028–0.31), respectively] compared to patients with high expression of HGFcyt and pMetcyt, and low pAktnuc [HR: 0.36 (0.19–0.67), 0.35 (0.20–0.64) and 0.47 (0.32–0.71), respectively; interaction analyses: P = 0.052, 0.035 and 0.013, respectively]. These differences remained in multivariable analysis when adjusting for patient age, tumour size, histological grade, St Gallen subtype and systemic treatment (interaction analysis, P‐values: 0.085, 0.027, and 0.023, respectively). This study suggests that patients with immunohistochemically low HGFcyt, low pMetcyt and high pAktnuc may derive an increased benefit from RT after breast‐conserving surgery concerning the risk of developing IBTR.
Collapse
Affiliation(s)
- Martin Sjöström
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Cynthia Veenstra
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Oncology, Linköping University, Linköping, Sweden
| | - Erik Holmberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Fredrika Killander
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Per Malmström
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Emma Niméus
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.,Division of Surgery, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Mårten Fernö
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Olle Stål
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Oncology, Linköping University, Linköping, Sweden
| |
Collapse
|
26
|
Biolatti V, Negrin L, Bellora N, Ibañez IL. High-throughput meta-analysis and validation of differentially expressed genes as potential biomarkers of ionizing radiation-response. Radiother Oncol 2020; 154:21-28. [PMID: 32931891 DOI: 10.1016/j.radonc.2020.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 08/20/2020] [Accepted: 09/06/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND AND PURPOSE The high-throughput analysis of gene expression in ionizing radiation (IR)-exposed human peripheral white blood cells (WBC) has emerged as a novel method for biodosimetry markers detection. We aimed to detect IR-exposure differential expressed genes (DEGs) as potential predictive biomarkers for biodosimetry and radioinduced-response. MATERIALS AND METHODS We performed a meta-analysis of raw data from public microarrays of ex vivo low linear energy transfer-irradiated human peripheral WBC. Functional enrichment and transcription factors (TF) detection from resulting DEGs were assessed. Six selected DEGs among studies were validated by qRT-PCR on mRNA from human peripheral blood samples from nine healthy human donors 24 h after ex vivo X-rays-irradiation. RESULTS We identified 275 DEGs after IR-exposure (parameters: |lfc| ≥ 0.7, q value <0.05), enriched in processes such as regulation after IR-exposure, DNA damage checkpoint, signal transduction by p53 and mitotic cell cycle checkpoint. Among these DEGs, DRAM1, NUDT15, PCNA, PLK2 and TIGAR were selected for qRT-PCR validation. Their expression levels significantly increased at 1-4 Gy respect to non-irradiated controls. Particularly, PCNA increased dose dependently. Curiously, TCF4 (Entrez Gene: 6925), detected as overrepresented TF in the radioinduced DEGs set, significantly decreased post-irradiation. CONCLUSION These six DEGs show potential to be proposed as candidates for IR-exposure biomarkers, considering their observed molecular radioinduced-response. Among them, TCF4, bioinformatically detected, was validated herein as an IR-responsive gene.
Collapse
Affiliation(s)
- Vanesa Biolatti
- National Atomic Energy Commission (CNEA), Bariloche Nuclear Medicine and Radiotherapy Integral Center - Institute of Nuclear Technologies for Health Foundation (INTECNUS); Laboratory of Radiobiology and Biodosimetry, S.C. de Bariloche, Argentina.
| | - Lara Negrin
- National Atomic Energy Commission (CNEA), Bariloche Nuclear Medicine and Radiotherapy Integral Center - Institute of Nuclear Technologies for Health Foundation (INTECNUS); Laboratory of Radiobiology and Biodosimetry, S.C. de Bariloche, Argentina.
| | - Nicolás Bellora
- National Scientific and Technical Research Council (CONICET), Scientific Technical Center CONICET - North Patagonia, Patagonian Andean Institute of Biological and Geo-Environmental Technologies (IPATEC), S.C. de Bariloche, Argentina.
| | - Irene L Ibañez
- National Scientific and Technical Research Council (CONICET), Institute of Nanocience and Nanotechnology (INN), Constituyentes Node (C1425FQB), CABA, Argentina; National Atomic Energy Commission (CNEA), Constituyentes Atomic Center, Research and Applications Management, Buenos Aires, Argentina.
| |
Collapse
|
27
|
Reuvers TGA, Kanaar R, Nonnekens J. DNA Damage-Inducing Anticancer Therapies: From Global to Precision Damage. Cancers (Basel) 2020; 12:E2098. [PMID: 32731592 PMCID: PMC7463878 DOI: 10.3390/cancers12082098] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/24/2020] [Accepted: 07/26/2020] [Indexed: 12/11/2022] Open
Abstract
DNA damage-inducing therapies are of tremendous value for cancer treatment and function by the direct or indirect formation of DNA lesions and subsequent inhibition of cellular proliferation. Of central importance in the cellular response to therapy-induced DNA damage is the DNA damage response (DDR), a protein network guiding both DNA damage repair and the induction of cancer-eradicating mechanisms such as apoptosis. A detailed understanding of DNA damage induction and the DDR has greatly improved our knowledge of the classical DNA damage-inducing therapies, radiotherapy and cytotoxic chemotherapy, and has paved the way for rational improvement of these treatments. Moreover, compounds targeting specific DDR proteins, selectively impairing DNA damage repair in cancer cells, form a promising novel therapy class that is now entering the clinic. In this review, we give an overview of the current state and ongoing developments, and discuss potential avenues for improvement for DNA damage-inducing therapies, with a central focus on the role of the DDR in therapy response, toxicity and resistance. Furthermore, we describe the relevance of using combination regimens containing DNA damage-inducing therapies and how they can be utilized to potentiate other anticancer strategies such as immunotherapy.
Collapse
Affiliation(s)
- Thom G. A. Reuvers
- Department of Molecular Genetics, Erasmus MC, Dr. Molenwaterplein 40, 3015 GD Rotterdam, The Netherlands; (T.G.A.R.); (R.K.)
