1
|
Chen JLY, Pan CK, Lin LC, Huang YS, Huang TH, Yang SJ, Kuo SH, Lin YL. Combination of ataxia telangiectasia and Rad3-related inhibition with ablative radiotherapy remodels the tumor microenvironment and enhances immunotherapy response in lung cancer. Cancer Immunol Immunother 2024; 74:8. [PMID: 39487895 PMCID: PMC11531452 DOI: 10.1007/s00262-024-03864-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
We investigated the combined effects of ataxia telangiectasia and Rad3-related (ATR) inhibition, ablative radiotherapy, and immune checkpoint inhibitor (ICI) therapy against lung cancer. ATR inhibitor was administered combined with ablative radiotherapy to assess its radiosensitizing effect on lung cancer cells. Treatment response and survival were evaluated in vivo using A549 xenograft flank tumor and synchronous LLC lung and flank tumor mouse models. Mice received ablative radiotherapy (12 Gy/d for 2 d), ATR inhibitor, and ICI. The tumor microenvironment was assessed in irradiated flank and non-irradiated lung tumors. Programmed death-ligand 1 expression was upregulated after irradiation. ATR inhibition attenuated this upregulation. ATR inhibitor pretreatment decreased cell survival after irradiation by inhibiting DNA double-strand break repair, inducing mitotic cell death, and altering cell cycle progression. ATR inhibition enhanced radiation-induced damage-associated molecular patterns determined by high mobility group box 1 quantification and activated the cyclic GMP-AMP synthase-stimulator of interferon genes pathway. Combined ATR inhibition and ablative radiotherapy inhibited tumor growth and improved survival in mice. Adding ICI therapy further enhanced local antitumor effects, reducing the metastatic lung tumor burden and remodeling the tumor microenvironment through immunogenic cell death induction and enhanced immune cell infiltration. Triple therapy increased immune cell infiltration in distant non-irradiated lung tumors and stimulated the generation of protective T-cell immunity in splenocytes. Safety analysis showed minimal toxicity. ATR inhibition enhanced the efficacy of ablative radiotherapy and immunotherapy in lung cancer. These findings underscore the importance of combination therapies for enhancing systemic antitumor immune responses and outcomes.
Collapse
Affiliation(s)
- Jenny Ling-Yu Chen
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Kai Pan
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung-Shan S. Rd., Taipei, 100, Taiwan
| | - Li-Cheng Lin
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung-Shan S. Rd., Taipei, 100, Taiwan
| | - Yu-Sen Huang
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsung-Hsuan Huang
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung-Shan S. Rd., Taipei, 100, Taiwan
| | - Shu-Jyuan Yang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Sung-Hsin Kuo
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Li Lin
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung-Shan S. Rd., Taipei, 100, Taiwan.
| |
Collapse
|
2
|
Dunne VL, Ghita-Pettigrew M, Redmond KM, Small DM, Weldon S, Taggart CC, Prise KM, Hanna GG, Butterworth KT. PTEN Depletion Increases Radiosensitivity in Response to Ataxia Telangiectasia-Related-3 (ATR) Inhibition in Non-Small Cell Lung Cancer (NSCLC). Int J Mol Sci 2024; 25:7817. [PMID: 39063060 PMCID: PMC11277409 DOI: 10.3390/ijms25147817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Radiotherapy (RT) treatment is an important strategy for the management of non-small cell lung cancer (NSCLC). Local recurrence amongst patients with late-stage NSCLC remains a challenge. The loss of PTEN has been associated with radio-resistance. This study aimed to examine the efficacy of RT combined with ataxia telangiectasia-mutated Rad3-related (ATR) inhibition using Ceralasertib in phosphatase and tensin homolog (PTEN)-depleted NSCLC cells and to assess early inflammatory responses indicative of radiation pneumonitis (RP) after combined-modality treatment. Small hairpin RNA (shRNA) transfections were used to generate H460 and A549 PTEN-depleted models. Ceralasertib was evaluated as a single agent and in combination with RT in vitro and in vivo. Histological staining was used to assess immune cell infiltration in pneumonitis-prone C3H/NeJ mice. Here, we report that the inhibition of ATR in combination with RT caused a significant reduction in PTEN-depleted NSCLC cells, with delayed DNA repair and reduced cell viability, as shown by an increase in cells in Sub G1. Combination treatment in vivo significantly inhibited H460 PTEN-depleted tumour growth in comparison to H460 non-targeting PTEN-expressing (NT) cell-line-derived xenografts (CDXs). Additionally, there was no significant increase in infiltrating macrophages or neutrophils except at 4 weeks, whereby combination treatment significantly increased macrophage levels relative to RT alone. Overall, our study demonstrates that ceralasertib and RT combined preferentially sensitises PTEN-depleted NSCLC models in vitro and in vivo, with no impact on early inflammatory response indicative of RP. These findings provide a rationale for evaluating ATR inhibition in combination with RT in NSCLC patients with PTEN mutations.
Collapse
Affiliation(s)
- Victoria L. Dunne
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Mihaela Ghita-Pettigrew
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Kelly M. Redmond
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Donna M. Small
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Sinéad Weldon
- Airway Innate Immunity Research Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (S.W.); (C.C.T.)
| | - Clifford C. Taggart
- Airway Innate Immunity Research Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (S.W.); (C.C.T.)
| | - Kevin M. Prise
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Gerard G. Hanna
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast BT9 7AB, UK;
| | - Karl T. Butterworth
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| |
Collapse
|
3
|
Brown KH, Ghita-Pettigrew M, Kerr BN, Mohamed-Smith L, Walls GM, McGarry CK, Butterworth KT. Characterisation of quantitative imaging biomarkers for inflammatory and fibrotic radiation-induced lung injuries using preclinical radiomics. Radiother Oncol 2024; 192:110106. [PMID: 38253201 DOI: 10.1016/j.radonc.2024.110106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024]
Abstract
BACKGROUND AND PURPOSE Radiomics is a rapidly evolving area of research that uses medical images to develop prognostic and predictive imaging biomarkers. In this study, we aimed to identify radiomics features correlated with longitudinal biomarkers in preclinical models of acute inflammatory and late fibrotic phenotypes following irradiation. MATERIALS AND METHODS Female C3H/HeN and C57BL6 mice were irradiated with 20 Gy targeting the upper lobe of the right lung under cone-beam computed tomography (CBCT) image-guidance. Blood samples and lung tissue were collected at baseline, weeks 1, 10 & 30 to assess changes in serum cytokines and histological biomarkers. The right lung was segmented on longitudinal CBCT scans using ITK-SNAP. Unfiltered and filtered (wavelet) radiomics features (n = 842) were extracted using PyRadiomics. Longitudinal changes were assessed by delta analysis and principal component analysis (PCA) was used to remove redundancy and identify clustering. Prediction of acute (week 1) and late responses (weeks 20 & 30) was performed through deep learning using the Random Forest Classifier (RFC) model. RESULTS Radiomics features were identified that correlated with inflammatory and fibrotic phenotypes. Predictive features for fibrosis were detected from PCA at 10 weeks yet overt tissue density was not detectable until 30 weeks. RFC prediction models trained on 5 features were created for inflammation (AUC 0.88), early-detection of fibrosis (AUC 0.79) and established fibrosis (AUC 0.96). CONCLUSIONS This study demonstrates the application of deep learning radiomics to establish predictive models of acute and late lung injury. This approach supports the wider application of radiomics as a non-invasive tool for detection of radiation-induced lung complications.
