197 results on '"Gary Middleton"'
Search Results
2. Tissue-Resident Memory T Cells in Pancreatic Ductal Adenocarcinoma Coexpress PD-1 and TIGIT and Functional Inhibition Is Reversible by Dual Antibody Blockade
- Author
-
Hayden Pearce, Wayne Croft, Samantha M. Nicol, Sandra Margielewska-Davies, Richard Powell, Richard Cornall, Simon J. Davis, Francesca Marcon, Matthew R. Pugh, Éanna Fennell, Sarah Powell-Brett, Brinder S. Mahon, Rachel M. Brown, Gary Middleton, Keith Roberts, and Paul Moss
- Subjects
Cancer Research ,Immunology - Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor clinical outlook. Responses to immune checkpoint blockade are suboptimal and a much more detailed understanding of the tumor immune microenvironment is needed if this situation is to be improved. Here, we characterized tumor-infiltrating T-cell populations in patients with PDAC using cytometry by time of flight (CyTOF) and single-cell RNA sequencing. T cells were the predominant immune cell subset observed within tumors. Over 30% of CD4+ T cells expressed a CCR6+CD161+ Th17 phenotype and 17% displayed an activated regulatory T-cell profile. Large populations of CD8+ tissue-resident memory (TRM) T cells were also present and expressed high levels of programmed cell death protein 1 (PD-1) and TIGIT. A population of putative tumor-reactive CD103+CD39+ T cells was also observed within the CD8+ tumor-infiltrating lymphocytes population. The expression of PD-1 ligands was limited largely to hemopoietic cells whilst TIGIT ligands were expressed widely within the tumor microenvironment. Programmed death-ligand 1 and CD155 were expressed within the T-cell area of ectopic lymphoid structures and colocalized with PD-1+TIGIT+ CD8+ T cells. Combinatorial anti–PD-1 and TIGIT blockade enhanced IFNγ secretion and proliferation of T cells in the presence of PD-1 and TIGIT ligands. As such, we showed that the PDAC microenvironment is characterized by the presence of substantial populations of TRM cells with an exhausted PD-1+TIGIT+ phenotype where dual checkpoint receptor blockade represents a promising avenue for future immunotherapy.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Fig. S1 - S12 from Tissue-Resident Memory T Cells in Pancreatic Ductal Adenocarcinoma Coexpress PD-1 and TIGIT and Functional Inhibition Is Reversible by Dual Antibody Blockade
- Author
-
Paul Moss, Keith Roberts, Gary Middleton, Rachel M. Brown, Brinder S. Mahon, Sarah Powell-Brett, Éanna Fennell, Matthew R. Pugh, Francesca Marcon, Simon J. Davis, Richard Cornall, Richard Powell, Sandra Margielewska-Davies, Samantha M. Nicol, Wayne Croft, and Hayden Pearce
- Abstract
Supplementary Figures S1 - S12
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Tables S1 - S5 from Tissue-Resident Memory T Cells in Pancreatic Ductal Adenocarcinoma Coexpress PD-1 and TIGIT and Functional Inhibition Is Reversible by Dual Antibody Blockade
- Author
-
Paul Moss, Keith Roberts, Gary Middleton, Rachel M. Brown, Brinder S. Mahon, Sarah Powell-Brett, Éanna Fennell, Matthew R. Pugh, Francesca Marcon, Simon J. Davis, Richard Cornall, Richard Powell, Sandra Margielewska-Davies, Samantha M. Nicol, Wayne Croft, and Hayden Pearce
- Abstract
Supplementary Tables 1 - 5
- Published
- 2023
- Full Text
- View/download PDF
5. Data from Tissue-Resident Memory T Cells in Pancreatic Ductal Adenocarcinoma Coexpress PD-1 and TIGIT and Functional Inhibition Is Reversible by Dual Antibody Blockade
- Author
-
Paul Moss, Keith Roberts, Gary Middleton, Rachel M. Brown, Brinder S. Mahon, Sarah Powell-Brett, Éanna Fennell, Matthew R. Pugh, Francesca Marcon, Simon J. Davis, Richard Cornall, Richard Powell, Sandra Margielewska-Davies, Samantha M. Nicol, Wayne Croft, and Hayden Pearce
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor clinical outlook. Responses to immune checkpoint blockade are suboptimal and a much more detailed understanding of the tumor immune microenvironment is needed if this situation is to be improved. Here, we characterized tumor-infiltrating T-cell populations in patients with PDAC using cytometry by time of flight (CyTOF) and single-cell RNA sequencing. T cells were the predominant immune cell subset observed within tumors. Over 30% of CD4+ T cells expressed a CCR6+CD161+ Th17 phenotype and 17% displayed an activated regulatory T-cell profile. Large populations of CD8+ tissue-resident memory (TRM) T cells were also present and expressed high levels of programmed cell death protein 1 (PD-1) and