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molenwaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Roland Kanaar
- Department of Molecular Genetics, Erasmus MC, Dr. Molenwaterplein 40, 3015 GD Rotterdam, The Netherlands; (T.G.A.R.); (R.K.)
- Oncode Institute, Office Jaarbeurs Innovation Mile (JIM), Jaarbeursplein 6, 3561 AL Utrecht, The Netherlands
| | - Julie Nonnekens
- Department of Molecular Genetics, Erasmus MC, Dr. Molenwaterplein 40, 3015 GD Rotterdam, The Netherlands; (T.G.A.R.); (R.K.)
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molenwaterplein 40, 3015 GD Rotterdam, The Netherlands
- Oncode Institute, Office Jaarbeurs Innovation Mile (JIM), Jaarbeursplein 6, 3561 AL Utrecht, The Netherlands
| |
Collapse
|
28
|
Masoudi-Khoram N, Abdolmaleki P, Hosseinkhan N, Nikoofar A, Mowla SJ, Monfared H, Baldassarre G. Differential miRNAs expression pattern of irradiated breast cancer cell lines is correlated with radiation sensitivity. Sci Rep 2020; 10:9054. [PMID: 32493932 PMCID: PMC7270150 DOI: 10.1038/s41598-020-65680-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/07/2020] [Indexed: 02/08/2023] Open
Abstract
Radiotherapy is a fundamental step in the treatment of breast cancer patients. The treatment efficiency is however reduced by the possible onset of radiation resistance. In order to develop the effective treatment approach, it is important to understand molecular basis of radiosensitivity in breast cancer. The purpose of the present study was to investigate different radiation response of breast cancer cell lines, and find out if this response may be related to change in the microRNAs expression profile. MDA-MB-231 and T47D cells were subjected to different doses of radiation, then MTT and clonogenic assays were performed to assess radiation sensitivity. Cytofluorometric and western blot analysis were performed to gain insight into cell cycle distribution and protein expression. MicroRNA sequencing and bioinformatics prediction methods were used to identify the difference in microRNAs expression between two breast cancer cells and the related genes and pathways. T47D cells were more sensitive to radiation respect to MDA-MB-231 cells as demonstrated by a remarkable G2 cell cycle arrest followed by a greater reduction in cell viability and colony forming ability. Accordingly, T47D cells showed higher increase in the phosphorylation of ATM, TP53 and CDK1 (markers of radiation response) and faster and more pronounced increase in RAD51 and γH2AX expression (markers of DNA damage), when compared to MDA-MB-231 cells. The two cell lines had different microRNAs expression profiles with a confirmed significant differential expression of miR-16-5p, which targets cell cycle related genes and predicts longer overall survival of breast cancer patients, as determined by bioinformatics analysis. These results suggest a possible role for miR-16-5p as radiation sensitizing microRNA and as prognostic/predictive biomarker in breast cancer.
Collapse
Affiliation(s)
- Nastaran Masoudi-Khoram
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Nazanin Hosseinkhan
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Alireza Nikoofar
- Department of Radiotherapy, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Seyed Javad Mowla
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamideh Monfared
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Gustavo Baldassarre
- Division of Experimental Oncology 2, Department of Translational Research, CRO, National Cancer Institute, Aviano, Italy
| |
Collapse
|
29
|
Joseph N, Kirkby NF, Hoskin PJ, West CML, Choudhury A, Dale RG. Radiobiologically derived biphasic fractionation schemes to overcome the effects of tumour hypoxia. Br J Radiol 2020; 93:20190250. [PMID: 32462907 DOI: 10.1259/bjr.20190250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE As a fractionated course of radiotherapy proceeds tumour shrinkage leads to resolution of hypoxia and the initiation of accelerated proliferation of radioresistant cancer cells with better repair capacity. We hypothesise that, in tumours with significant hypoxia, improved tumour control could be achieved with biphasic fractionation schedules that either use acceleration after 3-4 weeks of conventional radiotherapy or deliver a higher proportional dose towards the end of a course of treatment. We conducted a modelling study based on the concept of biological effective dose (BED) comparing such novel regimens with conventional fractionation. METHODS The comparator conventional fractionation schedule 70 Gy in 35 fractions delivered over 7 weeks was tested against the following novel regimens, both of which were designed to be isoeffective in terms of late normal tissue toxicity.40 Gy in 20 fractions over 4 weeks followed by 22.32 Gy in 6 consecutive daily fractions (delayed acceleration)30.4 Gy in 27 fractions over 4 weeks followed by 40 Gy in 15 fractions over 3 weeks (temporal dose redistribution)The delayed acceleration regimen is exactly identical to that of the comparator schedule over the first 28 days and the BED gains with the novel schedule are achieved during the second phase of treatment when reoxygenation is complete. For the temporal redistribution regimen, it was assumed that the reoxygenation fraction progressively increases during the first 4 weeks of treatment and an iterative approach was used to calculate the final tumour BED for varying hypoxic fractions. RESULTS Novel fractionation with delayed acceleration or temporal fractionation results in tumour BED gains equivalent to 3.5-8 Gy when delivered in 2 Gy fractions. CONCLUSION In hypoxic tumours, novel fractionation strategies result in significantly higher tumour BED in comparison to conventional fractionation. ADVANCES IN KNOWLEDGE We demonstrate that novel biphasic fractionation regimens could overcome the effects of tumour hypoxia resulting in biological dose escalation.
