1
|
Liu Y, Xu L, Dou Y, He Y. AXL: shapers of tumor progression and immunosuppressive microenvironments. Mol Cancer 2025; 24:11. [PMID: 39799359 PMCID: PMC11724481 DOI: 10.1186/s12943-024-02210-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/24/2024] [Indexed: 01/15/2025] Open
Abstract
As research progresses, our understanding of the tumor microenvironment (TME) has undergone profound changes. The TME evolves with the developmental stages of cancer and the implementation of therapeutic interventions, transitioning from an immune-promoting to an immunosuppressive microenvironment. Consequently, we focus intently on the significant role of the TME in tumor proliferation, metastasis, and the development of drug resistance. AXL is highly associated with tumor progression; however, previous studies on AXL have been limited to its impact on the biological behavior of cancer cells. An increasing body of research now demonstrates that AXL can influence the function and differentiation of immune cells, mediating immune suppression and thereby fostering tumor growth. A comprehensive analysis to identify and overcome the causes of immunosuppressive microenvironments represents a novel approach to conquering cancer. In this review, we focus on elucidating the role of AXL within the immunosuppressive microenvironments, discussing and analyzing the effects of AXL on tumor cells, T cells, macrophages, natural killer (NK) cells, fibroblasts, and other immune-stromal cells. We aim to clarify the contributions of AXL to the progression and drug resistance of cancer from its functional role in the immune microenvironment.
Collapse
Affiliation(s)
- Yihui Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lei Xu
- Department of Otolaryngology, Southwest Hospital, Army Medical University, Chongqing, 400000, China
| | - Yuanyao Dou
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
2
|
Stevenson L, Cairns L, Li X, Jammula S, Taylor H, Douglas R, McCabe N, Gavory G, Jacq X, Fitzgerald RC, Kennedy RD, Harrison T, Turkington RC. Inhibition of AKT enhances chemotherapy efficacy and synergistically interacts with targeting of the Inhibitor of apoptosis proteins in oesophageal adenocarcinoma. Sci Rep 2024; 14:32121. [PMID: 39739112 PMCID: PMC11686190 DOI: 10.1038/s41598-024-83912-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025] Open
Abstract
The incidence of oesophageal adenocarcinoma (OAC) has risen six-fold in western countries over the last forty years but survival rates have only marginally improved. Hyperactivation of the PI3K-AKT-mTOR pathway is a common occurrence in OAC, driving cell survival, proliferation and resistance to chemotherapeutic agents. Inhibition of AKT has been explored as a treatment strategy with limited success and current inhibitors have failed to progress through clinical trials. Our study, describes a novel allosteric AKT inhibitor, ALM301, and demonstrates an enhancement of the efficacy of conventional chemotherapy when combined with ALM301 in OAC. Reduced sensitivity to ALM301 is associated with high expression of the Inhibitor of Apoptosis (IAP) family of proteins, particularly XIAP. Combined AKT and IAP inhibition synergistically enhanced OAC cell death and successfully re-sensitized ALM301 and chemotherapy resistant cell lines. A high degree of synergism was also observed in patient-derived OAC organoids indicating the potential clinical relevance of the combination. This study demonstrates the role for dual AKT/IAP inhibition in OAC and provides a strong rationale for the further investigation of this highly efficacious combination strategy.
Collapse
Affiliation(s)
- Leanne Stevenson
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland
| | - Lauren Cairns
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland
| | - Xiaodun Li
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Sriganesh Jammula
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Harriet Taylor
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Rosalie Douglas
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland
| | - Niamh McCabe
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland
| | | | - Xavier Jacq
- Almac Discovery Ltd, Craigavon, Northern Ireland
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | | | | | - Richard C Turkington
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland.
| |
Collapse
|
3
|
Scanlon E, Lavery A, Albraikat M, Stevenson L, Kennedy C, Byrne R, Walker A, Mullan-Young B, McManus DT, Virdee PS, Elhussein L, Turbitt J, Collinson D, Miedzybrodzka Z, Van Schaeybroeck S, McQuaid S, James JA, Craig SG, Blayney JK, Petty RD, Harkin DP, Kennedy RD, Eatock MM, Middleton MR, Thomas A, Turkington RC. Immune microenvironment modulation following neoadjuvant therapy for oesophageal adenocarcinoma: a translational analysis of the DEBIOC clinical trial. ESMO Open 2024; 9:103930. [PMID: 39395265 PMCID: PMC11693431 DOI: 10.1016/j.esmoop.2024.103930] [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: 08/21/2024] [Accepted: 09/05/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND The Dual Erb B Inhibition in Oesophago-gastric Cancer (DEBIOC) trial reported an acceptable safety profile for neoadjuvant oxaliplatin and capecitabine (Xelox) ± AZD8931 in oesophageal adenocarcinoma (OAC) but limited efficacy. We evaluated the impact of neoadjuvant Xelox ± AZD8931, a novel small-molecule inhibitor with equipotent activity against epidermal growth factor receptor (EGFR), human epidermal growth factor receptor (HER)2 and HER3, on biological pathways using a unique software-driven solution. PATIENTS AND METHODS Transcriptomic profiles from 25 pre-treatment formalin-fixed paraffin-embedded OAC biopsies and 18 matched resection specimens, treated with Xelox + AZD8931 (n = 16) and Xelox alone (n = 9), were analysed using the Almac claraT total mRNA report analysing 92 gene signatures, 100 unique single-gene drug targets and 7337 single genes across 10 hallmarks of cancer. Gene-set enrichment analysis (GSEA) was utilised to investigate pathways governing pathological response. Tumour-infiltrating lymphocytes (TILs) were assessed digitally using the QuPath software. RESULTS Hierarchical clustering identified three molecular subgroups classified by activation of innate immune signalling. The immune-high subgroup was associated with HER2 positivity, increased pathological response and a marked reduction in immune signalling and TILs following neoadjuvant therapy. The immune-low cluster was predominantly HER2/EGFR-negative, and EGFR positivity was associated with the immune-mixed subgroup. Treatment with neoadjuvant therapy induced common resistance mechanisms, such as angiogenesis and epithelial-mesenchymal transition signalling, and a reduction in DNA repair signatures. Addition of AZD8931 was associated with reduction of expression of EGFR, HER2 and AKT pathways and also promoted an immunosuppressive microenvironment. GSEA showed that patients with a pathological response to treatment had increased immune signalling, whereas non-responders to neoadjuvant therapy were enriched for nucleotide repair and cellular growth through the action of E2F transcription factors. CONCLUSION OAC may be subdivided into three immune-related subgroups which undergo modulation in response to neoadjuvant therapy with marked suppression of the immune microenvironment in HER2-positive/immune-high tumours.