TIGIT. A population of putative tumor-reactive CD103+CD39+ T cells was also observed within the CD8+ tumor-infiltrating lymphocytes population. The expression of PD-1 ligands was limited largely to hemopoietic cells whilst TIGIT ligands were expressed widely within the tumor microenvironment. Programmed death-ligand 1 and CD155 were expressed within the T-cell area of ectopic lymphoid structures and colocalized with PD-1+TIGIT+ CD8+ T cells. Combinatorial anti–PD-1 and TIGIT blockade enhanced IFNγ secretion and proliferation of T cells in the presence of PD-1 and TIGIT ligands. As such, we showed that the PDAC microenvironment is characterized by the presence of substantial populations of TRM cells with an exhausted PD-1+TIGIT+ phenotype where dual checkpoint receptor blockade represents a promising avenue for future immunotherapy.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Material from Combined BRAF, EGFR, and MEK Inhibition in Patients with BRAFV600E-Mutant Colorectal Cancer
- Author
-
Eric Van Cutsem, Fatima Rangwala, Bijoyesh Mookerjee, Severine Bettinger, Savina Jaeger, Jan C. Brase, A. Scott Jung, Filip De Vos, Yves Humblet, Peter J. O'Dwyer, Rona Yaeger, Josep Tabernero, Michael S. Gordon, Kei Muro, Gary Middleton, Salvatore Siena, Autumn J. McRee, Antoine Hollebecque, Johanna C. Bendell, Takayuki Yoshino, Jan H.M. Schellens, Chloe E. Atreya, Thierry André, and Ryan B. Corcoran
- Abstract
Supplementary Material
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Development and Validation of a Combined Hypoxia and Immune Prognostic Classifier for Head and Neck Cancer
- Author
-
Hisham Mehanna, Tanguy Y. Seiwert, Benjamin E. Willcox, Jean-Baptiste Cazier, Catharine M. L. West, Jonathan Deeks, Andrew D. Beggs, Daniel A. Tennant, Gary Middleton, Kalu U.E. Ogbureke, Steven P. Lee, Arun Khattri, Riyue Bao, Gordon B. Ryan, Baksho Kaul, Margaret Hartley, Jennifer L. Bryant, Nikolaos Batis, Rachel J. Spruce, Helen R. Valentine, Sandra V. von Zeidler, Christopher J. Bagnall, Neeraj Lal, Lucinda Archer, Maha Ibrahim, Albert N. Menezes, and Jill M. Brooks
- Abstract
Purpose:Intratumoral hypoxia and immunity have been correlated with patient outcome in various tumor settings. However, these factors are not currently considered for treatment selection in head and neck cancer (HNC) due to lack of validated biomarkers. Here we sought to develop a hypoxia-immune classifier with potential application in patient prognostication and prediction of response to targeted therapy.Experimental Design:A 54-gene hypoxia-immune signature was constructed on the basis of literature review. Gene expression was analyzed in silico using the The Cancer Genome Atlas (TCGA) HNC dataset (n = 275) and validated using two independent cohorts (n = 130 and 123). IHC was used to investigate the utility of a simplified protein signature. The spatial distribution of hypoxia and immune markers was examined using multiplex immunofluorescence staining.Results:Unsupervised hierarchical clustering of TCGA dataset (development cohort) identified three patient subgroups with distinct hypoxia-immune phenotypes and survival profiles: hypoxialow/immunehigh, hypoxiahigh/immunelow, and mixed, with 5-year overall survival (OS) rates of 71%, 51%, and 49%, respectively (P = 0.0015). The prognostic relevance of the hypoxia-immune gene signature was replicated in two independent validation cohorts. Only PD-L1 and intratumoral CD3 protein expression were associated with improved OS on multivariate analysis. Hypoxialow/immunehigh and hypoxiahigh/immunelow tumors were overrepresented in “inflamed” and “immune-desert” microenvironmental profiles, respectively. Multiplex staining demonstrated an inverse correlation between CA-IX expression and prevalence of intratumoral CD3+ T cells (r = −0.5464; P = 0.0377), further corroborating the transcription-based classification.Conclusions:We developed and validated a hypoxia-immune prognostic transcriptional classifier, which may have clinical application to guide the use of hypoxia modification and targeted immunotherapies for the treatment of HNC.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Data from Development and Validation of a Combined Hypoxia and Immune Prognostic Classifier for Head and Neck Cancer
- Author
-