Collapse
Affiliation(s)
- Kathryn H Brown
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Northern Ireland, UK.
| | - Mihaela Ghita-Pettigrew
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Northern Ireland, UK
| | - Brianna N Kerr
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Northern Ireland, UK
| | - Letitia Mohamed-Smith
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Northern Ireland, UK
| | - Gerard M Walls
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Northern Ireland, UK; Northern Ireland Cancer Centre, Belfast Health & Social Care Trust, Northern Ireland, UK
| | - Conor K McGarry
- Northern Ireland Cancer Centre, Belfast Health & Social Care Trust, Northern Ireland, UK
| | - Karl T Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Northern Ireland, UK
| |
Collapse
|
4
|
Liu L, Hu X, Feng J, Lei A, Huang S, Liu X, Liu H, Luo L, Yao W. Suppression of DNMT1 combined with ATM or ATR inhibitor as a therapeutic combination of acute myeloid leukemia. Anticancer Drugs 2024; 35:251-262. [PMID: 38164802 PMCID: PMC10833198 DOI: 10.1097/cad.0000000000001564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 01/03/2024]
Abstract
The potential treatment option of targeting DNA methyltransferase 1 (DNMT1) has been explored, but further investigation is required to assess the efficacy of combination therapy in acute myeloid leukemia (AML). In this study, bioinformatics and online databases were utilized to select the combined therapeutic targets. The potential kinases associated with DNMT1-related genes in AML were analyzed using the Cancer Genome Atlas (TCGA) database and X2K Appyter (Expression2Kinases) database. In-vitro evaluations were conducted to assess the synergistic effects between DNMT1 and ATR/ATM in five AML cell lines (MOLM-16, NB-4, HEL 92.1.7, HEL, EOL-1). In our study, ATR and ATM are primarily the kinases associated with DNMT1-related genes in AML. We observed a significant upregulation of DNMT1, ATR, and ATM expression in AML tissues and cell lines. The five AML cell lines demonstrated sensitivity to monotherapy with GSK-368, AZD-1390, or AZD-6738 (EC50 value ranges from 5.461 to 7.349 nM, 5.821 to 10.120 nM, and 7.618 to 10.100 nM, respectively). A considerable synergistic effect was observed in AML cell lines when combining GSK-368 and AZD-1390, GSK-368 and AZD-6738, or AZD-1390 and AZD-6738, resulting in induced cell apoptosis and inhibited cell growth. DNMT1, ATM, and ATR possess potential as therapeutic targets for AML. Both individual targeting and combination targeting of these molecules have been confirmed as promising therapeutic approaches for AML.
Collapse
Affiliation(s)
- Lei Liu
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| | - Xiaoyan Hu
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| | - Jing Feng
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| | - Anhui Lei
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| | - Shiying Huang
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| | - Xian Liu
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| | - Hui Liu
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| | - Lan Luo
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| | - Wenyan Yao
- Department of Hematology and Oncology, The First People’s Hospital of Guiyang, Guiyang city, Guizhou Province, China
| |
Collapse
|
5
|
Khamidullina AI, Abramenko YE, Bruter AV, Tatarskiy VV. Key Proteins of Replication Stress Response and Cell Cycle Control as Cancer Therapy Targets. Int J Mol Sci 2024; 25:1263. [PMID: 38279263 PMCID: PMC10816012 DOI: 10.3390/ijms25021263] [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: 12/07/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
Replication stress (RS) is a characteristic state of cancer cells as they tend to exchange precision of replication for fast proliferation and increased genomic instability. To overcome the consequences of improper replication control, malignant cells frequently inactivate parts of their DNA damage response (DDR) pathways (the ATM-CHK2-p53 pathway), while relying on other pathways which help to maintain replication fork stability (ATR-CHK1). This creates a dependency on the remaining DDR pathways, vulnerability to further destabilization of replication and synthetic lethality of DDR inhibitors with common oncogenic alterations such as mutations of TP53, RB1, ATM, amplifications of MYC, CCNE1 and others. The response to RS is normally limited by coordination of cell cycle, transcription and replication. Inhibition of WEE1 and PKMYT1 kinases, which prevent unscheduled mitosis entry, leads to fragility of under-replicated sites. Recent evidence also shows that inhibition of Cyclin-dependent kinases (CDKs), such as CDK4/6, CDK2, CDK8/19 and CDK12/13 can contribute to RS through disruption of DNA repair and replication control. Here, we review the main causes of RS in cancers as well as main therapeutic targets-ATR, CHK1, PARP and their inhibitors.
Collapse
Affiliation(s)
- Alvina I. Khamidullina
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia; (A.I.K.); (Y.E.A.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Yaroslav E. Abramenko
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia; (A.I.K.); (Y.E.A.)
| | - Alexandra V. Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Victor V. Tatarskiy
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia; (A.I.K.); (Y.E.A.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| |
Collapse
|
6
|
Venugopala KN. Targeting the DNA Damage Response Machinery for Lung Cancer Treatment. Pharmaceuticals (Basel) 2022; 15:ph15121475. [PMID: 36558926 PMCID: PMC9781725 DOI: 10.3390/ph15121475] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Lung cancer is considered the most commonly diagnosed cancer and one of the leading causes of death globally. Despite the responses from small-cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) patients to conventional chemo- and radiotherapies, the current outcomes are not satisfactory. Recently, novel advances in DNA sequencing technologies have started to take off which have provided promising tools for studying different tumors for systematic mutation discovery. To date, a limited number of DDR inhibition trials have been conducted for the treatment of SCLC and NSCLC patients. However, strategies to test different DDR inhibitor combinations or to target multiple pathways are yet to be explored. With the various biomarkers that have either been recently discovered or are the subject of ongoing investigations, it is hoped that future trials would be designed to allow for studying targeted treatments in a biomarker-enriched population, which is defensible for the improvement of prognosis for SCLC and NSCLC patients. This review article sheds light on the different DNA repair pathways and some of the inhibitors targeting the proteins involved in the DNA damage response (DDR) machinery, such as ataxia telangiectasia and Rad3-related protein (ATR), DNA-dependent protein kinase (DNA-PK), and poly-ADP-ribose polymerase (PARP). In addition, the current status of DDR inhibitors in clinical settings and future perspectives are discussed.
Collapse
Affiliation(s)
- Katharigatta N. Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4000, South Africa
| |
Collapse
|
7
|
Kiesel B, Parise RA, Krishnamurthy A, Gore S, Beumer JH. Quantitation of the ataxia-telangiectasia-mutated and Rad3-related inhibitor elimusertib (BAY-1895344) in human plasma using LC-MS/MS. Biomed Chromatogr 2022; 36:e5455. [PMID: 35876841 PMCID: PMC9731518 DOI: 10.1002/bmc.5455] [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: 04/05/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/07/2022]
Abstract
Ataxia-telangiectasia-mutated and Rad3-related (ATR) is master regulator of the DNA-damage response that, through multiple mechanisms, can promote cancer cell survival in response to replication stress from sources, including chemotherapy and radiation. Elimusertib (BAY-1895344) is an orally available small-molecule ATR inhibitor currently in preclinical and clinical development for cancer treatment. To support these studies and define elimusertib pharmacokinetics, we developed a HPLC-MS method for its quantitation. A 50-μL volume of plasma was subjected to acetonitrile protein precipitation and then chromatographic separation using a Phenomenex Polar-RP column (2 × 50 mm, 4 μm) and a gradient mobile phase consisting of 0.1% formic acid in acetonitrile and water during a 7-min run time. Mass spectrometric detection was achieved using a SCIEX 4000 triple-stage mass spectrometer with electrospray positive-mode ionization. With a stable isotopic internal standard, the assay was linear from 30 to 5000 ng/mL and proved to be both accurate (93.5-108.2%) and precise (<6.3% coefficient of variation) fulfilling criteria from the Food and Drug Administration guidance on bioanalytical method validation. This LC-MS/MS assay will support several ongoing clinical studies by defining elimusertib pharmacokinetics.