Collapse
Affiliation(s)
- E Scanlon
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - A Lavery
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - M Albraikat
- Precision Medicine Centre of Excellence, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - L Stevenson
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - C Kennedy
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - R Byrne
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - A Walker
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - B Mullan-Young
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - D T McManus
- Department of Pathology, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast
| | - P S Virdee
- Centre for Statistics in Medicine, University of Oxford, Oxford
| | - L Elhussein
- Centre for Statistics in Medicine, University of Oxford, Oxford
| | - J Turbitt
- Medical Genetics, School of Medicine, Medical Sciences, Nutrition and Dentistry, University of Aberdeen, Aberdeen
| | - D Collinson
- Medical Genetics, School of Medicine, Medical Sciences, Nutrition and Dentistry, University of Aberdeen, Aberdeen
| | - Z Miedzybrodzka
- Medical Genetics, School of Medicine, Medical Sciences, Nutrition and Dentistry, University of Aberdeen, Aberdeen
| | - S Van Schaeybroeck
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - S McQuaid
- Precision Medicine Centre of Excellence, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast; Department of Pathology, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast; Northern Ireland Biobank, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - J A James
- Precision Medicine Centre of Excellence, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast; Department of Pathology, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast; Northern Ireland Biobank, The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - S G Craig
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - J K Blayney
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - R D Petty
- Division of Molecular and Clinical Medicine, Ninewells Hospital and School of Medicine, University of Dundee, Dundee
| | - D P Harkin
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast; Almac Diagnostic Services Ltd, Craigavon
| | - R D Kennedy
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast; Almac Diagnostic Services Ltd, Craigavon
| | - M M Eatock
- Northern Ireland Cancer Centre, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast
| | - M R Middleton
- Department of Oncology, University of Oxford, Oxford
| | - A Thomas
- University of Leicester, Leicester, UK
| | - R C Turkington
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast.
| |
Collapse
|
4
|
Hayes RM, O'Donovan TR, McKenna SL. Expression of MxA in esophageal cancer cell lines can influence sensitivity to chemotherapeutic agents but this does not require apoptosis. Cancer Med 2024; 13:e70173. [PMID: 39285636 PMCID: PMC11405456 DOI: 10.1002/cam4.70173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/09/2023] [Accepted: 10/26/2023] [Indexed: 09/20/2024] Open
Abstract
Esophageal cancer is a poor prognosis cancer characterized by intrinsic or acquired resistance to chemotherapeutic agents. The primary determinants of treatment failure are unknown. Expression of an anti-viral protein, myxovirus resistance protein A (MxA) is de-regulated in many cancers, including esophageal cancer, and its activity has been linked to apoptosis. This study has assessed whether MxA expression can influence the response of esophageal cancer cells to the chemotherapeutic agents 5-fluorouracil (5-FU) or oxaliplatin. MxA protein was differentially expressed in a panel of five esophageal cancer cell lines. KYSE450 and KYSE140 cells did not express MxA and were apoptosis incompetent. FLO-1, KYSE270, and OE21 cells expressed MxA, were more drug-sensitive and were apoptosis competent. MxA was artificially overexpressed in cell lines with no endogenous expression (KYSE450 and KYSE140). This increased the resistance of KYSE450 but not KYSE140 cells. Both cell lines remained apoptosis incompetent. We then evaluated siRNA knockdown of MxA in FLO-1 cells and CRISPR knockout in OE21 cells. Knockdown of MxA significantly increased drug sensitivity and caspase-3 activation in FLO-1 cells. OE21-MX1KO cells were also more drug-sensitive, but in contrast to FLO-1 cells, caspase-3 activation was reduced. Collectively these data indicate that MxA can promote resistance to chemotherapy, but this does not always correspond with effects on apoptosis. Effects on apoptosis are cell line specific, suggesting that other co-operating pathways determine the overall impact of MxA. Importantly, in cancer cells that overexpress the protein, drug sensitivity can be improved by interfering with MxA.
Collapse
Affiliation(s)
- R. M. Hayes
- Cancer Research @UCCCollege of Medicine and Health, University College CorkCorkIreland
| | - T. R. O'Donovan
- Cancer Research @UCCCollege of Medicine and Health, University College CorkCorkIreland
| | - S. L. McKenna
- Cancer Research @UCCCollege of Medicine and Health, University College CorkCorkIreland
| |
Collapse
|
5
|
Baxter MA, Spender LC, Cairns D, Walsh S, Oparka R, Porter RJ, Bray S, Skinner G, King S, Turbitt J, Collinson D, Miedzybrodzka ZH, Jellema G, Logan G, Kennedy RD, Turkington RC, McLean MH, Swinson D, Grabsch HI, Lord S, Seymour MJ, Hall PS, Petty RD. An investigation of the clinical impact and therapeutic relevance of a DNA damage immune response (DDIR) signature in patients with advanced gastroesophageal adenocarcinoma. ESMO Open 2024; 9:103450. [PMID: 38744099 PMCID: PMC11108838 DOI: 10.1016/j.esmoop.2024.103450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND An improved understanding of which gastroesophageal adenocarcinoma (GOA) patients respond to both chemotherapy and immune checkpoint inhibitors (ICI) is needed. We investigated the predictive role and underlying biology of a 44-gene DNA damage immune response (DDIR) signature in patients with advanced GOA. MATERIALS AND METHODS Transcriptional profiling was carried out on pretreatment tissue from 252 GOA patients treated with platinum-based chemotherapy (three dose levels) within the randomized phase III GO2 trial. Cross-validation was carried out in two independent GOA cohorts with transcriptional profiling, immune cell immunohistochemistry and epidermal growth factor receptor (EGFR) fluorescent in situ hybridization (FISH) (n = 430). RESULTS In the GO2 trial, DDIR-positive tumours had a greater radiological response (51.7% versus 28.5%, P = 0.022) and improved overall survival in a dose-dependent manner (P = 0.028). DDIR positivity was associated with a pretreatment inflamed tumour microenvironment (TME) and increased expression of biomarkers associated with ICI response such as CD274 (programmed death-ligand 1, PD-L1) and a microsatellite instability RNA signature. Consensus pathway analysis identified EGFR as a potential key determinant of the DDIR signature. EGFR amplification was associated with DDIR negativity and an immune cold TME. CONCLUSIONS Our results indicate the importance of the GOA TME in chemotherapy response, its relationship to DNA damage repair and EGFR as a targetable driver of an immune cold TME. Chemotherapy-sensitive inflamed GOAs could benefit from ICI delivered in combination with standard chemotherapy. Combining EGFR inhibitors and ICIs warrants further investigation in patients with EGFR-amplified tumours.
Collapse
Affiliation(s)
- M A Baxter
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee; Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee.
| | - L C Spender
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee
| | - D Cairns
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds
| | - S Walsh
- Department of Pathology, Ninewells Hospital and Medical School, NHS Tayside, Dundee
| | - R Oparka
- Department of Pathology, Ninewells Hospital and Medical School, NHS Tayside, Dundee
| | - R J Porter
- Department of Pathology, CRUK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh
| | - S Bray
- Tayside Biorepository, University of Dundee, Dundee
| | - G Skinner
- Tayside Biorepository, University of Dundee, Dundee
| | - S King
- Tayside Biorepository, University of Dundee, Dundee
| | - J Turbitt
- Genetics and Molecular Pathology Laboratory Services, NHS Grampian, Aberdeen
| | - D Collinson
- Genetics and Molecular Pathology Laboratory Services, NHS Grampian, Aberdeen
| | - Z H Miedzybrodzka
- Genetics and Molecular Pathology Laboratory Services, NHS Grampian, Aberdeen; School of Medicine, Medical Sciences, Nutrition and Dentistry, Polwarth Building, University of Aberdeen, Aberdeen
| | - G Jellema
- Almac Diagnostic Services, Craigavon
| | - G Logan
- Almac Diagnostic Services, Craigavon
| | - R D Kennedy
- Almac Diagnostic Services, Craigavon; Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - R C Turkington
- Almac Diagnostic Services, Craigavon; Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast
| | - M H McLean
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee; Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee
| | - D Swinson
- St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - H I Grabsch
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands; Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's University, University of Leeds, Leeds
| | - S Lord
- Department of Oncology, University of Oxford, Oxford
| | - M J Seymour
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds; St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - P S Hall
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, UK
| | - R D Petty
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee; Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee.