Collapse
Affiliation(s)
- Nuradh Joseph
- Ministry of Health, Colombo, Sri Lanka.,Sri Lanka Cancer Research Group, Maharagama, Sri Lanka
| | - Norman F Kirkby
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cancer Centre, Mount Vernon Hospital, Northwood, Middlesex, UK
| | - Catharine M L West
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ananya Choudhury
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Roger G Dale
- Faculty of Medicine, Imperial College, London, UK
| |
Collapse
|
30
|
Meehan J, Gray M, Martínez-Pérez C, Kay C, Pang LY, Fraser JA, Poole AV, Kunkler IH, Langdon SP, Argyle D, Turnbull AK. Precision Medicine and the Role of Biomarkers of Radiotherapy Response in Breast Cancer. Front Oncol 2020; 10:628. [PMID: 32391281 PMCID: PMC7193869 DOI: 10.3389/fonc.2020.00628] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/06/2020] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy remains an important treatment modality in nearly two thirds of all cancers, including the primary curative or palliative treatment of breast cancer. Unfortunately, largely due to tumor heterogeneity, tumor radiotherapy response rates can vary significantly, even between patients diagnosed with the same tumor type. Although in recent years significant technological advances have been made in the way radiation can be precisely delivered to tumors, it is proving more difficult to personalize radiotherapy regimens based on cancer biology. Biomarkers that provide prognostic or predictive information regarding a tumor's intrinsic radiosensitivity or its response to treatment could prove valuable in helping to personalize radiation dosing, enabling clinicians to make decisions between different treatment options whilst avoiding radiation-induced toxicity in patients unlikely to gain therapeutic benefit. Studies have investigated numerous ways in which both patient and tumor radiosensitivities can be assessed. Tumor molecular profiling has been used to develop radiosensitivity gene signatures, while the assessment of specific intracellular or secreted proteins, including circulating tumor cells, exosomes and DNA, has been performed to identify prognostic or predictive biomarkers of radiation response. Finally, the investigation of biomarkers related to radiation-induced toxicity could provide another means by which radiotherapy could become personalized. In this review, we discuss studies that have used these methods to identify or develop prognostic/predictive signatures of radiosensitivity, and how such assays could be used in the future as a means of providing personalized radiotherapy.
Collapse
Affiliation(s)
- James Meehan
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark Gray
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos Martínez-Pérez
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Charlene Kay
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Lisa Y Pang
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jennifer A Fraser
- School of Applied Science, Sighthill Campus, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Amy V Poole
- School of Applied Science, Sighthill Campus, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - David Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Arran K Turnbull
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
31
|
Ho V, Chung L, Singh A, Lea V, Abubakar A, Lim SH, Chua W, Ng W, Lee M, Roberts TL, de Souza P, Lee CS. Aberrant Expression of RAD52, Its Prognostic Impact in Rectal Cancer and Association with Poor Survival of Patients. Int J Mol Sci 2020; 21:ijms21051768. [PMID: 32143539 PMCID: PMC7084626 DOI: 10.3390/ijms21051768] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/29/2020] [Accepted: 03/01/2020] [Indexed: 12/18/2022] Open
Abstract
The DNA damage response enables cells to survive and maintain genome integrity. RAD52 is a DNA-binding protein involved in the homologous recombination in DNA repair, and is important for the maintenance of tumour genome integrity. We investigated possible correlations between RAD52 expression and cancer survival and response to preoperative radiotherapy. RAD52 expression was examined in tumour samples from 179 patients who underwent surgery for rectal cancer, including a sub-cohort of 40 patients who were treated with neoadjuvant therapy. A high score for RAD52 expression in the tumour centre was significantly associated with worse disease-free survival (DFS; p = 0.045). In contrast, reduced RAD52 expression in tumour centre samples from patients treated with neoadjuvant therapy (n = 40) significantly correlated with poor DFS (p = 0.025) and overall survival (OS; p = 0.048). Our results suggested that RAD52 may have clinical value as a prognostic marker of tumour response to neoadjuvant radiation and both disease-free status and overall survival in patients with rectal cancer.
Collapse
Affiliation(s)
- Vincent Ho
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (L.C.); (A.A.); (T.L.R.); (P.d.S.); (C.S.L.)
- Correspondence: ; Tel.: +61-2-4620-3845; Fax: +61-2-4520-3116
| | - Liping Chung
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (L.C.); (A.A.); (T.L.R.); (P.d.S.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.H.L.); (W.C.)
| | - Amandeep Singh
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia; (A.S.); (V.L.)
| | - Vivienne Lea
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia; (A.S.); (V.L.)
| | - Askar Abubakar
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (L.C.); (A.A.); (T.L.R.); (P.d.S.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.H.L.); (W.C.)
| | - Stephanie H. Lim
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.H.L.); (W.C.)
- Macarthur Cancer Therapy Centre, Campbelltown Hospital, NSW 2560, Australia
- Discipline of Medical Oncology, School of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia
| | - Wei Chua
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.H.L.); (W.C.)
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Weng Ng
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Mark Lee
- Department of Radiation Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Tara L. Roberts
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (L.C.); (A.A.); (T.L.R.); (P.d.S.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.H.L.); (W.C.)
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Paul de Souza
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (L.C.); (A.A.); (T.L.R.); (P.d.S.); (C.S.L.)
- Discipline of Medical Oncology, School of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (L.C.); (A.A.); (T.L.R.); (P.d.S.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.H.L.); (W.C.)
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia; (A.S.); (V.L.)
- Department of Radiation Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
- Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| |
Collapse
|
32
|
Does Direct and Indirect Exposure to Ionising Radiation Influence the Metastatic Potential of Breast Cancer Cells. Cancers (Basel) 2020; 12:cancers12010236. [PMID: 31963587 PMCID: PMC7016586 DOI: 10.3390/cancers12010236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
Ionising radiation (IR) is commonly used for cancer therapy; however, its potential influence on the metastatic ability of surviving cancer cells exposed directly or indirectly to IR remains controversial. Metastasis is a multistep process by which the cancer cells dissociate from the initial site, invade, travel through the blood stream or lymphatic system, and colonise distant sites. This complex process has been reported to require cancer cells to undergo epithelial-mesenchymal transition (EMT) by which the cancer cells convert from an adhesive, epithelial to motile, mesenchymal form and is also associated with changes in glycosylation of cell surface proteins, which may be functionally involved in metastasis. In this paper, we give an overview of metastatic mechanisms and of the fundamentals of cancer-associated glycosylation changes. While not attempting a comprehensive review of this wide and fast moving field, we highlight some of the accumulating evidence from in vitro and in vivo models for increased metastatic potential in cancer cells that survive IR, focusing on angiogenesis, cancer cell motility, invasion, and EMT and glycosylation. We also explore the indirect effects in cells exposed to exosomes released from irradiated cells. The results of such studies need to be interpreted with caution and there remains limited evidence that radiotherapy enhances the metastatic capacity of cancers in a clinical setting and undoubtedly has a very positive clinical benefit. However, there is potential that this therapeutic benefit may ultimately be enhanced through a better understanding of the direct and indirect effects of IR on cancer cell behaviour.