Collapse
Affiliation(s)
- Brian Kiesel
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
| | - Robert A. Parise
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Anuradha Krishnamurthy
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Steven Gore
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Jan H. Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
8
|
The Prognostic and Therapeutic Potential of DNA Damage Repair Pathway Alterations and Homologous Recombination Deficiency in Lung Cancer. Cancers (Basel) 2022; 14:cancers14215305. [DOI: 10.3390/cancers14215305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022] Open
Abstract
Lung cancer remains the second most commonly diagnosed cancer worldwide and the leading cause of cancer-related mortality. The mapping of genomic alterations and their role in lung-cancer progression has been followed by the development of new therapeutic options. Several novel drugs, such as targeted therapy and immunotherapy, have significantly improved outcomes. However, many patients with lung cancer do not benefit from existing therapies or develop progressive disease, leading to increased morbidity and mortality despite initial responses to treatment. Alterations in DNA-damage repair (DDR) genes represent a cancer hallmark that impairs a cell’s ability to prevent deleterious mutation accumulation and repair. These alterations have recently emerged as a therapeutic target in breast, ovarian, prostate, and pancreatic cancers. The role of DDR alterations remains largely unknown in lung cancer. Nevertheless, recent research efforts have highlighted a potential role of some DDR alterations as predictive biomarkers of response to treatment. Despite the failure of PARP inhibitors (main class of DDR targeting agents) to improve outcomes in lung cancer patients, there is some evidence suggesting a role of PARP inhibitors and other DDR targeting agents in benefiting a distinct subset of lung cancer patients. In this review, we will discuss the existing literature on DDR alterations and homologous recombination deficiency (HRD) state as predictive biomarkers and therapeutic targets in both non-small cell lung and small cell lung cancer.
Collapse
|
9
|
Brown KH, Ghita M, Dubois LJ, de Ruysscher D, Prise KM, Verhaegen F, Butterworth KT. A scoping review of small animal image-guided radiotherapy research: Advances, impact and future opportunities in translational radiobiology. Clin Transl Radiat Oncol 2022; 34:112-119. [PMID: 35496817 PMCID: PMC9046563 DOI: 10.1016/j.ctro.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/04/2022] [Indexed: 11/18/2022] Open
Abstract
Background and purpose To provide a scoping review of published studies using small animal irradiators and highlight the progress in preclinical radiotherapy (RT) studies enabled by these platforms since their development and commercialization in 2007. Materials and methods PubMed searches and manufacturer records were used to identify 907 studies that were screened with 359 small animal RT studies included in the analyses. These articles were classified as biology or physics contributions and into subgroups based on research aims, experimental models and other parameters to identify trends in the preclinical RT research landscape. Results From 2007 to 2021, most published articles were biology contributions (62%) whilst physics contributions accounted for 38% of the publications. The main research areas of physics articles were in dosimetry and calibration (24%), treatment planning and simulation (22%), and imaging (22%) and the studies predominantly used phantoms (41%) or in vivo models (34%). The majority of biology contributions were tumor studies (69%) with brain being the most commonly investigated site. The most frequently investigated areas of tumor biology were evaluating radiosensitizers (33%), model development (30%) and imaging (21%) with cell-line derived xenografts the most common model (82%). 31% of studies focused on normal tissue radiobiology and the lung was the most investigated site. Conclusions This study captures the trends in preclinical RT research using small animal irradiators from 2007 to 2021. Our data show the increased uptake and outputs from preclinical RT studies in important areas of biology and physics research that could inform translation to clinical trials.
Collapse
Affiliation(s)
- Kathryn H. Brown
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, United Kingdom
- Corresponding author at: Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom.
| | - Mihaela Ghita
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, United Kingdom
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW – School for Oncology, Maastricht University, Maastricht, The Netherlands
| | - Dirk de Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Kevin M. Prise
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, United Kingdom
| | - Frank Verhaegen
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Karl T. Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
10
|
Wang LW, Jiang S, Yuan YH, Duan J, Mao ND, Hui Z, Bai R, Xie T, Ye XY. Recent Advances in Synergistic Antitumor Effects Exploited from the Inhibition of Ataxia Telangiectasia and RAD3-Related Protein Kinase (ATR). Molecules 2022; 27:molecules27082491. [PMID: 35458687 PMCID: PMC9029554 DOI: 10.3390/molecules27082491] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/27/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
As one of the key phosphatidylinositol 3-kinase-related kinases (PIKKs) family members, ataxia telangiectasia and RAD3-related protein kinase (ATR) is crucial in maintaining mammalian cell genomic integrity in DNA damage response (DDR) and repair pathways. Dysregulation of ATR has been found across different cancer types. In recent years, the inhibition of ATR has been proven to be effective in cancer therapy in preclinical and clinical studies. Importantly, tumor-specific alterations such as ATM loss and Cyclin E1 (CCNE1) amplification are more sensitive to ATR inhibition and are being exploited in synthetic lethality (SL) strategy. Besides SL, synergistic anticancer effects involving ATRi have been reported in an increasing number in recent years. This review focuses on the recent advances in different forms of synergistic antitumor effects, summarizes the pharmacological benefits and ongoing clinical trials behind the biological mechanism, and provides perspectives for future challenges and opportunities. The hope is to draw awareness to the community that targeting ATR should have great potential in developing effective anticancer medicines.
Collapse
Affiliation(s)
- Li-Wei Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Songwei Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Ying-Hui Yuan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Jilong Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Nian-Dong Mao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zi Hui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (L.-W.W.); (S.J.); (Y.-H.Y.); (J.D.); (N.-D.M.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (R.B.); (T.X.); (X.-Y.Y.); Tel.: +86-571-28860236 (X.-Y.Y.)
| |
Collapse
|
11
|
Wilson Z, Odedra R, Wallez Y, Wijnhoven PW, Hughes AM, Gerrard J, Jones GN, Bargh-Dawson H, Brown E, Young LA, O'Connor MJ, Lau A. ATR Inhibitor AZD6738 (Ceralasertib) Exerts Antitumor Activity as a Monotherapy and in Combination with Chemotherapy and the PARP Inhibitor Olaparib. Cancer Res 2022; 82:1140-1152. [PMID: 35078817 PMCID: PMC9359726 DOI: 10.1158/0008-5472.can-21-2997] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/10/2021] [Accepted: 01/19/2022] [Indexed: 01/09/2023]
Abstract
AZD6738 (ceralasertib) is a potent and selective orally bioavailable inhibitor of ataxia telangiectasia and Rad3-related (ATR) kinase. ATR is activated in response to stalled DNA replication forks to promote G2-M cell-cycle checkpoints and fork restart. Here, we found AZD6738 modulated CHK1 phosphorylation and induced ATM-dependent signaling (pRAD50) and the DNA damage marker γH2AX. AZD6738 inhibited break-induced replication and homologous recombination repair. In vitro sensitivity to AZD6738 was elevated in, but not exclusive to, cells with defects in the ATM pathway or that harbor putative drivers of replication stress such as CCNE1 amplification. This translated to in vivo antitumor activity, with tumor control requiring continuous dosing and free plasma exposures, which correlated with induction of pCHK1, pRAD50, and γH2AX. AZD6738 showed combinatorial efficacy with agents associated with replication fork stalling and collapse such as carboplatin and irinotecan and the PARP inhibitor olaparib. These combinations required optimization of dose and schedules in vivo and showed superior antitumor activity at lower doses compared with that required for monotherapy. Tumor regressions required at least 2 days of daily dosing of AZD6738 concurrent with carboplatin, while twice daily dosing was required following irinotecan. In a BRCA2-mutant patient-derived triple-negative breast cancer (TNBC) xenograft model, complete tumor regression was achieved with 3 to5 days of daily AZD6738 per week concurrent with olaparib. Increasing olaparib dosage or AZD6738 dosing to twice daily allowed complete tumor regression even in a BRCA wild-type TNBC xenograft model. These preclinical data provide rationale for clinical evaluation of AZD6738 as a monotherapy or combinatorial agent. SIGNIFICANCE This detailed preclinical investigation, including pharmacokinetics/pharmacodynamics and dose-schedule optimizations, of AZD6738/ceralasertib alone and in combination with chemotherapy or PARP inhibitors can inform ongoing clinical efforts to treat cancer with ATR inhibitors.