| |
Collapse
|
6
|
Yan J, Wang S, Zhang J, Yuan Q, Gao X, Zhang N, Pan Y, Zhang H, Liu K, Yu J, Lu L, Liu H, Gao X, Zhao S, Zhang W, Reyila A, Qi Y, Zhang Q, Cang S, Lu Y, Pan Y, Kong Y, Nie Y. DNA damage response-related immune activation signature predicts the response to immune checkpoint inhibitors: from gastrointestinal cancer analysis to pan-cancer validation. Cancer Biol Med 2023; 21:j.issn.2095-3941.2023.0303. [PMID: 38164720 PMCID: PMC10976329 DOI: 10.20892/j.issn.2095-3941.2023.0303] [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: 08/15/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVE DNA damage response (DDR) deficiency has emerged as a prominent determinant of tumor immunogenicity. This study aimed to construct a DDR-related immune activation (DRIA) signature and evaluate the predictive accuracy of the DRIA signature for response to immune checkpoint inhibitor (ICI) therapy in gastrointestinal (GI) cancer. METHODS A DRIA signature was established based on two previously reported DNA damage immune response assays. Clinical and gene expression data from two published GI cancer cohorts were used to assess and validate the association between the DRIA score and response to ICI therapy. The predictive accuracy of the DRIA score was validated based on one ICI-treated melanoma and three pan-cancer published cohorts. RESULTS The DRIA signature includes three genes (CXCL10, IDO1, and IFI44L). In the discovery cancer cohort, DRIA-high patients with gastric cancer achieved a higher response rate to ICI therapy than DRIA-low patients (81.8% vs. 8.8%; P < 0.001), and the predictive accuracy of the DRIA score [area under the receiver operating characteristic curve (AUC) = 0.845] was superior to the predictive accuracy of PD-L1 expression, tumor mutational burden, microsatellite instability, and Epstein-Barr virus status. The validation cohort demonstrated that the DRIA score identified responders with microsatellite-stable colorectal and pancreatic adenocarcinoma who received dual PD-1 and CTLA-4 blockade with radiation therapy. Furthermore, the predictive performance of the DRIA score was shown to be robust through an extended validation in melanoma, urothelial cancer, and pan-cancer. CONCLUSIONS The DRIA signature has superior and robust predictive accuracy for the efficacy of ICI therapy in GI cancer and pan-cancer, indicating that the DRIA signature may serve as a powerful biomarker for guiding ICI therapy decisions.
Collapse
Affiliation(s)
- Junya Yan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
- Department of Oncology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, China
| | - Shibo Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Jing Zhang
- Faculty of Life Science, Northwest University, Xi’an 710069, China
| | - Qiangqiang Yuan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Xianchun Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Nannan Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Yan Pan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Haohao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Kun Liu
- Unit 73211 of the People’s Liberation Army, Nanjing 211800, China
| | - Jun Yu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Linbin Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Hui Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), School of Medicine, Northwest University, Xi’an 710069, China
| | - Xiaoliang Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Sheng Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Wenyao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Abudurousuli Reyila
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Yu Qi
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Qiujin Zhang
- Shaanxi University of Chinese Medicine, Second Clinical Medicine Faculty, Xi’an 712046, China
| | - Shundong Cang
- Department of Oncology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou 450003, China
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Yanglin Pan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Air Force Military Medical University, Xi’an 710032, China
| |
Collapse
|
7
|
Tsukamoto Y, Kurogi S, Fujishima H, Shibata T, Fumoto S, Nishiki K, Suzuki K, Etoh T, Shiraishi N, Fuchino T, Hirashita Y, Nakada C, Uchida T, Inomata M, Moriyama M, Hijiya N. Association of immune-related expression profile with sensitivity to chemotherapy in esophageal squamous cell carcinoma. Cancer Sci 2023; 114:4459-4474. [PMID: 37715346 PMCID: PMC10637075 DOI: 10.1111/cas.15942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/22/2023] [Accepted: 08/04/2023] [Indexed: 09/17/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) followed by surgery is one of the standard therapeutic approaches in Japan for patients with locally advanced esophageal carcinoma. Recently, the JCOG1109 study revealed that NAC with docetaxel, cisplatin and 5-fluorouracil (5-FU) (DCF-NAC) is superior to NAC with cisplatin and 5-FU, and has now become the standard preoperative chemotherapy. Using a microarray system, we have previously investigated the expression profiles of endoscopic biopsy samples from patients with esophageal squamous cell carcinoma (ESCC) before DCF-NAC (preNAC) and identified 17 molecules as biomarkers predictive of a pathologically complete response to DCF-NAC. Here, we re-grouped our previous dataset based on the histopathological response grade with the addition of several microarray profiles and conducted a re-analysis using bioinformatic web tools including DAVID, GSEA, UALCAN, and CIBERSORTx. We identified 204 genes that were differentially expressed between the highly resistant and sensitive groups. Some of these differentially expressed genes (DEGs) were related to the immune response and showed higher expression in the sensitive group. UALCAN showed that high expression of 28 of the top 50 DEGs was associated with a favorable prognosis (p < 0.25), and that this reached a significant (p < 0.05) level for 18 of them, suggesting that patients with high expression of these genes might have benefited from chemotherapy and thus had a better outcome. In preNAC biopsy tissues from a DCF-sensitive case, we demonstrated the presence of cells expressing mRNA for CXCL9, one of the prognosis-related DEGs. Our results highlight the association of immune-related expression profile in preNAC ESCC with the DCF-NAC efficacy.