Hisham Mehanna, Tanguy Y. Seiwert, Benjamin E. Willcox, Jean-Baptiste Cazier, Catharine M. L. West, Jonathan Deeks, Andrew D. Beggs, Daniel A. Tennant, Gary Middleton, Kalu U.E. Ogbureke, Steven P. Lee, Arun Khattri, Riyue Bao, Gordon B. Ryan, Baksho Kaul, Margaret Hartley, Jennifer L. Bryant, Nikolaos Batis, Rachel J. Spruce, Helen R. Valentine, Sandra V. von Zeidler, Christopher J. Bagnall, Neeraj Lal, Lucinda Archer, Maha Ibrahim, Albert N. Menezes, and Jill M. Brooks
- Abstract
Figure S1. Gene order within the cluster defined groups. Figure S2. Analysis of individual hypoxia and immune gene signatures. Figure S3. Flow diagram showing progress of patient samples through study for Correlate cohort Figure S4. Relationship between heatmap defined hypoxia-immune subgroups and T stage. Figure S5 Expression of individual hypoxia- or immune-response related genes within the heat-map defined subgroups. Figure S6. Correlation of PD-L1 or CA-IX protein expression with survival. Figure S7 Correlation of gene and protein expression. Figure S8. Spatial interactions of hypoxia and immune cells: Vectra images of multiplex stained HNC sections. Table S1 Treatment schedules Table S2. Genes in the RNASeq panel/TCGA dataset Table S3. Relationship of hypoxia-immune classifier to other prognostic variables Table S4. Summary of immunohistochemistry staining Table S5. Summary of statistical analyses for IHC staining
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Tumor-Derived GM-CSF Promotes Granulocyte Immunosuppression in Mesothelioma Patients
- Author
-
Raffit Hassan, Carmela De Santo, Gary Middleton, Maryalice Stetler-Stevenson, Seth M. Steinberg, Tim F. Greten, Firouzeh Korangy, Jingli Zhang, Betsy Morrow, Constance Yuan, Bahar Guliz Yenidunya, Neha Wali, Anish Thomas, Francis Mussai, Suzanne Graef, and Swati Khanna
- Abstract
Purpose: The cross-talk between tumor cells, myeloid cells, and T cells can play a critical role in tumor pathogenesis and response to immunotherapies. Although the etiology of mesothelioma is well understood, the impact of mesothelioma tumor cells on the surrounding immune microenvironment is less well studied. In this study, the effect of the mesothelioma tumor microenvironment on circulating and infiltrating granulocytes and T cells is investigated.Experimental Design: Tumor tissues and peripheral blood from mesothelioma patients were evaluated for presence of granulocytes, which were then tested for their T-cell suppression potential. Different cocultures of granulocytes and/or mesothelioma tumor cells and/or T cells were set up to identify the mechanism of T-cell inhibition.Results: Analysis of human tumors showed that the mesothelioma microenvironment is enriched in infiltrating granulocytes, which inhibit T-cell proliferation and activation. Characterization of the whole blood at diagnosis identified similar, circulating, immunosuppressive CD11b+CD15+HLADR− granulocytes at increased frequency compared with healthy controls. Culture of healthy-donor granulocytes with human mesothelioma cells showed that GM-CSF upregulates NOX2 expression and the release of reactive oxygen species (ROS) from granulocytes, resulting in T-cell suppression. Immunohistochemistry and transcriptomic analysis revealed that a majority of mesothelioma tumors express GM-CSF and that higher GM-CSF expression correlated with clinical progression. Blockade of GM-CSF with neutralizing antibody, or ROS inhibition, restored T-cell proliferation, suggesting that targeting of GM-CSF could be of therapeutic benefit in these patients.Conclusions: Our study presents the mechanism behind the cross-talk between mesothelioma tumors and the immune microenvironment and indicates that targeting GM-CSF could be a novel treatment strategy to augment immunotherapy in patients with mesothelioma. Clin Cancer Res; 24(12); 2859–72. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
10. Data from BRAF-Mutant Transcriptional Subtypes Predict Outcome of Combined BRAF, MEK, and EGFR Blockade with Dabrafenib, Trametinib, and Panitumumab in Patients with Colorectal Cancer
- Author
-
Ryan B. Corcoran, Eduard Gasal, Fatima Rangwala, Jan C. Brase, John M. Millholland, Eric Van Cutsem, Filip De Vos, Peter J. O'Dwyer, Rona Yaeger, Josep Tabernero, Michael S. Gordon, Salvatore Siena, Autumn J. McRee, Antoine Hollebecque, Johanna C. Bendell, Takayuki Yoshino, Jan H.M. Schellens, Chloe E. Atreya, Thierry André, Catarina D. Campbell, Yiqun Yang, and Gary Middleton
- Abstract