Collapse
|
33
|
Aguilar-Hernández I, Cárdenas-Chavez DL, López-Luke T, García-García A, Herrera-Domínguez M, Pisano E, Ornelas-Soto N. Discrimination of radiosensitive and radioresistant murine lymphoma cells by Raman spectroscopy and SERS. BIOMEDICAL OPTICS EXPRESS 2020; 11:388-405. [PMID: 32010523 PMCID: PMC6968773 DOI: 10.1364/boe.11.000388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/22/2019] [Accepted: 12/02/2019] [Indexed: 05/10/2023]
Abstract
Intrinsic radiosensitivity is a biological parameter known to influence the response to radiation therapy in cancer treatment. In this study, Raman spectroscopy and surface enhanced Raman spectroscopy (SERS) were successfully used in conjunction with principal component analysis (PCA) to discriminate between radioresistant (LY-R) and radiosensitive (LY-S) murine lymphoma sublines (L5178Y). PCA results for normal Raman analysis showed a differentiation between the radioresistant and radiosensitive cell lines based on their specific spectral fingerprint. In the case of SERS with gold nanoparticles (AuNPs), greater spectral enhancements were observed in the radioresistant subline in comparison to its radiosensitive counterpart, suggesting that each subline displays different interaction with AuNPs. Our results indicate that spectroscopic and chemometric techniques could be used as complementary tools for the prediction of intrinsic radiosensitivity of lymphoma samples.
Collapse
Affiliation(s)
- Iris Aguilar-Hernández
- Laboratorio de Nanotecnología Ambiental, Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. 64849, Mexico
| | - Diana L. Cárdenas-Chavez
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Atlixcáyotl 5718, Puebla, Pue., México, 72453, Mexico
| | - Tzarara López-Luke
- Instituto de Investigación en Metalurgia y Materiales, Universidad Michoacana de San Nicolás de Hidalgo, Edificio U, Ciudad Universitaria, 58030 Morelia, Mich., Mexico
| | - Alejandra García-García
- Laboratorio de síntesis y Modificación de Nanoestructuras y Materiales Bidimensionales. Centro de Investigación en Materiales Avanzados S.C. Parque PIIT. C.P. 66628, Apodaca N.L., Mexico
| | - Marcela Herrera-Domínguez
- Laboratorio de Nanotecnología Ambiental, Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. 64849, Mexico
| | - Eduardo Pisano
- Catedras CONACyT – Centro de Investigaciones en Óptica A.C., Alianza Centro 504, PIIT, Apodaca, N.L. 66629, Mexico
| | - Nancy Ornelas-Soto
- Laboratorio de Nanotecnología Ambiental, Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. 64849, Mexico
| |
Collapse
|
34
|
He K, Zhang S, Shao LL, Yin JC, Wu X, Shao YW, Yuan S, Yu J. Developing more sensitive genomic approaches to detect radioresponse in precision radiation oncology: From tissue DNA analysis to circulating tumor DNA. Cancer Lett 2019; 472:108-118. [PMID: 31837443 DOI: 10.1016/j.canlet.2019.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023]
Abstract
Despite the common application and considerable efforts to achieve precision radiotherapy (RT) in several types of cancer, RT has not yet entered the era of precision medicine; the ability to predict radiosensitivity and treatment responses in tumors and normal tissues is lacking. Therefore, development of genome-based methods for individual prognosis in radiation oncology is urgently required. Traditional DNA sequencing requires tissue samples collected during invasive operations; therefore, repeated tests are nearly impossible. Intra- and inter-tumoral heterogeneity may undermine the predictive power of a single assay from tumor samples. In contrast, analysis of circulating tumor DNA (ctDNA) allows for non-invasive and near real-time sampling of tumors. By investigating the genetic composition of tumors and monitoring dynamic changes during treatment, ctDNA analysis may potentially be clinically valuable in prediction of treatment responses prior to RT, surveillance of responses during RT, and evaluation of residual disease following RT. As a biomarker for RT response, ctDNA profiling may guide personalized treatments. In this review, we will discuss approaches of tissue DNA sequencing and ctDNA detection and summarize their clinical applications in both traditional RT and in combination with immunotherapy.
Collapse
Affiliation(s)
- Kewen He
- Department of Radiology, Shandong Cancer Hospital affiliated to Shandong University, Jinan, Shandong, 250117, People's Republic of China; Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People's Republic of China
| | - Shaotong Zhang
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, 250013, People's Republic of China
| | - Liang L Shao
- Geneseeq Technology Inc., Toronto, Ontario, M5G 1L7, Canada
| | - Jiani C Yin
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, 210032, People's Republic of China
| | - Xue Wu
- Geneseeq Technology Inc., Toronto, Ontario, M5G 1L7, Canada
| | - Yang W Shao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, 210032, People's Republic of China; School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Shuanghu Yuan
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People's Republic of China.
| | - Jinming Yu
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People's Republic of China.
| |
Collapse
|
35
|
Rassamegevanon T, Löck S, Baumann M, Krause M, von Neubeck C. Comparable radiation response of ex vivo and in vivo irradiated tumor samples determined by residual γH2AX. Radiother Oncol 2019; 139:94-100. [PMID: 31445839 DOI: 10.1016/j.radonc.2019.06.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/16/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE a) To investigate if an ex vivo cultured and irradiated tumor biopsy reflects and predicts the radiation response of the corresponding in vivo irradiated tumor measured with the DNA double strand break marker γH2AX foci. MATERIALS AND METHODS Five human head and neck squamous cell carcinoma (hHNSCC) xenograft models were used. Fine needle biopsies were taken from anesthetized tumor-bearing NMRI nude mice prior to in vivo single dose irradiation (0, 2, 4, or 8 Gy) under ambient blood flow. Biopsies were ex vivo reoxygenated and irradiated with equivalent doses. Tumors and biopsies were fixed 24 h post irradiation, and γH2AX foci were assessed in oxygenated tumor regions. RESULTS Linear regression analysis showed comparable slopes of the residual γH2AX foci dose-response curves in four out of five hHNSCC models when in vivo and ex vivo cohorts were compared. The slopes from ex vivo biopsies and in vivo tumors could classify the respective tumor model as sensitive or resistant according to the intrinsic radiation sensitivity (TCD50). CONCLUSION The ability of ex vivo irradiated tumor biopsies to reflect and predict the intrinsic radiation response of in vivo tumors increases the translational potential of the ex vivo γH2AX foci assay as a diagnostic tool for clinical practice.