Collapse
Affiliation(s)
- Zena Wilson
- Bioscience, Oncology R&D, AstraZeneca, Cheshire, United Kingdom
| | - Rajesh Odedra
- Bioscience, Oncology R&D, AstraZeneca, Cheshire, United Kingdom
| | - Yann Wallez
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | - Adina M. Hughes
- Bioscience, Oncology R&D, AstraZeneca, Cheshire, United Kingdom
| | - Joe Gerrard
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Gemma N. Jones
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Hannah Bargh-Dawson
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Elaine Brown
- Bioscience, Oncology R&D, AstraZeneca, Cheshire, United Kingdom
| | - Lucy A. Young
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Mark J. O'Connor
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Alan Lau
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, United Kingdom.,Corresponding Author: Alan Lau, Bioscience, Oncology R&D, AstraZeneca, Hodgkin Building, C/O Darwin Building, Unit 310, Cambridge Science Park, Milton Road, Cambridge CB4 OWG, United Kingdom. Phone: 4407-9171-88399; E-mail:
| |
Collapse
|
12
|
Karukonda P, Odhiambo D, Mowery YM. Pharmacologic inhibition of ataxia telangiectasia and Rad3-related (ATR) in the treatment of head and neck squamous cell carcinoma. Mol Carcinog 2022; 61:225-238. [PMID: 34964992 PMCID: PMC8799519 DOI: 10.1002/mc.23384] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 02/03/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) poses significant treatment challenges, with high recurrence rates for locally advanced disease despite aggressive therapy typically involving a combination of surgery, radiation therapy, and/or chemotherapy. HNSCCs commonly exhibit reduced or absent TP53 function due to genomic alterations or human papillomavirus (HPV) infection, leading to dependence on the S- and G2/M checkpoints for cell cycle regulation. Both of these checkpoints are activated by Ataxia Telangiectasia and Rad3-related (ATR), which tends to be overexpressed in HNSCC relative to adjacent normal tissues and represents a potentially promising therapeutic target, particularly in combination with other treatments. ATR is a DNA damage signaling kinase that is activated in response to replication stress and single-stranded DNA breaks, such as those induced by radiation therapy and certain chemotherapies. ATR kinase inhibitors are currently being investigated in several clinical trials as part of the management of locally advanced, recurrent, or metastatic HNSCC, along with other malignancies. In this review article, we summarize the rationale and preclinical data supporting incorporation of ATR inhibition into therapeutic regimens for HNSCC.
Collapse
Affiliation(s)
- Pooja Karukonda
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Diana Odhiambo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA,Department of Head and Neck Surgery & Communication Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
13
|
Petroni G, Cantley LC, Santambrogio L, Formenti SC, Galluzzi L. Radiotherapy as a tool to elicit clinically actionable signalling pathways in cancer. Nat Rev Clin Oncol 2022; 19:114-131. [PMID: 34819622 PMCID: PMC9004227 DOI: 10.1038/s41571-021-00579-w] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 02/03/2023]
Abstract
A variety of targeted anticancer agents have been successfully introduced into clinical practice, largely reflecting their ability to inhibit specific molecular alterations that are required for disease progression. However, not all malignant cells rely on such alterations to survive, proliferate, disseminate and/or evade anticancer immunity, implying that many tumours are intrinsically resistant to targeted therapies. Radiotherapy is well known for its ability to activate cytotoxic signalling pathways that ultimately promote the death of cancer cells, as well as numerous cytoprotective mechanisms that are elicited by cellular damage. Importantly, many cytoprotective mechanisms elicited by radiotherapy can be abrogated by targeted anticancer agents, suggesting that radiotherapy could be harnessed to enhance the clinical efficacy of these drugs. In this Review, we discuss preclinical and clinical data that introduce radiotherapy as a tool to elicit or amplify clinically actionable signalling pathways in patients with cancer.
Collapse
Affiliation(s)
- Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lewis C Cantley
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Bertho A, Dos Santos M, Braga-Cohen S, Buard V, Paget V, Guipaud O, Tarlet G, Milliat F, François A. Preclinical Model of Stereotactic Ablative Lung Irradiation Using Arc Delivery in the Mouse: Is Fractionation Worthwhile? Front Med (Lausanne) 2022; 8:794324. [PMID: 35004768 PMCID: PMC8739220 DOI: 10.3389/fmed.2021.794324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022] Open
Abstract
Lung stereotactic body radiation therapy is characterized by a reduction in target volumes and the use of severely hypofractionated schedules. Preclinical modeling became possible thanks to rodent-dedicated irradiation devices allowing accurate beam collimation and focal lung exposure. Given that a great majority of publications use single dose exposures, the question we asked in this study was as follows: in incremented preclinical models, is it worth using fractionated protocols or should we continue focusing solely on volume limitation? The left lungs of C57BL/6JRj mice were exposed to ionizing radiation using arc therapy and 3 × 3 mm beam collimation. Three-fraction schedules delivered over a period of 1 week were used with 20, 28, 40, and 50 Gy doses per fraction. Lung tissue opacification, global histological damage and the numbers of type II pneumocytes and club cells were assessed 6 months post-exposure, together with the gene expression of several lung cells and inflammation markers. Only the administration of 3 × 40 Gy or 3 × 50 Gy generated focal lung fibrosis after 6 months, with tissue opacification visible by cone beam computed tomography, tissue scarring and consolidation, decreased club cell numbers and a reactive increase in the number of type II pneumocytes. A fractionation schedule using an arc-therapy-delivered three fractions/1 week regimen with 3 × 3 mm beam requires 40 Gy per fraction for lung fibrosis to develop within 6 months, a reasonable time lapse given the mouse lifespan. A comparison with previously published laboratory data suggests that, in this focal lung irradiation configuration, administering a Biological Effective Dose ≥ 1000 Gy should be recommended to obtain lung fibrosis within 6 months. The need for such a high dose per fraction challenges the appropriateness of using preclinical highly focused fractionation schedules in mice.
Collapse
Affiliation(s)
- Annaïg Bertho
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Morgane Dos Santos
- Laboratory of Radiobiology of Accidental Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Sarah Braga-Cohen
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Valérie Buard
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Vincent Paget
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Olivier Guipaud
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Georges Tarlet
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Fabien Milliat
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Agnès François
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| |
Collapse
|
15
|
Rødland GE, Hauge S, Hasvold G, Bay LTE, Raabe TTH, Joel M, Syljuåsen RG. Differential Effects of Combined ATR/WEE1 Inhibition in Cancer Cells. Cancers (Basel) 2021; 13:cancers13153790. [PMID: 34359691 PMCID: PMC8345075 DOI: 10.3390/cancers13153790] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 07/13/2021] [Indexed: 01/14/2023] Open
Abstract
Simple Summary Cancer cells often show elevated replication stress and loss of cell cycle checkpoints. The ataxia telangiectasia and Rad3-related (ATR) and WEE1 kinases play roles in protecting cancer cells from high replication stress and in regulating the remaining cell cycle checkpoints. Inhibitors of ATR or WEE1 therefore have the potential to selectively kill cancer cells and are currently being tested in clinical trials. However, more studies are needed to understand how these inhibitors work in various types of cancer and to find the most effective ways of using them. Here, we have explored whether simultaneous treatment with ATR and WEE1 inhibitors is a promising approach. Effects were investigated in cell lines from osteosarcoma and lung cancer. We expect our results to be of importance for future treatment strategies with these inhibitors. Abstract Inhibitors of WEE1 and ATR kinases are considered promising for cancer treatment, either as monotherapy or in combination with chemo- or radiotherapy. Here, we addressed whether simultaneous inhibition of WEE1 and ATR might be advantageous. Effects of the WEE1 inhibitor MK1775 and ATR inhibitor VE822 were investigated in U2OS osteosarcoma cells and in four lung cancer cell lines, H460, A549, H1975, and SW900, with different sensitivities to the WEE1 inhibitor. Despite the differences in cytotoxic effects, the WEE1 inhibitor reduced the inhibitory phosphorylation of CDK, leading to increased CDK activity accompanied by ATR activation in all cell lines. However, combining ATR inhibition with WEE1 inhibition could not fully compensate for cell resistance to the WEE1 inhibitor and reduced cell viability to a variable extent. The decreased cell viability upon the combined treatment correlated with a synergistic induction of DNA damage in S-phase in U2OS cells but not in the lung cancer cells. Moreover, less synergy was found between ATR and WEE1 inhibitors upon co-treatment with radiation, suggesting that single inhibitors may be preferable together with radiotherapy. Altogether, our results support that combining WEE1 and ATR inhibitors may be beneficial for cancer treatment in some cases, but also highlight that the effects vary between cancer cell lines.