Purpose:The influence of the transcriptional and immunologic context of mutations on therapeutic outcomes with targeted therapy in cancer has not been well defined. BRAF V600E–mutant (BM) colorectal cancer comprises two main transcriptional subtypes, BM1 and BM2. We sought to determine the impact of BM subtype, as well as distinct biological features of those subtypes, on response to BRAF/MEK/EGFR inhibition in patients with colorectal cancer.Patients and Methods:Paired fresh tumor biopsies were acquired at baseline and on day 15 of treatment from all consenting patients with BM colorectal cancer enrolled in a phase II clinical trial of dabrafenib, trametinib, and panitumumab. For each sample, BM subtype, cell cycle, and immune gene signature expression were determined using RNA-sequencing (RNA-seq), and a Cox proportional hazards model was applied to determine association with progression-free survival (PFS).Results:Confirmed response rates, median PFS, and median overall survival (OS) were higher in BM1 subtype patients compared with BM2 subtype patients. Evaluation of immune contexture identified greater immune reactivity in BM1, whereas cell-cycle signatures were more highly expressed in BM2. A multivariate model of PFS incorporating BM subtype plus immune and cell-cycle signatures revealed that BM subtype encompasses the majority of the effect.Conclusions:BM subtype is significantly associated with the outcome of combination dabrafenib, trametinib, and panitumumab therapy and may serve as a standalone predictive biomarker beyond mutational status. Our findings support a more nuanced approach to targeted therapeutic decisions that incorporates assessment of transcriptional context.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Data from BRAF-Mutant Transcriptional Subtypes Predict Outcome of Combined BRAF, MEK, and EGFR Blockade with Dabrafenib, Trametinib, and Panitumumab in Patients with Colorectal Cancer
- Author
-
Ryan B. Corcoran, Eduard Gasal, Fatima Rangwala, Jan C. Brase, John M. Millholland, Eric Van Cutsem, Filip De Vos, Peter J. O'Dwyer, Rona Yaeger, Josep Tabernero, Michael S. Gordon, Salvatore Siena, Autumn J. McRee, Antoine Hollebecque, Johanna C. Bendell, Takayuki Yoshino, Jan H.M. Schellens, Chloe E. Atreya, Thierry André, Catarina D. Campbell, Yiqun Yang, and Gary Middleton
- Abstract
Supplementary Data
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary figure 2 from Tumor-Derived GM-CSF Promotes Granulocyte Immunosuppression in Mesothelioma Patients
- Author
-
Raffit Hassan, Carmela De Santo, Gary Middleton, Maryalice Stetler-Stevenson, Seth M. Steinberg, Tim F. Greten, Firouzeh Korangy, Jingli Zhang, Betsy Morrow, Constance Yuan, Bahar Guliz Yenidunya, Neha Wali, Anish Thomas, Francis Mussai, Suzanne Graef, and Swati Khanna
- Abstract
Mesothelioma granulocytes suppress T cell activation
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figs and Tables from Tumor-Derived GM-CSF Promotes Granulocyte Immunosuppression in Mesothelioma Patients
- Author
-
Raffit Hassan, Carmela De Santo, Gary Middleton, Maryalice Stetler-Stevenson, Seth M. Steinberg, Tim F. Greten, Firouzeh Korangy, Jingli Zhang, Betsy Morrow, Constance Yuan, Bahar Guliz Yenidunya, Neha Wali, Anish Thomas, Francis Mussai, Suzanne Graef, and Swati Khanna
- Abstract
Supplementary Figs and Tables
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary figure 3 from Tumor-Derived GM-CSF Promotes Granulocyte Immunosuppression in Mesothelioma Patients
- Author
-
Raffit Hassan, Carmela De Santo, Gary Middleton, Maryalice Stetler-Stevenson, Seth M. Steinberg, Tim F. Greten, Firouzeh Korangy, Jingli Zhang, Betsy Morrow, Constance Yuan, Bahar Guliz Yenidunya, Neha Wali, Anish Thomas, Francis Mussai, Suzanne Graef, and Swati Khanna
- Abstract
Mesothelioma upregulates ROS production by granulocytes
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary figure 1 from Tumor-Derived GM-CSF Promotes Granulocyte Immunosuppression in Mesothelioma Patients
- Author
-
Raffit Hassan, Carmela De Santo, Gary Middleton, Maryalice Stetler-Stevenson, Seth M. Steinberg, Tim F. Greten, Firouzeh Korangy, Jingli Zhang, Betsy Morrow, Constance Yuan, Bahar Guliz Yenidunya, Neha Wali, Anish Thomas, Francis Mussai, Suzanne Graef, and Swati Khanna
- Abstract
Arginase activity does not drive immunosuppressive granulocyte function
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary figure 4 from Tumor-Derived GM-CSF Promotes Granulocyte Immunosuppression in Mesothelioma Patients
- Author
-
Raffit Hassan, Carmela De Santo, Gary Middleton, Maryalice Stetler-Stevenson, Seth M. Steinberg, Tim F. Greten, Firouzeh Korangy, Jingli Zhang, Betsy Morrow, Constance Yuan, Bahar Guliz Yenidunya, Neha Wali, Anish Thomas, Francis Mussai, Suzanne Graef, and Swati Khanna