Collapse
Affiliation(s)
- Yoshiyuki Tsukamoto
- Department of Molecular Pathology, Faculty of MedicineOita UniversityOitaJapan
| | - Shusaku Kurogi
- Department of Molecular Pathology, Faculty of MedicineOita UniversityOitaJapan
| | - Hajime Fujishima
- Department of Gastroenterological and Pediatric Surgery, Faculty of MedicineOita UniversityOitaJapan
| | - Tomotaka Shibata
- Department of Gastroenterological and Pediatric Surgery, Faculty of MedicineOita UniversityOitaJapan
| | | | - Kohei Nishiki
- Department of SurgeryOita Nakamura HospitalOitaJapan
| | - Kosuke Suzuki
- Department of Gastroenterological and Pediatric Surgery, Faculty of MedicineOita UniversityOitaJapan
| | - Tsuyoshi Etoh
- Department of Gastroenterological and Pediatric Surgery, Faculty of MedicineOita UniversityOitaJapan
| | - Norio Shiraishi
- Comprehensive Surgery for Community Medicine, Oita University Faculty of MedicineOita UniversityOitaJapan
| | - Takafumi Fuchino
- Department of Molecular Pathology, Faculty of MedicineOita UniversityOitaJapan
- Department of Gastroenterology, Faculty of MedicineOita UniversityOitaJapan
| | - Yuka Hirashita
- Department of Molecular Pathology, Faculty of MedicineOita UniversityOitaJapan
- Department of Gastroenterology, Faculty of MedicineOita UniversityOitaJapan
| | - Chisato Nakada
- Department of Molecular Pathology, Faculty of MedicineOita UniversityOitaJapan
- Department of Urology, Faculty of MedicineOita UniversityOitaJapan
| | - Tomohisa Uchida
- Department of Molecular Pathology, Faculty of MedicineOita UniversityOitaJapan
| | - Masafumi Inomata
- Department of Gastroenterological and Pediatric Surgery, Faculty of MedicineOita UniversityOitaJapan
| | - Masatsugu Moriyama
- Department of Molecular Pathology, Faculty of MedicineOita UniversityOitaJapan
| | - Naoki Hijiya
- Department of Molecular Pathology, Faculty of MedicineOita UniversityOitaJapan
| |
Collapse
|
8
|
Arbore G, Albarello L, Bucci G, Punta M, Cossu A, Fanti L, Maurizio A, Di Mauro F, Bilello V, Arrigoni G, Bonfiglio S, Biancolini D, Puccetti F, Elmore U, Vago L, Cascinu S, Tonon G, Rosati R, Casorati G, Dellabona P. Preexisting Immunity Drives the Response to Neoadjuvant Chemotherapy in Esophageal Adenocarcinoma. Cancer Res 2023; 83:2873-2888. [PMID: 37350667 PMCID: PMC10472105 DOI: 10.1158/0008-5472.can-23-0356] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/18/2023] [Accepted: 06/20/2023] [Indexed: 06/24/2023]
Abstract
Current treatment for patients with locally advanced esophageal adenocarcinoma (EAC) is neoadjuvant chemotherapy (nCT), alone or combined with radiotherapy, before surgery. However, fewer than 30% of treated patients show a pathologic complete response to nCT, which correlates with increased 5-year survival compared with nonresponders. Understanding the mechanisms of response to nCT is pivotal to better stratify patients and inform more efficacious therapies. Here, we investigated the immune mechanisms involved in nCT response by multidimensional profiling of pretreatment tumor biopsies and blood from 68 patients with EAC (34 prospectively and 34 retrospectively collected), comparing complete responders versus nonresponders to nCT. At the tumor level, complete response to nCT was associated with molecular signatures of immune response and proliferation, increased putative antitumor tissue-resident memory CD39+ CD103+ CD8+ T cells, and reduced immunosuppressive T regulatory cells (Treg) and M2-like macrophages. Systemically, complete responders showed higher frequencies of immunostimulatory CD14+ CD11c+ HLA-DRhigh cells, and reduced programmed cell death ligand 1-positive (PD-L1+) monocytic myeloid-derived suppressor cells, along with high plasma GM-CSF (proinflammatory) and low IL4, CXCL10, C3a, and C5a (suppressive). Plasma proinflammatory and suppressive cytokines correlated directly and inversely, respectively, with the frequency of tumor-infiltrating CD39+ CD103+ CD8+ T cells. These results suggest that preexisting immunity in baseline tumor drives the clinical activity of nCT in locally advanced EAC. Furthermore, it may be possible to stratify patients based on predictive immune signatures, enabling tailored neoadjuvant and/or adjuvant regimens. SIGNIFICANCE Multidimensional profiling of pretreatment esophageal adenocarcinoma shows patient response to nCT is correlated with active preexisting immunity and indicates molecular pathways of resistance that may be targeted to improve clinical outcomes.
Collapse
Affiliation(s)
- Giuseppina Arbore
- Experimental Immunology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Albarello
- Department of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gabriele Bucci
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Punta
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Cossu
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorella Fanti
- Division of Gastroenterology & Gastrointestinal Endoscopy, San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Maurizio
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Di Mauro
- Experimental Immunology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vito Bilello
- Experimental Immunology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianluigi Arrigoni
- Department of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Bonfiglio
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Donatella Biancolini
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Puccetti
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ugo Elmore
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Vago
- Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Cascinu
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Tonon
- Vita-Salute San Raffaele University, Milan, Italy
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Riccardo Rosati
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Dellabona
- Experimental Immunology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
9
|
McCabe NH, Stevenson L, Scanlon E, Douglas R, Kennedy S, Keminer O, Windshügel B, Zisterer D, Kennedy RD, Blayney JK, Turkington RC. Identification of Src as a Therapeutic Target in Oesophageal Adenocarcinoma through Functional Genomic and High-Throughput Drug Screening Approaches. Cancers (Basel) 2022; 14:cancers14153726. [PMID: 35954391 PMCID: PMC9367554 DOI: 10.3390/cancers14153726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/15/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Drug resistance limits the effectiveness of oesophageal adenocarcinoma (OAC) chemotherapies, leading to a poor prognosis for this disease. Elucidation of the underlying resistance mechanisms is key to enabling the identification of more effective treatments. This study, therefore, aims to identify novel therapeutic and/or chemotherapy sensitising drug targets in OAC. Transcriptional data from a cohort of 273 pre-treatment OAC biopsies, from patients who received neoadjuvant chemotherapy followed by surgical resection, were analysed using gene set enrichment analysis (GSEA) to determine differential gene expression between responding and non-responding OAC tumours. From this, 80 genes were selected for high-throughput siRNA screening in OAC cell lines with or without standard chemotherapy treatment. In parallel, cell viability assays were performed using a panel of FDA-approved drugs and combination index (CI) values were calculated to evaluate drug synergy with standard chemotherapy. Mechanisms of synergy were investigated using western blot, propidium iodide flow cytometry, and proliferation assays. Taken together, the screens identified that targeting Src, using either siRNA or the small molecule inhibitor dasatinib, enhanced the efficacy of chemotherapy in OAC cells. Further in vitro functional analysis confirmed Src inhibition to be synergistic with standard OAC chemotherapies, 5-fluorouracil (5-FU), and cisplatin (CDDP). In conclusion, a compound screen together with a functional genomic approach identified Src as a potential chemosensitising target in OAC, which could be assessed in a clinical study for poor prognosis OAC patients.
Collapse
Affiliation(s)
- Niamh H. McCabe
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (N.H.M.); (L.S.); (E.S.); (R.D.); (S.K.); (J.K.B.)
| | - Leanne Stevenson
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (N.H.M.); (L.S.); (E.S.); (R.D.); (S.K.); (J.K.B.)
| | - Enya Scanlon
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (N.H.M.); (L.S.); (E.S.); (R.D.); (S.K.); (J.K.B.)
| | - Rosalie Douglas
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (N.H.M.); (L.S.); (E.S.); (R.D.); (S.K.); (J.K.B.)
| | - Susanna Kennedy
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (N.H.M.); (L.S.); (E.S.); (R.D.); (S.K.); (J.K.B.)
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525 Hamburg, Germany; (O.K.); (B.W.)
- Department of Life Sciences and Chemistry, Jacobs University Bremen, 28759 Bremen, Germany
| | - Björn Windshügel
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525 Hamburg, Germany; (O.K.); (B.W.)
- Department of Life Sciences and Chemistry, Jacobs University Bremen, 28759 Bremen, Germany
| | - Daniela Zisterer
- Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, D08 XW7X Dublin, Ireland;
| | | | - Jaine K. Blayney
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (N.H.M.); (L.S.); (E.S.); (R.D.); (S.K.); (J.K.B.)
| | - Richard C. Turkington
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (N.H.M.); (L.S.); (E.S.); (R.D.); (S.K.); (J.K.B.)