Collapse
|
16
|
Goff PH, Bhakuni R, Pulliam T, Lee JH, Hall ET, Nghiem P. Intersection of Two Checkpoints: Could Inhibiting the DNA Damage Response Checkpoint Rescue Immune Checkpoint-Refractory Cancer? Cancers (Basel) 2021; 13:3415. [PMID: 34298632 PMCID: PMC8307089 DOI: 10.3390/cancers13143415] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022] Open
Abstract
Metastatic cancers resistant to immunotherapy require novel management strategies. DNA damage response (DDR) proteins, including ATR (ataxia telangiectasia and Rad3-related), ATM (ataxia telangiectasia mutated) and DNA-PK (DNA-dependent protein kinase), have been promising therapeutic targets for decades. Specific, potent DDR inhibitors (DDRi) recently entered clinical trials. Surprisingly, preclinical studies have now indicated that DDRi may stimulate anti-tumor immunity to augment immunotherapy. The mechanisms governing how DDRi could promote anti-tumor immunity are not well understood; however, early evidence suggests that they can potentiate immunogenic cell death to recruit and activate antigen-presenting cells to prime an adaptive immune response. Merkel cell carcinoma (MCC) is well suited to test these concepts. It is inherently immunogenic as ~50% of patients with advanced MCC persistently benefit from immunotherapy, making MCC one of the most responsive solid tumors. As is typical of neuroendocrine cancers, dysfunction of p53 and Rb with upregulation of Myc leads to the very rapid growth of MCC. This suggests high replication stress and susceptibility to DDRi and DNA-damaging agents. Indeed, MCC tumors are particularly radiosensitive. Given its inherent immunogenicity, cell cycle checkpoint deficiencies and sensitivity to DNA damage, MCC may be ideal for testing whether targeting the intersection of the DDR checkpoint and the immune checkpoint could help patients with immunotherapy-refractory cancers.
Collapse
Affiliation(s)
- Peter H. Goff
- Department of Radiation Oncology, University of Washington, Seattle, WA 98195, USA;
| | - Rashmi Bhakuni
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.B.); (T.P.); (J.H.L.)
| | - Thomas Pulliam
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.B.); (T.P.); (J.H.L.)
| | - Jung Hyun Lee
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.B.); (T.P.); (J.H.L.)
- Institute for Stem Cell and Regenerative Medicine, Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Evan T. Hall
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98109, USA;
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Paul Nghiem
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.B.); (T.P.); (J.H.L.)
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
17
|
Liu L, Chen Y, Huang Y, Cao K, Liu T, Shen H, Cui J, Li B, Cai J, Gao F, Yang Y. Long non-coding RNA ANRIL promotes homologous recombination-mediated DNA repair by maintaining ATR protein stability to enhance cancer resistance. Mol Cancer 2021; 20:94. [PMID: 34225755 PMCID: PMC8256557 DOI: 10.1186/s12943-021-01382-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 05/31/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Lei Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China.,Department of Oncology, General Hospital of Central Theater Command of Chinese People's Liberation Army, No.627 Wuluo Road, Wuchang District, Wuhan, Hubei, 430070, P. R. China
| | - Yuanyuan Chen
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China
| | - Yijuan Huang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China
| | - Kun Cao
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China
| | - Tingting Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China
| | - Hui Shen
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China
| | - Jianguo Cui
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China
| | - Bailong Li
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China
| | - Jianming Cai
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China. .,School of Public Health and Management, Wenzhou Medical University, University Town, Wenzhou, Zhejiang, P. R. China.
| | - Fu Gao
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China.
| | - Yanyong Yang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, P. R. China.
| |
Collapse
|
18
|
Davison C, Morelli R, Knowlson C, McKechnie M, Carson R, Stachtea X, McLaughlin KA, Prise VE, Savage K, Wilson RH, Mulligan KA, Wilson PM, Ladner RD, LaBonte MJ. Targeting nucleotide metabolism enhances the efficacy of anthracyclines and anti-metabolites in triple-negative breast cancer. NPJ Breast Cancer 2021; 7:38. [PMID: 33824328 PMCID: PMC8024381 DOI: 10.1038/s41523-021-00245-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) remains the most lethal breast cancer subtype with poor response rates to the current chemotherapies and a lack of additional effective treatment options. We have identified deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase) as a critical gatekeeper that protects tumour DNA from the genotoxic misincorporation of uracil during treatment with standard chemotherapeutic agents commonly used in the FEC regimen. dUTPase catalyses the hydrolytic dephosphorylation of deoxyuridine triphosphate (dUTP) to deoxyuridine monophosphate (dUMP), providing dUMP for thymidylate synthase as part of the thymidylate biosynthesis pathway and maintaining low intracellular dUTP concentrations. This is crucial as DNA polymerase cannot distinguish between dUTP and deoxythymidylate triphosphate (dTTP), leading to dUTP misincorporation into DNA. Targeting dUTPase and inducing uracil misincorporation during the repair of DNA damage induced by fluoropyrimidines or anthracyclines represents an effective strategy to induce cell lethality. dUTPase inhibition significantly sensitised TNBC cell lines to fluoropyrimidines and anthracyclines through imbalanced nucleotide pools and increased DNA damage leading to decreased proliferation and increased cell death. These results suggest that repair of treatment-mediated DNA damage requires dUTPase to prevent uracil misincorporation and that inhibition of dUTPase is a promising strategy to enhance the efficacy of TNBC chemotherapy.
Collapse
Affiliation(s)
- Craig Davison
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Roisin Morelli
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Catherine Knowlson
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Melanie McKechnie
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Robbie Carson
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Xanthi Stachtea
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | | | | | - Kienan Savage
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Richard H Wilson
- Translational Research Centre, University of Glasgow, Glasgow, UK
| | | | | | - Robert D Ladner
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Melissa J LaBonte
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK.
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
19
|
Effect of ATR Inhibition in RT Response of HPV-Negative and HPV-Positive Head and Neck Cancers. Int J Mol Sci 2021; 22:ijms22041504. [PMID: 33546122 PMCID: PMC7913134 DOI: 10.3390/ijms22041504] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Radiotherapy (RT) has a central role in head and neck squamous cell carcinoma (HNSCC) treatment. Targeted therapies modulating DNA damage response (DDR) and more specific cell cycle checkpoints can improve the radiotherapeutic response. Here, we assessed the influence of ataxia-telangiectasia mutated and Rad3-related (ATR) inhibition with the ATR inhibitor AZD6738 on RT response in both human papillomavirus (HPV)-negative and HPV-positive HNSCC. We found that ATR inhibition enhanced RT response in HPV-negative and HPV-positive cell lines independent of HPV status. The radiosensitizing effect of AZD6738 was correlated with checkpoint kinase 1 (CHK1)-mediated abrogation of G2/M-arrest. This resulted in the inhibition of RT-induced DNA repair and in an increase in the percentage of micronucleated cells. We validated the enhanced RT response in HPV-negative and HPV-positive xenograft models. These data demonstrate the potential use of ATR inhibition in combination with RT as a treatment option for both HPV-negative and HPV-positive HNSCC patients.