- Abstract
GM-CSF drives ROS release in granulocytes
- Published
- 2023
- Full Text
- View/download PDF
17. Spatial determination and prognostic impact of the fibroblast transcriptome in pancreatic ductal adenocarcinoma
- Author
-
Wayne Croft, Hayden Pearce, Sandra Margielewska-Davies, Lindsay Lim, Samantha M. Nicol, Fouzia Zayou, Daniel Blakeway, Francesca Marcon, Sarah Powell-Brett, Brinder Mahon, Reena Merard, Jianmin Zuo, Gary Middleton, Keith Roberts, Rachel M. Brown, and Paul Moss
- Abstract
Pancreatic ductal adenocarcinoma has a poor clinical outcome and responses to immunotherapy are suboptimal. Stromal fibroblasts are a dominant but heterogenous population within the tumor microenvironment and therapeutic targeting of stromal subsets may have therapeutic utility. Here we combined spatial transcriptomics and scRNA-Seq to define the transcriptome of tumor-proximal and tumor-distal cancer-associated fibroblasts (CAFs) and linked this to clinical outcome. Tumor-proximal fibroblasts comprised large populations of myofibroblasts, strongly expressed podoplanin, and were enriched for Wnt ligand signaling. In contrast, inflammatory CAFs were dominant within tumor-distal subsets and expressed complement components and the Wnt-inhibitor SFRP2. Poor clinical outcome was correlated with elevated HIF-1α and podoplanin whilst high level expression of inflammatory and complement genes was predictive of extended survival. These findings demonstrate the extreme transcriptional heterogeneity of CAFs and its determination by apposition to tumor. Selective targeting of tumor-proximal subsets, potentially combined with HIF-1α inhibition and immune stimulation, may offer a multimodal therapeutic approach for this disease.Graphical Abstract
- Published
- 2023
- Full Text
- View/download PDF
18. Capecitabine Versus Active Monitoring in Stable or Responding Metastatic Colorectal Cancer After 16 Weeks of First-Line Therapy: Results of the Randomized FOCUS4-N Trial
- Author
-
Janet Graham, Mahesh K. B. Parmar, Emma Yates, Tim Maughan, Jenny F. Seligmann, Louise Brown, David Fisher, Kai-Keen Shiu, Fiona Collinson, Ewan Brown, Philip Quirke, Stephen Falk, Richard H. Wilson, Susan D. Richman, Gary Middleton, Harpreet Wasan, Richard Kaplan, Richard Adams, Rachel Butler, Matthew T. Seymour, and Leslie Samuel
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,business.industry ,Active monitoring ,MEDLINE ,medicine.disease ,Capecitabine ,First line therapy ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
PURPOSE Despite extensive randomized evidence supporting the use of treatment breaks in metastatic colorectal cancer (mCRC), they are not universally offered to patients despite improvements in quality of life without detriment to overall survival (OS). FOCUS4-N was set up to explore the impact of oral maintenance therapy in patients who are responding to first-line therapy. METHODS FOCUS4 was a molecularly stratified trial program that registered patients with newly diagnosed mCRC. The FOCUS4-N trial was offered to patients in whom a targeted subtrial was unavailable or biomarker tests failed. Patients were randomly assigned using a 1:1 ratio between maintenance capecitabine and active monitoring (AM). The primary outcome was progression-free survival (PFS) with secondary outcomes including OS toxicity and tolerability. RESULTS Between March 2014 and March 2020, 254 patients were randomly assigned (127 to capecitabine and 127 to AM) across 88 UK sites. Baseline characteristics were balanced. There was strong evidence of efficacy for PFS (hazard ratio = 0.40; 95% CI, 0.21 to 0.75; P < .0001), but no significant improvement in OS (hazard ratio, 0.93; 95% CI, 0.69 to 1.27; P = .66) was observed. Compliance with treatment was good, and toxicity from capecitabine versus AM was as expected with grade ≥ 2 fatigue (25% v 12%), diarrhea (23% v 13%), and hand-foot syndrome (26% v 3%). Quality of life showed little difference between the groups. CONCLUSION Despite strong evidence of disease control with maintenance therapy, OS remains unaffected and FOCUS4-N provides additional evidence to support the use of treatment breaks as safe management alternatives for patients who are stable or responding to first-line treatment for mCRC. Capecitabine without bevacizumab may be used to extend PFS in the interval after 16 weeks of first-line therapy.