- Correspondence: ; Tel.: +44-(0)28-9097-2756
| |
Collapse
|
10
|
Baxter MA, Middleton F, Cagney HP, Petty RD. Resistance to immune checkpoint inhibitors in advanced gastro-oesophageal cancers. Br J Cancer 2021; 125:1068-1079. [PMID: 34230609 PMCID: PMC8505606 DOI: 10.1038/s41416-021-01425-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/17/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have altered the treatment paradigm across a range of tumour types, including gastro-oesophageal cancers. For patients with any cancer type who respond, ICIs can confer long-term disease control and significantly improve survival and quality of life, but for patients with gastro-oesophageal cancer, ICIs can be transformative, as durable responses in advanced disease have hitherto been rare, especially in those patients who are resistant to first-line cytotoxic therapies. Results from trials in patients with advanced-stage gastro-oesophageal cancer have raised hopes that ICIs will be successful as adjuvant and neoadjuvant treatments in early-stage disease, when the majority of patients relapse after potential curative treatments, and several trials are ongoing. Unfortunately, however, ICI-responding patients appear to constitute a minority subgroup within gastro-oesophageal cancer, and resistance to ICI therapy (whether primary or acquired) is common. Understanding the biological mechanisms of ICI resistance is a current major research challenge and involves investigation of both tumour and patient-specific factors. In this review, we discuss the mechanisms underlying ICI resistance and their potential specific applications of this knowledge towards precision medicine strategies in the management of gastro-oesophageal cancers in clinical practice.
Collapse
Affiliation(s)
- Mark A Baxter
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK.
| | - Fearghas Middleton
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK
| | - Hannah P Cagney
- School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Russell D Petty
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK.
| |
Collapse
|
11
|
Izadi F, Sharpe BP, Breininger SP, Secrier M, Gibson J, Walker RC, Rahman S, Devonshire G, Lloyd MA, Walters ZS, Fitzgerald RC, Rose-Zerilli MJJ, Underwood TJ. Genomic Analysis of Response to Neoadjuvant Chemotherapy in Esophageal Adenocarcinoma. Cancers (Basel) 2021; 13:3394. [PMID: 34298611 PMCID: PMC8308111 DOI: 10.3390/cancers13143394] [Citation(s) in RCA: 4] [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: 06/10/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 01/04/2023] Open
Abstract
Neoadjuvant therapy followed by surgery is the standard of care for locally advanced esophageal adenocarcinoma (EAC). Unfortunately, response to neoadjuvant chemotherapy (NAC) is poor (20-37%), as is the overall survival benefit at five years (9%). The EAC genome is complex and heterogeneous between patients, and it is not yet understood whether specific mutational patterns may result in chemotherapy sensitivity or resistance. To identify associations between genomic events and response to NAC in EAC, a comparative genomic analysis was performed in 65 patients with extensive clinical and pathological annotation using whole-genome sequencing (WGS). We defined response using Mandard Tumor Regression Grade (TRG), with responders classified as TRG1-2 (n = 27) and non-responders classified as TRG4-5 (n =38). We report a higher non-synonymous mutation burden in responders (median 2.08/Mb vs. 1.70/Mb, p = 0.036) and elevated copy number variation in non-responders (282 vs. 136/patient, p < 0.001). We identified copy number variants unique to each group in our cohort, with cell cycle (CDKN2A, CCND1), c-Myc (MYC), RTK/PIK3 (KRAS, EGFR) and gastrointestinal differentiation (GATA6) pathway genes being specifically altered in non-responders. Of note, NAV3 mutations were exclusively present in the non-responder group with a frequency of 22%. Thus, lower mutation burden, higher chromosomal instability and specific copy number alterations are associated with resistance to NAC.
Collapse
Affiliation(s)
- Fereshteh Izadi
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
- Centre for NanoHealth, Swansea University Medical School, Singleton Campus, Swansea SA2 8PP, UK
| | - Benjamin P. Sharpe
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Stella P. Breininger
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
| | - Maria Secrier
- UCL Genetics Institute, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK;
| | - Jane Gibson
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Robert C. Walker
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
| | - Saqib Rahman
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
| | - Ginny Devonshire
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK;
| | - Megan A. Lloyd
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
| | - Zoë S. Walters
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Rebecca C. Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge CB2 OXZ, UK;
| | - Matthew J. J. Rose-Zerilli
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Tim J. Underwood
- School of Cancer Sciences, Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (F.I.); (B.P.S.); (S.P.B.); (J.G.); (R.C.W.); (S.R.); (M.A.L.); (Z.S.W.); (M.J.J.R.-Z.)
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | | |
Collapse
|
12
|
Foley KG, Lavery A, Napier E, Campbell D, Eatock MM, Kennedy RD, Bradley KM, Turkington RC. A DNA-damage immune response assay combined with PET biomarkers predicts response to neo-adjuvant chemotherapy and survival in oesophageal adenocarcinoma. Sci Rep 2021; 11:13061. [PMID: 34158588 PMCID: PMC8219719 DOI: 10.1038/s41598-021-92545-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/07/2021] [Indexed: 11/24/2022] Open
Abstract
18F-fluorodeoxyglucose PET-CT may guide treatment decisions in patients with oesophageal adenocarcinoma (OAC). This study evaluated the added value of maximum standardised uptake value (SUVmax) to a novel DNA-damage immune response (DDIR) assay to improve pathological response prediction. The diagnostic accuracy of PET response and the prognostic significance of PET metrics for recurrence-free survival (RFS) and overall survival (OS) were assessed. This was a retrospective, single-centre study of OAC patients treated with neo-adjuvant chemotherapy from 2003 to 2014. SUVmax was recorded from baseline and repeat PET-CT after completion of pre-operative chemotherapy. Logistic regression models tested the additional predictive value of PET metrics combined with the DDIR assay for pathological response. Cox regression models tested the prognostic significance of PET metrics for RFS and OS. In total, 113 patients were included; 25 (22.1%) were DDIR positive and 88 (77.9%) were DDIR negative. 69 (61.1%) were PET responders (SUVmax reduction of 35%) and 44 (38.9%) were PET non-responders. After adding PET metrics to DDIR status, post-chemotherapy SUVmax (hazard ratio (HR) 0.75, p = 0.02), SUVmax change (HR 1.04, p = 0.003) and an optimum SUVmax reduction of 46.5% (HR 4.36, p = 0.021) showed additional value for predicting pathological response. The optimised SUVmax threshold was independently significant for RFS (HR 0.47, 95% CI 0.26–0.85, p = 0.012) and OS (HR 0.51, 95% CI 0.26–0.99, p = 0.047). This study demonstrated the additional value of PET metrics, when combined with a novel DDIR assay, to predict pathological response in OAC patients treated with neo-adjuvant chemotherapy. Furthermore, an optimised SUVmax reduction threshold for pathological response was calculated and was independently significant for RFS and OS.