Collapse
Affiliation(s)
- Treewut Rassamegevanon
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany.
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, 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, and; Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, 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, and; Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Cläre von Neubeck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
Walker AK, Karaszi K, Valentine H, Strauss VY, Choudhury A, McGill S, Wen K, Brown MD, Ramani V, Bhattarai S, Teo MTW, Yang L, Myers KA, Deshmukh N, Denley H, Browning L, Love SB, Iyer G, Clarke NW, Hall E, Huddart R, James ND, Hoskin PJ, West CML, Kiltie AE. MRE11 as a Predictive Biomarker of Outcome After Radiation Therapy in Bladder Cancer. Int J Radiat Oncol Biol Phys 2019; 104:809-818. [PMID: 30885775 PMCID: PMC6588678 DOI: 10.1016/j.ijrobp.2019.03.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/06/2019] [Accepted: 03/09/2019] [Indexed: 01/08/2023]
Abstract
PURPOSE Organ-confined muscle-invasive bladder cancer is treated with cystectomy or bladder preservation techniques, including radiation therapy. There are currently no biomarkers to inform management decisions and aid patient choice. Previously we showed high levels of MRE11 protein, assessed by immunohistochemistry (IHC), predicted outcome after radiation therapy, but not cystectomy. Therefore, we sought to develop the MRE11 IHC assay for clinical use and define its relationship to clinical outcome in samples from 2 major clinical trials. METHODS AND MATERIALS Samples from the BCON and BC2001 randomized controlled trials and a cystectomy cohort were stained using automated IHC methods and scored for MRE11 in 3 centers in the United Kingdom. RESULTS Despite step-wise creation of scoring cards and standard operating procedures for staining and interpretation, there was poor intercenter scoring agreement (kappa, 0.32; 95% confidence interval, 0.17-0.47). No significant associations between MRE11 scores and cause-specific survival were identified in BCON (n = 132) and BC2001 (n = 221) samples. Reoptimized staining improved agreement between scores from BCON tissue microarrays (n = 116), but MRE11 expression was not prognostic for cause-specific survival. CONCLUSIONS Manual IHC scoring of MRE11 was not validated as a reproducible biomarker of radiation-based bladder preservation success. There is a need for automated quantitative methods or a reassessment of how DNA-damage response relates to clinical outcomes.
Collapse
Affiliation(s)
- Alexandra K Walker
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Katalin Karaszi
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Helen Valentine
- Translational Radiobiology Group, Division of Cancer Sciences, Christie Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Victoria Y Strauss
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Diseases, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ananya Choudhury
- Translational Radiobiology Group, Division of Cancer Sciences, Christie Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Shaun McGill
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Kaisheng Wen
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Michael D Brown
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| | - Vijay Ramani
- Department of Urology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Selina Bhattarai
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Mark T W Teo
- Leeds Cancer Centre, St James's University Hospital, Leeds, United Kingdom
| | - Lingjian Yang
- Translational Radiobiology Group, Division of Cancer Sciences, Christie Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Kevin A Myers
- Experimental Cancer Medicine Centre, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Nayneeta Deshmukh
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Helen Denley
- Department of Cellular Pathology, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Lisa Browning
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom; NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Sharon B Love
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Diseases, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Gopa Iyer
- Weill Cornell Medical College, Cornell University, New York, New York
| | - Noel W Clarke
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom; Department of Urology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Emma Hall
- Clinical Trials and Statistics Unit, Institute of Cancer Research, London, United Kingdom
| | - Robert Huddart
- Academic Uro-Oncology Unit, The Royal Marsden NHS Foundation Trust, Sutton, London, United Kingdom; The Institute of Cancer Research, London, United Kingdom
| | - Nicholas D James
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Peter J Hoskin
- Cancer Centre, Mount Vernon Hospital, Northwood, Middlesex, United Kingdom; Manchester Cancer Research Centre, University of Manchester, Manchester, United Kingdom
| | - Catharine M L West
- Translational Radiobiology Group, Division of Cancer Sciences, Christie Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Anne E Kiltie
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
37
|
Towards optimal stopping in radiation therapy. Radiother Oncol 2019; 134:96-100. [DOI: 10.1016/j.radonc.2019.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 12/25/2022]
|
38
|
Mechanistic Modelling of Radiation Responses. Cancers (Basel) 2019; 11:cancers11020205. [PMID: 30744204 PMCID: PMC6406300 DOI: 10.3390/cancers11020205] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 12/30/2022] Open
Abstract
Radiobiological modelling has been a key part of radiation biology and therapy for many decades, and many aspects of clinical practice are guided by tools such as the linear-quadratic model. However, most of the models in regular clinical use are abstract and empirical, and do not provide significant scope for mechanistic interpretation or making predictions in novel cell lines or therapies. In this review, we will discuss the key areas of ongoing mechanistic research in radiation biology, including physical, chemical, and biological steps, and review a range of mechanistic modelling approaches which are being applied in each area, highlighting the possible opportunities and challenges presented by these techniques.