- Published
- 2021
- Full Text
- View/download PDF
19. A highly predictive autoantibody-based biomarker panel for prognosis in early-stage NSCLC with potential therapeutic implications
- Author
-
Gary Middleton, Alex G. Richter, Akshay J Patel, Ti-Myen Tan, Jonathan M. Blackburn, and Babu Naidu
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Protein Array Analysis ,Disease ,Article ,Tumour biomarkers ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Survivorship curve ,Biomarkers, Tumor ,medicine ,Adjuvant therapy ,Humans ,Stage (cooking) ,Lung cancer ,Survival rate ,Aged ,Autoantibodies ,business.industry ,Autoantibody ,Computational Biology ,Prognosis ,medicine.disease ,ROC Curve ,Biomarker (medicine) ,Female ,business ,Non-small-cell lung cancer - Abstract
Background Lung cancer is the leading cause of cancer-related death worldwide. Surgical resection remains the definitive curative treatment for early-stage disease offering an overall 5-year survival rate of 62%. Despite careful case selection, a significant proportion of early-stage cancers relapse aggressively within the first year post-operatively. Identification of these patients is key to accurate prognostication and understanding the biology that drives early relapse might open up potential novel adjuvant therapies. Methods We performed an unsupervised interrogation of >1600 serum-based autoantibody biomarkers using an iterative machine-learning algorithm. Results We identified a 13 biomarker signature that was highly predictive for survivorship in post-operative early-stage lung cancer; this outperforms currently used autoantibody biomarkers in solid cancers. Our results demonstrate significantly poor survivorship in high expressers of this biomarker signature with an overall 5-year survival rate of 7.6%. Conclusions We anticipate that the data will lead to the development of an off-the-shelf prognostic panel and further that the oncogenic relevance of the proteins recognised in the panel may be a starting point for a new adjuvant therapy.
- Published
- 2021
- Full Text
- View/download PDF
20. Germline genetic variation and predicting immune checkpoint inhibitor induced toxicity
- Author
-
Ik Shin Chin, Aman Khan, Anna Olsson-Brown, Sophie Papa, Gary Middleton, and Claire Palles
- Subjects
Genetics ,Molecular Biology ,Genetics (clinical) - Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionised the treatment of various cancer types. ICIs reinstate T-cell function to elicit an anti-cancer immune response. The resulting immune response can however have off-target effects which manifest as autoimmune type serious immune-related adverse events (irAE) in ~10–55% of patients treated. It is currently challenging to predict both who will experience irAEs and to what severity. Identification of patients at high risk of serious irAE would revolutionise patient care. While the pathogenesis driving irAE development is still unclear, host genetic factors are proposed to be key determinants of these events. This review presents current evidence supporting the role of the host genome in determining risk of irAE. We summarise the spectrum and timing of irAEs following treatment with ICIs and describe currently reported germline genetic variation associated with expression of immuno-modulatory factors within the cancer immunity cycle, development of autoimmune disease and irAE occurrence. We propose that germline genetic determinants of host immune function and autoimmune diseases could also explain risk of irAE development. We also endorse genome-wide association studies of patients being treated with ICIs to identify genetic variants that can be used in polygenic risk scores to predict risk of irAE.
- Published
- 2022
- Full Text
- View/download PDF
21. Using DNA sequencing data to quantify T cell fraction and therapy response
- Author
-
Javier Herrero, Gary Middleton, Charles Swanton, John Edwards, Ricky Thakrar, David Pearce, Domenic Marrone, Mickael Escudero, Christian Ottensmeier, Francisco Gimeno Valiente, Jonas Demeulemeester, James Black, Kevin Litchfield, Claire Wilson, Neal Navani, Helen Lowe, JAMES READING, Nicholas McGranahan, Diana Johnson, Hugo Aerts, Benny Chain, Miljana Tanic, and Robert Hynds
- Subjects
Multidisciplinary ,Tumor-infiltrating lymphocytes ,Somatic cell ,T-cell receptor excision circles ,medicine.medical_treatment ,T cell ,Ipilimumab ,Immunotherapy ,Biology ,medicine.anatomical_structure ,Immune system ,medicine ,Cancer research ,Exome ,medicine.drug - Abstract
The immune microenvironment influences tumour evolution and can be both prognostic and predict response to immunotherapy1,2. However, measurements of tumour infiltrating lymphocytes (TILs) are limited by a shortage of appropriate data. Whole-exome sequencing (WES) of DNA is frequently performed to calculate tumour mutational burden and identify actionable mutations. Here we develop T cell exome TREC tool (T cell ExTRECT), a method for estimation of T cell fraction from WES samples using a signal from T cell receptor excision circle (TREC) loss during V(D)J recombination of the T cell receptor-α gene (TCRA (also known as TRA)). TCRA T cell fraction correlates with orthogonal TIL estimates and is agnostic to sample type. Blood TCRA T cell fraction is higher in females than in males and correlates with both tumour immune infiltrate and presence of bacterial sequencing reads. Tumour TCRA T cell fraction is prognostic in lung adenocarcinoma. Using a meta-analysis of tumours treated with immunotherapy, we show that tumour TCRA T cell fraction predicts immunotherapy response, providing value beyond measuring tumour mutational burden. Applying T cell ExTRECT to a multi-sample pan-cancer cohort reveals a high diversity of the degree of immune infiltration within tumours. Subclonal loss of 12q24.31–32, encompassing SPPL3, is associated with reduced TCRA T cell fraction. T cell ExTRECT provides a cost-effective technique to characterize immune infiltrate alongside somatic changes. A robust, cost-effective technique based on whole-exome sequencing data can be used to characterize immune infiltrates, relate the extent of these infiltrates to somatic changes in tumours, and enables prediction of tumour responses to immune checkpoint inhibition therapy.