Collapse
Affiliation(s)
| | | | - Eoin Napier
- Belfast Health and Social Care Trust, Belfast, UK
| | | | - Martin M Eatock
- Queen's University Belfast, Belfast, UK.,Belfast Health and Social Care Trust, Belfast, UK
| | | | - Kevin M Bradley
- Wales Research & Diagnostic Positron Emission Tomography Imaging Centre (PETIC), Cardiff University, Cardiff, UK
| | | |
Collapse
|
13
|
Koemans WJ, van Dieren JM, van den Berg JG, Meijer GA, Snaebjornsson P, Chalabi M, Lecot F, Riedl R, Krijgsman O, Hofland I, Broeks A, Voncken FEM, Peppelenbosch MP, Sosef MN, van Sandick JW, Kodach LL. High CD8 + tumour-infiltrating lymphocyte density associates with unfavourable prognosis in oesophageal adenocarcinoma following poor response to neoadjuvant chemoradiotherapy. Histopathology 2021; 79:238-251. [PMID: 33660299 DOI: 10.1111/his.14361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 02/04/2021] [Accepted: 03/02/2021] [Indexed: 11/29/2022]
Abstract
AIMS Determining prognosis following poor response to neoadjuvant chemoradiotherapy (nCRT) in oesophageal adenocarcinoma (OAC) remains challenging. An immunosuppressive tumour microenvironment (TME) as well as immune infiltrate density and composition are considered to play a critical role in the immune interaction between host and tumour and can predict therapy response and survival in many cancers, including gastrointestinal malignancies. The aim of this study was to establish the TME characteristics associated with survival following a poor response to nCRT. METHODS AND RESULTS The prognostic significance of OAC-associated CD3+ , CD4+ , CD8+ , forkhead box protein 3 (FoxP3+ ) and programmed cell death ligand 1 (PD-L1) expression was studied by immunohistochemistry and quantified by automated image analysis in 123 patients who underwent nCRT and curative resection. Results from good and poor responders were contrasted and immune infiltration was related to disease course in both groups. Subsequently a cohort of 57 patients with a moderate response to nCRT was analysed in a similar fashion. Tumour cell percentage positively correlated to immune infiltration markers. In good and moderate responders, none of the immune infiltrate parameters was associated with survival; in poor responders CD8+ was an independent negative predictor of OS in univariate analysis (P = 0.03) and high CD8+ infiltration was associated with worse OS (15 versus 32 months, P = 0.042). CONCLUSION A high CD8+ density is an independent biomarker of poor OS in poor responders to nCRT, but not in good and moderate responders. Our results suggest that patients with a poor response to nCRT but concomitant high CD8+ counts in the resection specimen require adjuvant therapy.
Collapse
Affiliation(s)
- Willem J Koemans
- Department of Surgical Oncology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jolanda M van Dieren
- Department of Gastrointestinal Oncology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jose G van den Berg
- Department of Pathology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Gerrit A Meijer
- Department of Pathology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Myriam Chalabi
- Department of Gastrointestinal Oncology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Frederig Lecot
- Department of Surgery, Zuyderland Hospital, Heerlen, the Netherlands
| | - Robert Riedl
- Department of Pathology, Zuyderland Hospital, Heerlen, the Netherlands
| | - Oscar Krijgsman
- Departments of Molecular Oncology and Immunology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ingrid Hofland
- Departments of Core Facility, Molecular Pathology and Biobanking, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Departments of Core Facility, Molecular Pathology and Biobanking, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Francine E M Voncken
- Department of Radiotherapy, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maikel P Peppelenbosch
- Departments of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Meindert N Sosef
- Department of Surgery, Zuyderland Hospital, Heerlen, the Netherlands
| | - Johanna W van Sandick
- Department of Surgical Oncology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Liudmila L Kodach
- Department of Pathology, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Lavery A, Blayney JK, Turkington RC. Response to: 'Unified definition of relapse-free survival should be used for evaluating survival benefit in oesophageal adenocarcinoma' by Li. Gut 2021; 70:626. [PMID: 32606210 DOI: 10.1136/gutjnl-2020-322096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/08/2022]
Affiliation(s)
- Anita Lavery
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Jaine K Blayney
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Richard C Turkington
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| |
Collapse
|
15
|
Doultsinos D, Mills IG. Derivation and Application of Molecular Signatures to Prostate Cancer: Opportunities and Challenges. Cancers (Basel) 2021; 13:495. [PMID: 33525365 PMCID: PMC7865812 DOI: 10.3390/cancers13030495] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer is a high-incidence cancer that requires improved patient stratification to ensure accurate predictions of risk and treatment response. Due to the significant contributions of transcription factors and epigenetic regulators to prostate cancer progression, there has been considerable progress made in developing gene signatures that may achieve this. Some of these are aligned to activities of key drivers such as the androgen receptor, whilst others are more agnostic. In this review, we present an overview of these signatures, the strategies for their derivation, and future perspectives on their continued development and evolution.
Collapse
Affiliation(s)
- Dimitrios Doultsinos
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
| | - Ian G. Mills
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Patrick G Johnston Centre for Cancer Research, Queen’s University of Belfast, Belfast BT9 7AE, UK
| |
Collapse
|
16
|
Malla SB, Fisher DJ, Domingo E, Blake A, Hassanieh S, Redmond KL, Richman SD, Youdell M, Walker SM, Logan GE, Chatzipli A, Amirkhah R, Humphries MP, Craig SG, McDermott U, Seymour MT, Morton DG, Quirke P, West NP, Salto-Tellez M, Kennedy RD, Johnston PG, Tomlinson I, Koelzer VH, Campo L, Kaplan RS, Longley DB, Lawler M, Maughan TS, Brown LC, Dunne PD. In-depth Clinical and Biological Exploration of DNA Damage Immune Response as a Biomarker for Oxaliplatin Use in Colorectal Cancer. Clin Cancer Res 2021; 27:288-300. [PMID: 33028592 PMCID: PMC7614625 DOI: 10.1158/1078-0432.ccr-20-3237] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE The DNA damage immune response (DDIR) assay was developed in breast cancer based on biology associated with deficiencies in homologous recombination and Fanconi anemia pathways. A positive DDIR call identifies patients likely to respond to platinum-based chemotherapies in breast and esophageal cancers. In colorectal cancer, there is currently no biomarker to predict response to oxaliplatin. We tested the ability of the DDIR assay to predict response to oxaliplatin-based chemotherapy in colorectal cancer and characterized the biology in DDIR-positive colorectal cancer. EXPERIMENTAL DESIGN Samples and clinical data were assessed according to DDIR status from patients who received either 5-fluorouracil (5-FU) or 5FUFA (bolus and infusion 5-FU with folinic acid) plus oxaliplatin (FOLFOX) within the FOCUS trial (n = 361, stage IV), or neoadjuvant FOLFOX in the FOxTROT trial (n = 97, stage II/III). Whole transcriptome, mutation, and IHC data of these samples were used to interrogate the biology of DDIR in colorectal cancer. RESULTS Contrary to our hypothesis, DDIR-negative patients displayed a trend toward improved outcome for oxaliplatin-based chemotherapy compared with DDIR-positive patients. DDIR positivity was associated with microsatellite instability (MSI) and colorectal molecular subtype 1. Refinement of the DDIR signature, based on overlapping IFN-related chemokine signaling associated with DDIR positivity across colorectal cancer and breast cancer cohorts, further confirmed that the DDIR assay did not have predictive value for oxaliplatin-based chemotherapy in colorectal cancer. CONCLUSIONS DDIR positivity does not predict improved response following oxaliplatin treatment in colorectal cancer. However, data presented here suggest the potential of the DDIR assay in identifying immune-rich tumors that may benefit from immune checkpoint blockade, beyond current use of MSI status.