Collapse
|
39
|
Cytokine Modulation in Breast Cancer Patients Undergoing Radiotherapy: A Revision of the Most Recent Studies. Int J Mol Sci 2019; 20:ijms20020382. [PMID: 30658426 PMCID: PMC6359111 DOI: 10.3390/ijms20020382] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/13/2022] Open
Abstract
Breast cancer (BC) is the most common tumor and the second cause for cancer-related death in women worldwide, although combined treatments are well-established interventions. Several effects seem to be responsible for poor outcomes in advanced or triple-negative BC patients. Focusing on the interaction of ionizing radiation with tumor and normal tissues, the role of cytokine modulation as a surrogate of immunomodulation must still be explored. In this work, we carried out an overview of studies published in the last five years involving the cytokine profile in BC patients undergoing radiotherapy. The goal of this review was to evaluate the profile and modulation of major cytokines and interleukins as potential biomarkers of survival, treatment response, and toxicity in BC patient undergoing radiotherapy. Out of 47 retrieved papers selected using PubMed search, 15 fulfilled the inclusion criteria. Different studies reported that the modulation of specific cytokines was time- and treatment-dependent. Radiotherapy (RT) induces the modulation of inflammatory cytokines up to 6 months for most of the analyzed cytokines, which in some cases can persist up to several years post-treatment. The role of specific cytokines as prognostic and predictive of radiotherapy outcome is critically discussed.
Collapse
|
40
|
Story MD, Durante M. Radiogenomics. Med Phys 2018; 45:e1111-e1122. [DOI: 10.1002/mp.13064] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/23/2018] [Accepted: 04/27/2018] [Indexed: 12/24/2022] Open
Affiliation(s)
- Michael D. Story
- Department of Radiation Oncology University of Texas, Southwestern Medical Center Dallas TX USA
- Simmons Comprehensive Cancer Center University of Texas, Southwestern Medical Center Dallas TX USA
| | - Marco Durante
- Trento Institute for Fundamental Physics Applications National Institute for Nuclear Physics Trento Italy
- Department of Physics University of Trento Trento Italy
| |
Collapse
|
41
|
Wen P, Xia J, Cao X, Chen B, Tao Y, Wu L, Xu A, Zhao G. dbCRSR: a manually curated database for regulation of cancer radiosensitivity. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2018; 2018:5025485. [PMID: 29860480 PMCID: PMC6007213 DOI: 10.1093/database/bay049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/26/2018] [Indexed: 12/11/2022]
Abstract
Radiotherapy is used to treat approximately 50% of all cancer patients, with varying prognoses. Intrinsic radiosensitivity is an important factor underlying the radiotherapeutic efficacy of this precise treatment. During the past decades, great efforts have been made to improve radiotherapy treatment through multiple strategies. However, invaluable data remains buried in the extensive radiotherapy literature, making it difficult to obtain an overall view of the detailed mechanisms leading to radiosensitivity, thus limiting advances in radiotherapy. To address this issue, we collected data from the relevant literature contained in the PubMed database and developed a literature-based database that we term the cancer radiosensitivity regulation factors database (dbCRSR). dbCRSR is a manually curated catalogue of radiosensitivity, containing multiple radiosensitivity regulation factors (395 coding genes, 119 non-coding RNAs and 306 chemical compounds) with appropriate annotation. To illustrate the value of the data we collected, data mining was performed including functional annotation and network analysis. In summary, dbCRSR is the first literature-based database to focus on radiosensitivity and provides a resource to better understand the detailed mechanisms of radiosensitivity. We anticipate dbCRSR will be a useful resource to enrich our knowledge and to promote further study of radiosensitivity. Database URL: http://bioinfo.ahu.edu.cn:8080/dbCRSR/
Collapse
Affiliation(s)
- Pengbo Wen
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei, Anhui, People's Republic of China.,University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Junfeng Xia
- Institute of Physical Science and Information Technology, School of Computer Science and Technology, Anhui University, Hefei, Anhui, People's Republic of China
| | - Xianbin Cao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei, Anhui, People's Republic of China
| | - Bin Chen
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei, Anhui, People's Republic of China
| | - Yinping Tao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei, Anhui, People's Republic of China
| | - Lijun Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei, Anhui, People's Republic of China
| | - An Xu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei, Anhui, People's Republic of China
| | - Guoping Zhao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei, Anhui, People's Republic of China
| |
Collapse
|
42
|
Thompson MK, Poortmans P, Chalmers AJ, Faivre-Finn C, Hall E, Huddart RA, Lievens Y, Sebag-Montefiore D, Coles CE. Practice-changing radiation therapy trials for the treatment of cancer: where are we 150 years after the birth of Marie Curie? Br J Cancer 2018; 119:389-407. [PMID: 30061587 PMCID: PMC6117262 DOI: 10.1038/s41416-018-0201-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
As we mark 150 years since the birth of Marie Curie, we reflect on the global advances made in radiation oncology and the current status of radiation therapy (RT) research. Large-scale international RT clinical trials have been fundamental in driving evidence-based change and have served to improve cancer management and to reduce side effects. Radiation therapy trials have also improved practice by increasing quality assurance and consistency in treatment protocols across multiple centres. This review summarises some of the key RT practice-changing clinical trials over the last two decades, in four common cancer sites for which RT is a crucial component of curative treatment: breast, lung, urological and lower gastro-intestinal cancer. We highlight the global inequality in access to RT, and the work of international organisations, such as the International Atomic Energy Agency (IAEA), the European SocieTy for Radiotherapy and Oncology (ESTRO), and the United Kingdom National Cancer Research Institute Clinical and Translational Radiotherapy Research Working Group (CTRad), that aim to improve access to RT and facilitate radiation research. We discuss some emerging RT technologies including proton beam therapy and magnetic resonance linear accelerators and predict likely future directions in clinical RT research.