- Published
- 2021
- Full Text
- View/download PDF
22. COVID-19: Third dose booster vaccine effectiveness against breakthrough coronavirus infection, hospitalisations and death in patients with cancer: A population-based study
- Author
-
Lennard Y.W. Lee, Maria C. Ionescu, Thomas Starkey, Martin Little, Michael Tilby, Arvind R. Tripathy, Hayley S. Mckenzie, Youssra Al-Hajji, Nathan Appanna, Matthew Barnard, Liza Benny, Alexander Burnett, Emma L. Cattell, James J. Clark, Sam Khan, Qamar Ghafoor, Hari Panneerselvam, George Illsley, Catherine Harper-Wynne, Rosie J. Hattersley, Alvin JX. Lee, Oliver Lomas, Justin KH. Liu, Amanda McCauley, Matthew Pang, Jennifer S. Pascoe, James R. Platt, Grisma Patel, Vijay Patel, Vanessa A. Potter, Amelia Randle, Anne S. Rigg, Tim M. Robinson, Tom W. Roques, René L. Roux, Stefan Rozmanowski, Harriet Taylor, Mark H. Tuthill, Isabella Watts, Sarah Williams, Andrew Beggs, Tim Iveson, Siow M. Lee, Gary Middleton, Mark Middleton, Andrew Protheroe, Matthew W. Fittall, Tom Fowler, Peter Johnson, Emma Kinloch, Emily Lam, Gillian Murphy, Malcolm Rhodes, Kate Robinson, Sanskriti Swarup, Keeley Bernhardt, Jola Bytyci, Yuxin Ying, Sukhmunni Johal, and Remarez Sheehan
- Subjects
Hospitalization ,Cancer Research ,Oncology ,Neoplasms ,Vaccination ,COVID-19 ,Humans ,Vaccine Efficacy ,Pandemics - Abstract
People living with cancer and haematological malignancies are at an increased risk of hospitalisation and death following infection with acute respiratory syndrome coronavirus 2. Coronavirus third dose vaccine boosters are proposed to boost waning immune responses in immunocompromised individuals and increase coronavirus protection; however, their effectiveness has not yet been systematically evaluated.This study is a population-scale real-world evaluation of the United Kingdom's third dose vaccine booster programme for cancer patients from 8th December 2020 to 7th December 2021. The cancer cohort comprises individuals from Public Health England's national cancer dataset, excluding individuals less than 18 years. A test-negative case-control design was used to assess the third dose booster vaccine effectiveness. Multivariable logistic regression models were fitted to compare risk in the cancer cohort relative to the general population.The cancer cohort comprised of 2,258,553 tests from 361,098 individuals. Third dose boosters were evaluated by reference to 87,039,743 polymerase chain reaction coronavirus tests. Vaccine effectiveness against breakthrough infections, symptomatic infections, coronavirus hospitalisation and death in cancer patients were 59.1%, 62.8%, 80.5% and 94.5%, respectively. Lower vaccine effectiveness was associated with a cancer diagnosis within 12 months, lymphoma, recent systemic anti-cancer therapy (SACT) or radiotherapy. Patients with lymphoma had low levels of protection from symptomatic disease. In spite of third dose boosters, following multivariable adjustment, individuals with cancer remain at an increased risk of coronavirus hospitalisation and death compared to the population control (OR 3.38, 3.01, respectively. p lt; 0.001 for both).Third dose boosters are effective for most individuals with cancer, increasing protection from coronavirus. However, their effectiveness is heterogenous and lower than the general population. Many patients with cancer will remain at the increased risk of coronavirus infections even after 3 doses. In the case of patients with lymphoma, there is a particularly strong disparity of vaccine effectiveness against breakthrough infection and severe disease. Breakthrough infections will disrupt cancer care and treatment with potentially adverse consequences on survival outcomes. The data support the role of vaccine boosters in preventing severe disease, and further pharmacological intervention to prevent transmission and aid viral clearance to limit the disruption of cancer care as the delivery of care continues to evolve during the coronavirus pandemic.