Collapse
Affiliation(s)
- Sudhir B Malla
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - David J Fisher
- MRC Clinical Trials Unit, University College London, London, United Kingdom
| | - Enric Domingo
- MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Andrew Blake
- MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Sylvana Hassanieh
- MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Keara L Redmond
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Susan D Richman
- Pathology and data analytics, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Michael Youdell
- MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Gemma E Logan
- Almac Diagnostic Services, Craigavon, United Kingdom
| | - Aikaterina Chatzipli
- Cancer, Ageing and Somatic Mutation (CASM), Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Raheleh Amirkhah
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Matthew P Humphries
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Stephanie G Craig
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Ultan McDermott
- Cancer, Ageing and Somatic Mutation (CASM), Wellcome Sanger Institute, Cambridge, United Kingdom
- AstraZeneca, United Kingdom
| | | | - Dion G Morton
- University of Birmingham, Birmingham, United Kingdom
| | - Philip Quirke
- Cancer, Ageing and Somatic Mutation (CASM), Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Nicholas P West
- Cancer, Ageing and Somatic Mutation (CASM), Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Manuel Salto-Tellez
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Richard D Kennedy
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Patrick G Johnston
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | | | | | - Letitia Campo
- MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Richard S Kaplan
- MRC Clinical Trials Unit, University College London, London, United Kingdom
| | - Daniel B Longley
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Mark Lawler
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Timothy S Maughan
- MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom.
| | - Louise C Brown
- MRC Clinical Trials Unit, University College London, London, United Kingdom
| | | |
Collapse
|
17
|
Li W. Unified definition of relapse-free survival should be used for evaluating survival benefit in oesophageal adenocarcinoma. Gut 2021; 70:228. [PMID: 32404310 DOI: 10.1136/gutjnl-2020-321482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Weizheng Li
- Department of Gastroenterology, Maoming People's Hospital, Maoming 525000, China
| |
Collapse
|
18
|
Zhang C, Shi Z, Kalendralis P, Whybra P, Parkinson C, Berbee M, Spezi E, Roberts A, Christian A, Lewis W, Crosby T, Dekker A, Wee L, Foley KG. Prediction of lymph node metastases using pre-treatment PET radiomics of the primary tumour in esophageal adenocarcinoma: an external validation study. Br J Radiol 2020; 94:20201042. [PMID: 33264032 DOI: 10.1259/bjr.20201042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES To improve clinical lymph node staging (cN-stage) in oesophageal adenocarcinoma by developing and externally validating three prediction models; one with clinical variables only, one with positron emission tomography (PET) radiomics only, and a combined clinical and radiomics model. METHODS Consecutive patients with fluorodeoxyglucose (FDG) avid tumours treated with neoadjuvant therapy between 2010 and 2016 in two international centres (n = 130 and n = 60, respectively) were included. Four clinical variables (age, gender, clinical T-stage and tumour regression grade) and PET radiomics from the primary tumour were used for model development. Diagnostic accuracy, area under curve (AUC), discrimination and calibration were calculated for each model. The prognostic significance was also assessed. RESULTS The incidence of lymph node metastases was 58% in both cohorts. The areas under the curve of the clinical, radiomics and combined models were 0.79, 0.69 and 0.82 in the developmental cohort, and 0.65, 0.63 and 0.69 in the external validation cohort, with good calibration demonstrated. The area under the curve of current cN-stage in development and validation cohorts was 0.60 and 0.66, respectively. For overall survival, the combined clinical and radiomics model achieved the best discrimination performance in the external validation cohort (X2 = 6.08, df = 1, p = 0.01). CONCLUSION Accurate diagnosis of lymph node metastases is crucial for prognosis and guiding treatment decisions. Despite finding improved predictive performance in the development cohort, the models using PET radiomics derived from the primary tumour were not fully replicated in an external validation cohort. ADVANCES IN KNOWLEDGE This international study attempted to externally validate a new prediction model for lymph node metastases using PET radiomics. A model combining clinical variables and PET radiomics improved discrimination of lymph node metastases, but these results were not externally replicated.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Zhenwei Shi
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Petros Kalendralis
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Phil Whybra
- School of Engineering, Cardiff University, Cardiff, UK
| | | | - Maaike Berbee
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | - Ashley Roberts
- Department of Radiology, University Hospital of Wales, Cardiff, UK
| | - Adam Christian
- Department of Pathology, University Hospital of Wales, Cardiff, UK
| | - Wyn Lewis
- Department of Upper GI Surgery, University Hospital of Wales, Cardiff, UK
| | - Tom Crosby
- Department of Clinical Oncology, Velindre Cancer Centre, Cardiff, UK
| | - Andre Dekker
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Kieran G Foley
- Department of Radiology, Velindre Cancer Centre, Cardiff, UK
| |
Collapse
|
19
|
Lavery A, Turkington RC. Transcriptomic biomarkers for predicting response to neoadjuvant treatment in oesophageal cancer. Gastroenterol Rep (Oxf) 2020; 8:411-424. [PMID: 33442473 PMCID: PMC7793050 DOI: 10.1093/gastro/goaa065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/21/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
Oesophageal cancer is a devastating disease with poor outcomes and is the sixth leading cause of cancer death worldwide. In the setting of resectable disease, there is clear evidence that neoadjuvant chemotherapy and chemoradiotherapy result in improved survival. Disappointingly, only 15%-30% of patients obtain a histopathological response to neoadjuvant therapy, often at the expense of significant toxicity. There are no predictive biomarkers in routine clinical use in this setting and the ability to stratify patients for treatment could dramatically improve outcomes. In this review, we aim to outline current progress in evaluating predictive transcriptomic biomarkers for neoadjuvant therapy in oesophageal cancer and discuss the challenges facing biomarker development in this setting. We place these issues in the wider context of recommendations for biomarker development and reporting. The majority of studies focus on messenger RNA (mRNA) and microRNA (miRNA) biomarkers. These studies report a range of different genes involved in a wide variety of pathways and biological processes, and this is explained to a large extent by the different platforms and analysis methods used. Many studies are also vastly underpowered so are not suitable for identifying a candidate biomarker. Multiple molecular subtypes of oesophageal cancer have been proposed, although little is known about how these relate to clinical outcomes. We anticipate that the accumulating wealth of genomic and transcriptomic data and clinical trial collaborations in the coming years will provide unique opportunities to stratify patients in this poor-prognosis disease and recommend that future biomarker development incorporates well-designed retrospective and prospective analyses.
Collapse
Affiliation(s)
- Anita Lavery
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK
| | - Richard C Turkington
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
20
|
Donlon NE, Sheppard A, Davern M, O’Connell F, Phelan JJ, Power R, Nugent T, Dinneen K, Aird J, Greene J, Nevins Selvadurai P, Bhardwaj A, Foley EK, Ravi N, Donohoe CL, Reynolds JV, Lysaght J, O’Sullivan J, Dunne MR. Linking Circulating Serum Proteins with Clinical Outcomes in Esophageal Adenocarcinoma-An Emerging Role for Chemokines. Cancers (Basel) 2020; 12:3356. [PMID: 33202734 PMCID: PMC7698106 DOI: 10.3390/cancers12113356] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) is an aggressive cancer with poor prognosis and incidence is increasing rapidly in the Western world. Multi-modal treatment has improved survival outcomes but only for a minority of patients. Currently no markers have been identified to predict treatment response. This study investigated the association between clinical outcomes and pre-treatment levels of 54 serum proteins in n = 80 patients with EAC. Low tumor regression grade (TRG), corresponding to a favorable treatment response, was linked to prolonged overall survival (OS). CCL4 was higher in patients with a favorable treatment response, while Tie2 and CRP were higher in poor responders. Elevated CCL22 and CCL26 was associated with improved OS, while elevated IL-10 showed a negative association. CCL3, CCL4, IL-1α and IL-12/IL23p40 were highest in individuals with no adverse features of tumor biology, whereas levels of Tie2 and VEGF were lowest in this cohort. CCL4 was also elevated in patients with high tumor lymphocyte infiltration. Comparison of matched pre- and post-treatment serum (n = 28) showed a large reduction in VEGFC, and a concomitant increase in other cytokines, including CCL4. These data link several serum markers with clinical outcomes, highlighting an important role for immune cell trafficking in the EAC antitumor immune response.