Collapse
Affiliation(s)
- Mareike K Thompson
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | | | - Anthony J Chalmers
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, University of Manchester; The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Emma Hall
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, Sutton, London, SM2 5NG, UK
| | - Robert A Huddart
- Section of Radiotherapy and Imaging, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Yolande Lievens
- Department of Radiation Oncology, Ghent University Hospital and Ghent University, C. Heymanslaan, 9000, Ghent, Belgium
| | - David Sebag-Montefiore
- Radiotherapy Research Group, Leeds Institute of Cancer and Pathology, University of Leeds; Leeds Cancer Centre, St James's University Hospitals, Leeds, LS9 7TF, UK
| | - Charlotte E Coles
- Department of Oncology, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
43
|
Zhou ZR, Yang ZZ, Yu XL, Guo XM. Highlights on molecular targets for radiosensitization of breast cancer cells: Current research status and prospects. Cancer Med 2018; 7:3110-3117. [PMID: 29856131 PMCID: PMC6051209 DOI: 10.1002/cam4.1588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/16/2022] Open
Abstract
In the past, searching for effective radiotherapy sensitization molecular targets and improving the radiation sensitivity of malignant tumors was the hot topic for the oncologists, but with little achievements. We will summarize the research results about breast cancer irradiation sensitization molecular targets over the past two decades; we mainly focus on the following aspects: DNA damage repair and radiation sensitization, cell cycle regulation and radiation sensitization, cell autophagy regulation and radiation sensitization, and radiation sensitivity prediction and breast cancer radiotherapy scheme making. And based on this summary, we will put forward some of our viewpoints.
Collapse
Affiliation(s)
- Zhi-Rui Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhao-Zhi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Li Yu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Mao Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
44
|
Ikegwuonu T, Haddow G, Tait J, Murray AF, Kunkler IH. Horizon scanning implanted biosensors in personalising breast cancer management: First pilot study of breast cancer patients views. Health Sci Rep 2018; 1:30. [PMID: 30613798 PMCID: PMC6266376 DOI: 10.1002/hsr2.30] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/24/2017] [Accepted: 01/25/2018] [Indexed: 12/13/2022] Open
Abstract
AIMS This study aimed to explore breast cancer patients' understanding and acceptability of implanted biosensors (BS) within the primary tumour to personalise adjuvant radiotherapy, and to determine optimal design and number of BS, and evaluate potential clinical benefits as well as concerns about tolerance, toxicity, dwell time, and confidentiality of data. PATIENTS AND METHODS A total of 32 patients treated by surgery (29 breast conserving, 3 mastectomy), postoperative radiotherapy and systemic therapy for early breast cancer, were recruited from a posttreatment radiotherapy clinic at a cancer centre. Patients participated in semistructured interviews. Interview transcripts were analysed using qualitative methods. RESULTS Participants were aged 39 to 87 years, with a median age of 62 years. Most (N = 23[72%]) were unfamiliar with biosensors. The majority (N = 29[90.6%]) were supportive of the technology's potential use in future breast cancer treatment and were willing to accept biosensors (N = 28[88%]) if they were endorsed by their breast cancer consultant. Only 3 patients expressed concerns, predominantly about uncertainties on their role in the diagnostic and treatment pathway. Patients were flexible about the size and shape of BS, but had a preference for small size (N = 28 [87.5%]). Most (N = 22[69%]) would accept implantation of more than 5 BS and were flexible (N = 22[69%]) about indefinite dwell time. Patients had a strong preference for wireless powering of the BS (N = 28[87.5%]). Few had concerns about loss of confidentiality of data collected. All patients considered biosensors to be potentially of important clinical benefit. CONCLUSIONS While knowledge of biosensors was limited, patients were generally supportive of biosensors implanted within the primary tumour to collect data that might personalise and improve breast cancer radiotherapy in future.
Collapse
Affiliation(s)
| | - Gill Haddow
- Science, Technology and Innovation StudiesThe University of EdinburghEdinburghScotland
| | - Joyce Tait
- Innogen InstituteThe University of EdinburghEdinburghScotland
| | - Alan F. Murray
- Institute for Bioengineering, School of EngineeringUniversity of EdinburghEdinburghScotland
| | - Ian H. Kunkler
- Edinburgh Cancer Research Centre, Institute of Genetic and Molecular MedicineWestern General HospitalEdinburghScotland
| |
Collapse
|
45
|
Meksiarun P, Aoki PHB, Van Nest SJ, Sobral-Filho RG, Lum JJ, Brolo AG, Jirasek A. Breast cancer subtype specific biochemical responses to radiation. Analyst 2018; 143:3850-3858. [DOI: 10.1039/c8an00345a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
External beam radiotherapy is a common form of treatment for breast cancer.
Collapse
Affiliation(s)
- Phiranuphon Meksiarun
- Department of Physics
- I.K. Barber School of Arts and Sciences
- University of British Columbia – Okanagan
- Kelowna
- Canada
| | - Pedro H. B. Aoki
- São Paulo State University (UNESP)
- School of Sciences
- Humanities and Languages
- Campus Assis
- Brazil
| | | | | | - Julian J. Lum
- University of Victoria
- Department of Biochemistry and Microbiology
- Victoria
- Canada
- Trev and Joyce Deeley Research Centre
| | | | - Andrew Jirasek
- Department of Physics
- I.K. Barber School of Arts and Sciences
- University of British Columbia – Okanagan
- Kelowna
- Canada
| |
Collapse
|
46
|
Early Postoperative Low Expression of RAD50 in Rectal Cancer Patients Associates with Disease-Free Survival. Cancers (Basel) 2017; 9:cancers9120163. [PMID: 29189711 PMCID: PMC5742811 DOI: 10.3390/cancers9120163] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Molecular biomarkers have the potential to predict response to the treatment of rectal cancer. In this study, we aimed to evaluate the prognostic and clinicopathological implication of RAD50 (DNA repair protein RAD50 homolog) expression in rectal cancer. METHODS A total of 266 rectal cancer patients who underwent surgery and received chemo- and radiotherapy between 2000 and 2011 were involved in the study. Postoperative RAD50 expression was determined by immunohistochemistry in surgical samples (n = 266). RESULTS Using Kaplan-Meier survival analysis, we found that low RAD50 expression in postoperative samples was associated with worse disease free survival (p = 0.001) and overall survival (p < 0.001) in early stage/low-grade tumors. In a comparison of patients with low vs. high RAD50 expression, we found that low levels of postoperative RAD50 expression in rectal cancer tissues were significantly associated with perineural invasion (p = 0.002). CONCLUSION Expression of RAD50 in rectal cancer may serve as a prognostic biomarker for long-term survival of patients with perineural invasion-positive tumors and for potential use in early stage and low-grade rectal cancer assessment.