Collapse
|
20
|
Walls GM, Oughton JB, Chalmers AJ, Brown S, Collinson F, Forster MD, Franks KN, Gilbert A, Hanna GG, Hannaway N, Harrow S, Haswell T, Hiley CT, Hinsley S, Krebs M, Murden G, Phillip R, Ryan AJ, Salem A, Sebag-Montefoire D, Shaw P, Twelves CJ, Walker K, Young RJ, Faivre-Finn C, Greystoke A. CONCORDE: A phase I platform study of novel agents in combination with conventional radiotherapy in non-small-cell lung cancer. Clin Transl Radiat Oncol 2020; 25:61-66. [PMID: 33072895 PMCID: PMC7548952 DOI: 10.1016/j.ctro.2020.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
Lung cancer is the leading cause of cancer mortality worldwide and most patients are unsuitable for 'gold standard' treatment, which is concurrent chemoradiotherapy. CONCORDE is a platform study seeking to establish the toxicity profiles of multiple novel radiosensitisers targeting DNA repair proteins in patients treated with sequential chemoradiotherapy. Time-to-event continual reassessment will facilitate efficient dose-finding.
Collapse
Key Words
- ATM, Ataxia telangiectasia mutated
- ATR, Ataxia telangiectasia and Rad3 related
- CRT, Chemoradiotherapy
- CT, Computed tomography
- CTCAE, Common terminology criteria for adverse events
- CTRad, Clinical and Translational Radiotherapy Research Working Group
- Continual reassessment method
- DDRi, DNA damage response inhibitor
- DLT, Dose limiting toxicity
- DNA damage repair inhibitor
- DNA, Deoxyribonucleic acid
- DNA-PK, DNA-dependent protein kinase
- ECOG, Eastern Cooperative Oncology Group
- EORTC, European Organisation for Research and Treatment of Cancer
- ICRU, International Commission on Radiation Units and Measurements
- IMPs, Investigational medicinal products
- LA, Locally advanced
- MRC, Medical Research Council
- NCRI, National Cancer Research Institute
- NSCLC, Non-small cell lung cancer
- Non-small cell lung cancer
- PARP, Poly (ADP-ribose) polymerase
- PET, Positron emission tomography
- PFS, Progression free survival
- PROMs, Patient-reported outcome measures
- Platform trial
- RECIST, Response evaluation criteria in solid tumours
- RP2D, Recommended phase II dose
- RT, Radiotherapy
- SACT, Systemic anti-cancer therapy
- SRC, Safety review committee
- Sequential chemoradiotherapy
- TNM, Tumour node metastasis
- TiTE-CRM, Time to event continual reassessment method
- cfDNA, Cell-free DNA
Collapse
Affiliation(s)
- Gerard M. Walls
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Northern Ireland, UK
| | - Jamie B. Oughton
- Leeds Institute of Clinical Trials Research, University of Leeds, England, UK
| | | | - Sarah Brown
- Leeds Institute of Clinical Trials Research, University of Leeds, England, UK
| | - Fiona Collinson
- Leeds Institute of Clinical Trials Research, University of Leeds, England, UK
| | | | - Kevin N. Franks
- St James’ Institute of Oncology, University of Leeds, England, UK
| | | | - Gerard G. Hanna
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Australia
| | | | - Stephen Harrow
- The Beatson West of Scotland Cancer Centre, Glasgow, Scotland, UK
| | | | | | - Samantha Hinsley
- Leeds Institute of Clinical Trials Research, University of Leeds, England, UK
- Institute of Cancer Sciences, University of Glasgow, Scotland, UK
| | - Matthew Krebs
- Faculty of Biology, Medicine and Health, University of Manchester, England, UK
| | - Geraldine Murden
- Leeds Institute of Clinical Trials Research, University of Leeds, England, UK
| | - Rachel Phillip
- Leeds Institute of Clinical Trials Research, University of Leeds, England, UK
| | - Anderson J. Ryan
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, England, UK
| | - Ahmed Salem
- The Christie NHS Foundation Trust/University of Manchester, Manchester, England, UK
| | | | - Paul Shaw
- Velindre University NHS Trust, Cardiff, Wales, UK
| | - Chris J. Twelves
- St James’ Institute of Oncology, University of Leeds, England, UK
| | - Katrina Walker
- Leeds Institute of Clinical Trials Research, University of Leeds, England, UK
| | - Robin J. Young
- Academic Unit of Clinical Oncology, Weston Park Hospital, Sheffield, England, UK
| | - Corinne Faivre-Finn
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, England, UK
| | | |
Collapse
|
21
|
Li H, Che J, Jiang M, Cui M, Feng G, Dong J, Zhang S, Lu L, Liu W, Fan S. CLPTM1L induces estrogen receptor β signaling-mediated radioresistance in non-small cell lung cancer cells. Cell Commun Signal 2020; 18:152. [PMID: 32943060 PMCID: PMC7499972 DOI: 10.1186/s12964-020-00571-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/01/2020] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Radioresistance is a major challenge in lung cancer radiotherapy, and new radiosensitizers are urgently needed. Estrogen receptor β (ERβ) is involved in the progression of non-small cell lung cancer (NSCLC), however, the role of ERβ in the response to radiotherapy in lung cancer remains elusive. In the present study, we investigated the mechanism underlying ERβ-mediated transcriptional activation and radioresistance of NSCLC cells. METHODS Quantitative real-time PCR, western blot and immunohistochemistry were used to detect the expression of CLPTM1L, ERβ and other target genes. The mechanism of CLPTM1L in modulation of radiosensitivity was investigated by chromatin immunoprecipitation assay, luciferase reporter gene assay, immunofluorescence staining, confocal microscopy, coimmunoprecipitation and GST pull-down assays. The functional role of CLPTM1L was detected by function assays in vitro and in vivo. RESULTS CLPTM1L expression was negatively correlated with the radiosensitivity of NSCLC cell lines, and irradiation upregulated CLPTM1L in radioresistant (A549) but not in radiosensitive (H460) NSCLC cells. Meanwhile, IR induced the translocation of CLPTM1L from the cytoplasm into the nucleus in NSCLC cells. Moreover, CLPTM1L induced radioresistance in NSCLC cells. iTRAQ-based analysis and cDNA microarray identified irradiation-related genes commonly targeted by CLPTM1L and ERβ, and CLPTM1L upregulated ERβ-induced genes CDC25A, c-Jun, and BCL2. Mechanistically, CLPTM1L coactivated ERβ by directly interacting with ERβ through the LXXLL NR (nuclear receptor)-binding motif. Functionally, ERβ silencing was sufficient to block CLPTM1L-enhanced radioresistance of NSCLC cells in vitro. CLPTM1L shRNA treatment in combination with irradiation significantly inhibited cancer cell growth in NSCLC xenograft tumors in vivo. CONCLUSIONS The present results indicate that CLPTM1L acts as a critical coactivator of ERβ to promote the transcription of its target genes and induce radioresistance of NSCLC cells, suggesting a new target for radiosensitization in NSCLC therapy. Video Abstract.