Collapse
Affiliation(s)
- Noel E. Donlon
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Andrew Sheppard
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Maria Davern
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Fiona O’Connell
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - James J. Phelan
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Robert Power
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Timothy Nugent
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Kate Dinneen
- Department of Histopathology, St James’s Hospital, Dublin 8, Ireland; (K.D.); (J.A.)
| | - John Aird
- Department of Histopathology, St James’s Hospital, Dublin 8, Ireland; (K.D.); (J.A.)
| | - John Greene
- Department of Medical Oncology, St James’s Hospital, Dublin 8, Ireland; (J.G.); (P.N.S.)
| | - Paul Nevins Selvadurai
- Department of Medical Oncology, St James’s Hospital, Dublin 8, Ireland; (J.G.); (P.N.S.)
| | - Anshul Bhardwaj
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Emma K. Foley
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Narayanasamy Ravi
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Claire L. Donohoe
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - John V. Reynolds
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Jacintha O’Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| | - Margaret R. Dunne
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (N.E.D.); (A.S.); (M.D.); (F.O.); (J.J.P.); (R.P.); (T.N.); (A.B.); (E.K.F.); (N.R.); (C.L.D.); (J.V.R.); (J.L.); (J.O.)
- Trinity St James’s Cancer Institute, St James’s Hospital, Dublin 8, Ireland
| |
Collapse
|
21
|
Wang S, Liu Y, Feng Y, Zhang J, Swinnen J, Li Y, Ni Y. A Review on Curability of Cancers: More Efforts for Novel Therapeutic Options Are Needed. Cancers (Basel) 2019; 11:E1782. [PMID: 31766180 PMCID: PMC6896199 DOI: 10.3390/cancers11111782] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer remains a major cause of death globally. Given its relapsing and fatal features, curing cancer seems to be something hardly possible for the majority of patients. In view of the development in cancer therapies, this article summarizes currently available cancer therapeutics and cure potential by cancer type and stage at diagnosis, based on literature and database reviews. Currently common cancer therapeutics include surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. However, treatment with curative intent by these methods are mainly eligible for patients with localized disease or treatment-sensitive cancers and therefore their contributions to cancer curability are relatively limited. The prognosis for cancer patients varies among different cancer types with a five-year relative survival rate (RSR) of more than 80% in thyroid cancer, melanoma, breast cancer, and Hodgkin's lymphoma. The most dismal prognosis is observed in patients with small-cell lung cancer, pancreatic cancer, hepatocellular carcinoma, oesophagal cancer, acute myeloid leukemia, non-small cell lung cancer, and gastric cancer with a five-year RSR ranging between 7% and 28%. The current review is intended to provide a general view about how much we have achieved in curing cancer as regards to different therapies and cancer types. Finally, we propose a small molecule dual-targeting broad-spectrum anticancer strategy called OncoCiDia, in combination with emerging highly sensitive liquid biopsy, with theoretical curative potential for the management of solid malignancies, especially at the micro-cancer stage.
Collapse
Affiliation(s)
- Shuncong Wang
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| | - Yewei Liu
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| | - Yuanbo Feng
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| | - Jian Zhang
- Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China;
| | - Johan Swinnen
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| | - Yue Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yicheng Ni
- KU Leuven, Campus Gasthuisberg, Faculty of Medicine, 3000 Leuven, Belgium; (S.W.); (Y.L.); (Y.F.); (J.S.)
| |
Collapse
|
22
|
Miyake M, Hori S, Ohnishi S, Toritsuka M, Fujii T, Shimizu T, Owari T, Morizawa Y, Gotoh D, Itami Y, Nakai Y, Anai S, Torimoto K, Tanaka N, Fujimoto K. Supplementary granulocyte macrophage colony-stimulating factor to chemotherapy and programmed death-ligand 1 blockade decreases local recurrence after surgery in bladder cancer. Cancer Sci 2019; 110:3315-3327. [PMID: 31385407 PMCID: PMC6778624 DOI: 10.1111/cas.14158] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/16/2022] Open
Abstract
Despite advances and refinements in surgery and perioperative chemotherapy, there are still unmet medical needs with respect to radical cystectomy for muscle‐invasive bladder cancer (MIBC). We investigated the potential benefit of supplementary granulocyte macrophage colony‐stimulating factor (GM‐CSF) to chemoimmunotherapy with programmed cell death protein‐1 (PD‐1)/programmed death‐ligand 1 (PD‐L1) axis blockade and standard neoadjuvant chemotherapy in bladder cancer. We inoculated 2 × 105MBT2 cells s.c. in C3H mice to create a syngeneic animal model of local recurrence (LR). When the tumor diameter reached 12 mm, the mice were allocated randomly as follows: (i) non‐treated control (vehicle only); (ii) anti‐mPD‐L1 monotherapy; (iii) mGM‐CSF monotherapy; (iv) anti‐mPD‐L1 plus mGM‐CSF; (v) gemcitabine and cisplatin (GC); (vi) GC plus anti‐mPD‐L1; (vii) GC plus mGM‐CSF; and (viii) GC plus anti‐mPD‐L1 plus mGM‐CSF. After completing 2‐week neoadjuvant therapy, tumors were resected for resection margin evaluation and immunohistochemical staining and blood was collected for flow cytometry and ELISA. Operative wounds were sutured, and the operative site was monitored to detect LR. Addition of anti‐mPD‐L1 and mGM‐CSF to neoadjuvant GC chemotherapy enhanced the antitumor effect and reduced positive resection margins (50% vs 12.5%). Combination of GC, anti‐mPD‐L1, and mGM‐CSF resulted in longer LR‐free survival and cancer‐specific survival compared to those in other groups. These effects involved an immunotherapy‐related decrease in oncological properties such as tumor invasion capacity and epithelial‐mesenchymal transition. mGM‐CSF significantly decreased the accumulation of myeloid‐derived suppressor cells in both the blood and tumor microenvironment and blood interleukin‐6 levels. Supplementary GM‐CSF to neoadjuvant GC plus PD‐L1 blockade could decrease LR after radical surgery by immune modulation in the blood and tumor microenvironment.
Collapse
Affiliation(s)
- Makito Miyake
- Department of Urology, Nara Medical University, Nara, Japan
| | - Shunta Hori
- Department of Urology, Nara Medical University, Nara, Japan
| | - Sayuri Ohnishi
- Department of Urology, Nara Medical University, Nara, Japan
| | | | - Tomomi Fujii
- Department of Diagnostic Pathology, Nara Medical University, Nara, Japan
| | - Takuto Shimizu
- Department of Urology, Nara Medical University, Nara, Japan
| | - Takuya Owari
- Department of Urology, Nara Medical University, Nara, Japan
| | | | - Daisuke Gotoh
- Department of Urology, Nara Medical University, Nara, Japan
| | | | - Yasushi Nakai
- Department of Urology, Nara Medical University, Nara, Japan
| | - Satoshi Anai
- Department of Urology, Nara Medical University, Nara, Japan
| | | | | | | |
Collapse
|