Collapse
|
47
|
Habash M, Bohorquez LC, Kyriakou E, Kron T, Martin OA, Blyth BJ. Clinical and Functional Assays of Radiosensitivity and Radiation-Induced Second Cancer. Cancers (Basel) 2017; 9:cancers9110147. [PMID: 29077012 PMCID: PMC5704165 DOI: 10.3390/cancers9110147] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 01/10/2023] Open
Abstract
Whilst the near instantaneous physical interaction of radiation energy with living cells leaves little opportunity for inter-individual variation in the initial yield of DNA damage, all the downstream processes in how damage is recognized, repaired or resolved and therefore the ultimate fate of cells can vary across the population. In the clinic, this variability is observed most readily as rare extreme sensitivity to radiotherapy with acute and late tissue toxic reactions. Though some radiosensitivity can be anticipated in individuals with known genetic predispositions manifest through recognizable phenotypes and clinical presentations, others exhibit unexpected radiosensitivity which nevertheless has an underlying genetic cause. Currently, functional assays for cellular radiosensitivity represent a strategy to identify patients with potential radiosensitivity before radiotherapy begins, without needing to discover or evaluate the impact of the precise genetic determinants. Yet, some of the genes responsible for extreme radiosensitivity would also be expected to confer susceptibility to radiation-induced cancer, which can be considered another late adverse event associated with radiotherapy. Here, the utility of functional assays of radiosensitivity for identifying individuals susceptible to radiotherapy-induced second cancer is discussed, considering both the common mechanisms and important differences between stochastic radiation carcinogenesis and the range of deterministic acute and late toxic effects of radiotherapy.
Collapse
Affiliation(s)
- Mohammad Habash
- Cancer Research Division, Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC 3000, Australia.
- Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Luis C Bohorquez
- Physical Sciences, Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC 3000, Australia.
| | - Elizabeth Kyriakou
- Physical Sciences, Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC 3000, Australia.
| | - Tomas Kron
- Physical Sciences, Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC 3000, Australia.
| | - Olga A Martin
- Cancer Research Division, Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC 3000, Australia.
- Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC 3000, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Benjamin J Blyth
- Cancer Research Division, Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC 3000, Australia.
- Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC 3000, Australia.
| |
Collapse
|
48
|
Flippot R, Massard C, Auclin E, Azria D, Bourien H, Rochigneux P, Schernberg A, Verlingue L, Zafrani L, Vignot S. Quelle vision des biomarqueurs en 2017 ? Promesses et défis de la médecine personnalisée en oncologie. Bull Cancer 2017; 104:735-743. [DOI: 10.1016/j.bulcan.2017.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/18/2017] [Indexed: 10/19/2022]
|
49
|
MRE11 and ATM Expression Levels Predict Rectal Cancer Survival and Their Association with Radiotherapy Response. PLoS One 2016; 11:e0167675. [PMID: 27930716 PMCID: PMC5145179 DOI: 10.1371/journal.pone.0167675] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/20/2016] [Indexed: 01/02/2023] Open
Abstract
Background Aberrant expression of DNA repair proteins is associated with poor survival in cancer patients. We investigated the combined expression of MRE11 and ATM as a predictive marker of response to radiotherapy in rectal cancer. Methods MRE11 and ATM expression were examined in tumor samples from 262 rectal cancer patients who underwent surgery for rectal cancer, including a sub-cohort of 54 patients who were treated with neoadjuvant radiotherapy. The relationship between expression of the two-protein panel and tumor regression grade (TRG) was assessed by Mann–Whitney U test and receiver operating characteristics area under curve (ROC-AUC) analysis. The association between expression of the two-protein panel and clinicopathologic variables and survival was examined by Kaplan-Meier methods and Cox regression analysis. Results A high score for two-protein combined expression in the tumor center (TC) was significantly associated with worse disease-free survival (DFS) (P = 0.035) and overall survival (OS) (P = 0.003) in the whole cohort, and with DFS (P = 0.028) and OS (P = 0.024) in the neoadjuvant subgroup (n = 54). In multivariate analysis, the two-protein combination panel (HR = 2.178, 95% CI 1.115–4.256, P = 0.023) and perineural invasion (HR = 2.183, 95% CI 1.222–3.899, P = 0.008) were significantly associated with DFS. Using ROC-AUC analysis of good versus poor histological tumor response among patients treated preoperatively with radiotherapy, the average ROC-AUC was 0.745 for the combined panel, 0.618 for ATM alone, and 0.711 for MRE11 alone. Conclusions The MRE11/ATM two-protein panel developed in this study may have clinical value as a predictive marker of tumor response to neoadjuvant radiotherapy, and a prognostic marker for disease-free and overall survival.
Collapse
|
50
|
Differences in DNA Repair Capacity, Cell Death and Transcriptional Response after Irradiation between a Radiosensitive and a Radioresistant Cell Line. Sci Rep 2016; 6:27043. [PMID: 27245205 PMCID: PMC4887990 DOI: 10.1038/srep27043] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/13/2016] [Indexed: 12/14/2022] Open
Abstract
Normal tissue toxicity after radiotherapy shows variability between patients, indicating inter-individual differences in radiosensitivity. Genetic variation probably contributes to these differences. The aim of the present study was to determine if two cell lines, one radiosensitive (RS) and another radioresistant (RR), showed differences in DNA repair capacity, cell viability, cell cycle progression and, in turn, if this response could be characterised by a differential gene expression profile at different post-irradiation times. After irradiation, the RS cell line showed a slower rate of γ-H2AX foci disappearance, a higher frequency of incomplete chromosomal aberrations, a reduced cell viability and a longer disturbance of the cell cycle when compared to the RR cell line. Moreover, a greater and prolonged transcriptional response after irradiation was induced in the RS cell line. Functional analysis showed that 24 h after irradiation genes involved in “DNA damage response”, “direct p53 effectors” and apoptosis were still differentially up-regulated in the RS cell line but not in the RR cell line. The two cell lines showed different response to IR and can be distinguished with cell-based assays and differential gene expression analysis. The results emphasise the importance to identify biomarkers of radiosensitivity for tailoring individualized radiotherapy protocols.
Collapse
|