Collapse
Affiliation(s)
- Hang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| | - Jun Che
- grid.459328.10000 0004 1758 9149Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, 200 Hui-He Road, Wuxi, 214062 Jiangsu P.R. China
| | - Mian Jiang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| | - Guoxing Feng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| | - Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| | - Shuqin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| | - Lu Lu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| | - Weili Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Bai-Di Road, Tianjin, 300192 P.R. China
| |
Collapse
|
22
|
Targeting the DNA Damage Response for Radiosensitization. CANCER DRUG DISCOVERY AND DEVELOPMENT 2020. [DOI: 10.1007/978-3-030-49701-9_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Goundry WRF, Dai K, Gonzalez M, Legg D, O’Kearney-McMullan A, Morrison J, Stark A, Siedlecki P, Tomlin P, Yang J. Development and Scale-up of a Route to ATR Inhibitor AZD6738. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.9b00075] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- William R. F. Goundry
- Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield SK102NA, United Kingdom
| | - Kuangchu Dai
- Changzhou SynTheAll Pharmaceutical Co. Ltd, No 589, North Yulong Road, Chunjiang Town, Xinbei
District, Changzhou 213127, Jiangsu P. R. China
| | - Miguel Gonzalez
- Asymchem Laboratories (Tianjin) Co. Ltd, No. 6 Dongting, Third Avenue, TEDA, Tianjin, 300457, P. R. China
| | - Daniel Legg
- Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield SK102NA, United Kingdom
| | - Anne O’Kearney-McMullan
- Pharmaceutical Technology and Development, Operations, AstraZeneca, Macclesfield SK102NA, United Kingdom
| | - James Morrison
- Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield SK102NA, United Kingdom
| | - Andrew Stark
- Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield SK102NA, United Kingdom
| | - Paul Siedlecki
- Pharmaceutical Technology and Development, Operations, AstraZeneca, Macclesfield SK102NA, United Kingdom
| | - Paula Tomlin
- Pharmaceutical Technology and Development, Operations, AstraZeneca, Macclesfield SK102NA, United Kingdom
| | - Jianbo Yang
- Asymchem Laboratories (Tianjin) Co. Ltd, No. 6 Dongting, Third Avenue, TEDA, Tianjin, 300457, P. R. China
| |
Collapse
|
24
|
Dillon MT, Bergerhoff KF, Pedersen M, Whittock H, Crespo-Rodriguez E, Patin EC, Pearson A, Smith HG, Paget JTE, Patel RR, Foo S, Bozhanova G, Ragulan C, Fontana E, Desai K, Wilkins AC, Sadanandam A, Melcher A, McLaughlin M, Harrington KJ. ATR Inhibition Potentiates the Radiation-induced Inflammatory Tumor Microenvironment. Clin Cancer Res 2019; 25:3392-3403. [PMID: 30770349 PMCID: PMC6551222 DOI: 10.1158/1078-0432.ccr-18-1821] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/09/2018] [Accepted: 02/11/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE ATR inhibitors (ATRi) are in early phase clinical trials and have been shown to sensitize to chemotherapy and radiotherapy preclinically. Limited data have been published about the effect of these drugs on the tumor microenvironment.Experimental Design: We used an immunocompetent mouse model of HPV-driven malignancies to investigate the ATR inhibitor AZD6738 in combination with fractionated radiation (RT). Gene expression analysis and flow cytometry were performed posttherapy. RESULTS Significant radiosensitization to RT by ATRi was observed alongside a marked increase in immune cell infiltration. We identified increased numbers of CD3+ and NK cells, but most of this infiltrate was composed of myeloid cells. ATRi plus radiation produced a gene expression signature matching a type I/II IFN response, with upregulation of genes playing a role in nucleic acid sensing. Increased MHC I levels were observed on tumor cells, with transcript-level data indicating increased antigen processing and presentation within the tumor. Significant modulation of cytokine gene expression (particularly CCL2, CCL5, and CXCL10) was found in vivo, with in vitro data indicating CCL3, CCL5, and CXCL10 are produced from tumor cells after ATRi + RT. CONCLUSIONS We show that DNA damage by ATRi and RT leads to an IFN response through activation of nucleic acid-sensing pathways. This triggers increased antigen presentation and innate immune cell infiltration. Further understanding of the effect of this combination on the immune response may allow modulation of these effects to maximize tumor control through antitumor immunity.
Collapse
Affiliation(s)
| | | | - Malin Pedersen
- The Institute of Cancer Research, London, United Kingdom
| | | | | | | | - Alex Pearson
- The Institute of Cancer Research, London, United Kingdom
| | - Henry G Smith
- The Institute of Cancer Research, London, United Kingdom
| | | | | | - Shane Foo
- The Institute of Cancer Research, London, United Kingdom
| | | | | | - Elisa Fontana
- The Institute of Cancer Research, London, United Kingdom
| | - Krisha Desai
- The Institute of Cancer Research, London, United Kingdom
| | - Anna C Wilkins
- The Institute of Cancer Research, London, United Kingdom
| | | | - Alan Melcher
- The Institute of Cancer Research, London, United Kingdom
| | | | | |
Collapse
|
25
|
Evolution of the Supermodel: Progress in Modelling Radiotherapy Response in Mice. Clin Oncol (R Coll Radiol) 2019; 31:272-282. [PMID: 30871751 DOI: 10.1016/j.clon.2019.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
Mouse models are essential tools in cancer research that have been used to understand the genetic basis of tumorigenesis, cancer progression and to test the efficacies of anticancer treatments including radiotherapy. They have played a critical role in our understanding of radiotherapy response in tumours and normal tissues and continue to evolve to better recapitulate the underlying biology of humans. In addition, recent developments in small animal irradiators have significantly improved in vivo irradiation techniques, allowing previously unimaginable experimental approaches to be explored in the laboratory. The combination of contemporary mouse models with small animal irradiators represents a major step forward for the radiobiology field in being able to much more accurately replicate clinical exposure scenarios. As radiobiology studies become ever more sophisticated in reflecting developments in the clinic, it is increasingly important to understand the basis and potential limitations of extrapolating data from mice to humans. This review provides an overview of mouse models and small animal radiotherapy platforms currently being used as advanced radiobiological research tools towards improving the translational power of preclinical studies.
Collapse
|
26
|
Ghita M, Dunne V, Hanna GG, Prise KM, Williams JP, Butterworth KT. Preclinical models of radiation-induced lung damage: challenges and opportunities for small animal radiotherapy. Br J Radiol 2019; 92:20180473. [PMID: 30653332 DOI: 10.1259/bjr.20180473] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Despite a major paradigm shift in radiotherapy planning and delivery over the past three decades with continuing refinements, radiation-induced lung damage (RILD) remains a major dose limiting toxicity in patients receiving thoracic irradiations. Our current understanding of the biological processes involved in RILD which includes DNA damage, inflammation, senescence and fibrosis, is based on clinical observations and experimental studies in mouse models using conventional radiation exposures. Whilst these studies have provided vital information on the pulmonary radiation response, the current implementation of small animal irradiators is enabling refinements in the precision and accuracy of dose delivery to mice which can be applied to studies of RILD. This review presents the current landscape of preclinical studies in RILD using small animal irradiators and highlights the challenges and opportunities for the further development of this emerging technology in the study of normal tissue damage in the lung.
Collapse
Affiliation(s)
- Mihaela Ghita
- 1 Centre for Cancer Research and Cell Biology, Queen's University Belfast , Belfast , Northern Ireland, UK
| | - Victoria Dunne
- 1 Centre for Cancer Research and Cell Biology, Queen's University Belfast , Belfast , Northern Ireland, UK
| | - Gerard G Hanna
- 1 Centre for Cancer Research and Cell Biology, Queen's University Belfast , Belfast , Northern Ireland, UK.,2 Northern Ireland Cancer Centre, Belfast City Hospital , Belfast , Northern Ireland, UK
| | - Kevin M Prise
- 1 Centre for Cancer Research and Cell Biology, Queen's University Belfast , Belfast , Northern Ireland, UK
| | - Jaqueline P Williams
- 3 University of Rochester Medical Centre, University of Rochester , Rochester , USA
| | - Karl T Butterworth
- 1 Centre for Cancer Research and Cell Biology, Queen's University Belfast , Belfast , Northern Ireland, UK
| |
Collapse
|
27
|
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
|
28
|
Integrating Small Animal Irradiators withFunctional Imaging for Advanced Preclinical Radiotherapy Research. Cancers (Basel) 2019; 11:cancers11020170. [PMID: 30717307 PMCID: PMC6406472 DOI: 10.3390/cancers11020170] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/23/2019] [Accepted: 01/29/2019] [Indexed: 12/16/2022] Open
Abstract
Translational research aims to provide direct support for advancing novel treatment approaches in oncology towards improving patient outcomes. Preclinical studies have a central role in this process and the ability to accurately model biological and physical aspects of the clinical scenario in radiation oncology is critical to translational success. The use of small animal irradiators with disease relevant mouse models and advanced in vivo imaging approaches offers unique possibilities to interrogate the radiotherapy response of tumors and normal tissues with high potential to translate to improvements in clinical outcomes. The present review highlights the current technology and applications of small animal irradiators, and explores how these can be combined with molecular and functional imaging in advanced preclinical radiotherapy research.
Collapse
|
29
|
Lavigne J, Suissa A, Verger N, Dos Santos M, Benadjaoud M, Mille-Hamard L, Momken I, Soysouvanh F, Buard V, Guipaud O, Paget V, Tarlet G, Milliat F, François A. Lung Stereotactic Arc Therapy in Mice: Development of Radiation Pneumopathy and Influence of HIF-1α Endothelial Deletion. Int J Radiat Oncol Biol Phys 2019; 104:279-290. [PMID: 30703512 DOI: 10.1016/j.ijrobp.2019.01.081] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/10/2019] [Accepted: 01/21/2019] [Indexed: 01/24/2023]
Abstract
PURPOSE Stereotactic body radiation therapy offers good lung local tumor control by the administration of a high dose per fraction in small volumes. Stereotactic body radiation therapy preclinical modeling is now possible, and our aim was to develop a model of focal irradiation of the mouse lung and to investigate the impact of conditional hypoxia-inducible factor 1α (HIF-1α) deletion in the endothelium on radiation-induced tissue damage. METHODS AND MATERIALS The Small Animal Radiation Research Platform was used to create a mouse model of focal irradiation of the lung using arc therapy. HIF-1α conditional deletion was obtained by crossing mice expressing Cre recombinase under the endothelial promoter VE-cadherin (VECad-Cre+/+ mice) with HIF-1α floxed mice. RESULTS Lung stereotactic arc therapy allows thoracic wall sparing and long-term studies. However, isodose curves showed that neighboring organs received significant doses of radiation, as revealed by ipsilateral lung acute red hepatization and major gene expression level modifications. Conditional HIF-1α deletion reduced acute lung edema and tended to diminish neutrophil infiltrate, but it had no impact on long-term global tissue damage. CONCLUSIONS Arc therapy for focal high-dose irradiation of mouse lung is an efficient model for long-term studies. However, irradiation may have a strong impact on the structure and function of neighboring organs, which must be considered. HIF-1α conditional deletion has no beneficial impact on lung damage in this irradiation schedule.
Collapse
Affiliation(s)
- Jérémy Lavigne
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France; Sorbonne Université, Collège Doctoral, Paris, France
| | - Alexandra Suissa
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Nicolas Verger
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Morgane Dos Santos
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Accidentelles, Fontenay-aux-Roses, France
| | - Mohamedamine Benadjaoud
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Fontenay-aux-Roses, France
| | - Laurence Mille-Hamard
- Unité de Biologie Intégrative des Adaptations à l'Exercice, Université Évry-Val-d'Essonne, Université Paris-Saclay, Evry, France
| | - Iman Momken
- Unité de Biologie Intégrative des Adaptations à l'Exercice, Université Évry-Val-d'Essonne, Université Paris-Saclay, Evry, France
| | - Frédéric Soysouvanh
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France; Sorbonne Université, Collège Doctoral, Paris, France
| | - Valérie Buard
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Olivier Guipaud
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Vincent Paget
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Georges Tarlet
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Fabien Milliat
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Agnès François
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France.
| |
Collapse
|
30
|
Walb MC, Carlson BL, Sarkaria JN, Tryggestad EJ. Quantifying the setup uncertainty of a stereotactic murine micro-image guided radiation therapy system. Br J Radiol 2018; 92:20180487. [PMID: 30299986 DOI: 10.1259/bjr.20180487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE: Investigate the reproducibility of murine cranial positioning using solely a stereotactic stage, and quantify the potential improvements from the on-board image guidance of the X-RAD SmART irradiator. METHODS: For intermouse reproducibility, athymic nude mice (N = 5, ×4 groups) were cranially fixed on a stereotactic stage. Each mouse was imaged via cone-beam CT (CBCT). A virtual isocenter target was placed in the brain, the stage shifted to that target, and the couch positions recorded. The mouse was removed from the stage and this process repeated twice (N=60 measurements). The first acquired CBCT coordinates (within each group of five mice) were used to define "stereotactic couch coordinates." CBCT shifts were calculated to quantify the accuracy of setup based on couch coordinates alone. For intramouse reproducibility, C57BL/6 mice (N=4) were imaged daily for 7 days. Each mouse had individual stereotactic coordinates defined from their first day of CBCT localization, and positional shifts required on the six subsequent days of imaging were quantified (N = 24 measurements). RESULTS: The mean vector shift between stereotactic setup and CBCT alignment for inter and intramouse analysis was 0.78 ± 0.27 mm and 0.82 ± 0.34 mm, respectively. CONCLUSION: Cranial irradiation that can permit positional uncertainties on the order of a millimeter can rely solely on stereotactic coordinates derived from a single daily CBCT. Irradiations of subregions requiring submillimeter accuracy require daily image guidance for each mouse. ADVANCES IN KNOWLEDGE: This is the first investigation of stereotactic reproducibility using the X-RAD SmART and it suggests a method for increased efficiency in high-throughput experiments.
Collapse
Affiliation(s)
- Matthew C Walb
- Department of Radiation Oncology, Mayo Clinic , Rochester , MN
| | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic , Rochester , MN
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic , Rochester , MN
| | | |
Collapse
|
31
|
Foote KM, Nissink JWM, McGuire T, Turner P, Guichard S, Yates JWT, Lau A, Blades K, Heathcote D, Odedra R, Wilkinson G, Wilson Z, Wood CM, Jewsbury PJ. Discovery and Characterization of AZD6738, a Potent Inhibitor of Ataxia Telangiectasia Mutated and Rad3 Related (ATR) Kinase with Application as an Anticancer Agent. J Med Chem 2018; 61:9889-9907. [PMID: 30346772 DOI: 10.1021/acs.jmedchem.8b01187] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The kinase ataxia telangiectasia mutated and rad3 related (ATR) is a key regulator of the DNA-damage response and the apical kinase which orchestrates the cellular processes that repair stalled replication forks (replication stress) and associated DNA double-strand breaks. Inhibition of repair pathways mediated by ATR in a context where alternative pathways are less active is expected to aid clinical response by increasing replication stress. Here we describe the development of the clinical candidate 2 (AZD6738), a potent and selective sulfoximine morpholinopyrimidine ATR inhibitor with excellent preclinical physicochemical and pharmacokinetic (PK) characteristics. Compound 2 was developed improving aqueous solubility and eliminating CYP3A4 time-dependent inhibition starting from the earlier described inhibitor 1 (AZ20). The clinical candidate 2 has favorable human PK suitable for once or twice daily dosing and achieves biologically effective exposure at moderate doses. Compound 2 is currently being tested in multiple phase I/II trials as an anticancer agent.
Collapse
Affiliation(s)
- Kevin M Foote
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - J Willem M Nissink
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Thomas McGuire
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Paul Turner
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Sylvie Guichard
- Bioscience, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - James W T Yates
- DMPK, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - Alan Lau
- Bioscience, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - Kevin Blades
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Dan Heathcote
- Discovery Sciences, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Rajesh Odedra
- Bioscience, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - Gary Wilkinson
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Zena Wilson
- Bioscience, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - Christine M Wood
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Philip J Jewsbury
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| |
Collapse
|