- Published
- 2022
23. Paradox breaker BRAF inhibitors have comparable potency and MAPK pathway reactivation to encorafenib in BRAF mutant colorectal cancer
- Author
-
Oliver J Pickles, Andrew D Beggs, Gary Middleton, Louise Tee, and Aneta Drozd
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,medicine.medical_treatment ,Context (language use) ,colorectal cancer ,encorafenib ,Targeted therapy ,BRAF ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Encorafenib ,medicine ,Vemurafenib ,neoplasms ,paradox breaker ,Cetuximab ,business.industry ,Melanoma ,Binimetinib ,medicine.disease ,digestive system diseases ,PLX8394 ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,business ,medicine.drug ,Research Paper - Abstract
The BEACON CRC trial demonstrated a survival advantage over chemotherapy for a combination of targeted agents comprising the potent BRAF inhibitor encorafenib together with cetuximab and binimetinib. Resistance to BRAF inhibition in CRC arises in part through the generation and activation of RAF dimers resulting in MEK-ERK pathway reactivation. Paradox breaker BRAF inhibitors, such as PLX8394, are designed to inhibit RAF dimer formation. We analyzed whether paradox breakers reduce pathway reactivation and so have enhanced potency compared with encorafenib in BRAF mutant CRC. The potency of encorafenib and PLX8394 was greater than vemurafenib and the degree of pathway reactivation somewhat less. However, dose response curves for encorafenib and PLX8394 were similar and there was no significant differences in degree of pathway reactivation. To our knowledge these data represent the first comparative data of encorafenib and paradox breaker inhibitors in BRAF mutant CRC. Whilst these results support further investigation of PLX8394, all three agents tested reactivated the pathway in melanoma cells, a disease in which monotherapy is effective. Strategies focused on restricting RAF dimerization fail to address the impact that specific context of BRAF mutation in CRC has on targeted therapy outcomes.
- Published
- 2020
24. BRAF-Mutant Transcriptional Subtypes Predict Outcome of Combined BRAF, MEK, and EGFR Blockade with Dabrafenib, Trametinib, and Panitumumab in Patients with Colorectal Cancer
- Author
-
Autumn J. McRee, Takayuki Yoshino, Filip de Vos, Chloe E. Atreya, Eduard Gasal, Jan H.M. Schellens, Salvatore Siena, Eric Van Cutsem, Rona Yaeger, Thierry André, John Millholland, Ryan B. Corcoran, Antoine Hollebecque, Catarina D. Campbell, Josep Tabernero, Peter J. O'Dwyer, Fatima Rangwala, Johanna C. Bendell, Jan C. Brase, Yiqun Yang, Gary Middleton, and Michael S. Gordon
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pyridones ,Colorectal cancer ,medicine.medical_treatment ,Oncology and Carcinogenesis ,MAP Kinase Kinase 1 ,Context (language use) ,Pyrimidinones ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Clinical Research ,Internal medicine ,Oximes ,Antineoplastic Combined Chemotherapy Protocols ,Genetics ,medicine ,Humans ,Panitumumab ,Oncology & Carcinogenesis ,Cancer ,Trametinib ,Neoplastic ,Tumor ,business.industry ,Imidazoles ,Dabrafenib ,Cell cycle ,Prognosis ,medicine.disease ,Colo-Rectal Cancer ,Survival Rate ,ErbB Receptors ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Mutation ,Colorectal Neoplasms ,Digestive Diseases ,business ,Biomarkers ,medicine.drug - Abstract
Purpose: The influence of the transcriptional and immunologic context of mutations on therapeutic outcomes with targeted therapy in cancer has not been well defined. BRAF V600E–mutant (BM) colorectal cancer comprises two main transcriptional subtypes, BM1 and BM2. We sought to determine the impact of BM subtype, as well as distinct biological features of those subtypes, on response to BRAF/MEK/EGFR inhibition in patients with colorectal cancer. Patients and Methods: Paired fresh tumor biopsies were acquired at baseline and on day 15 of treatment from all consenting patients with BM colorectal cancer enrolled in a phase II clinical trial of dabrafenib, trametinib, and panitumumab. For each sample, BM subtype, cell cycle, and immune gene signature expression were determined using RNA-sequencing (RNA-seq), and a Cox proportional hazards model was applied to determine association with progression-free survival (PFS). Results: Confirmed response rates, median PFS, and median overall survival (OS) were higher in BM1 subtype patients compared with BM2 subtype patients. Evaluation of immune contexture identified greater immune reactivity in BM1, whereas cell-cycle signatures were more highly expressed in BM2. A multivariate model of PFS incorporating BM subtype plus immune and cell-cycle signatures revealed that BM subtype encompasses the majority of the effect. Conclusions: BM subtype is significantly associated with the outcome of combination dabrafenib, trametinib, and panitumumab therapy and may serve as a standalone predictive biomarker beyond mutational status. Our findings support a more nuanced approach to targeted therapeutic decisions that incorporates assessment of transcriptional context.
- Published
- 2020
- Full Text
- View/download PDF
25. Characterising the impact of pneumonia on outcome in non-small cell lung cancer: identifying preventative strategies
- Author
-
Peter Nightingale, Alex G. Richter, Gary Middleton, Mark T. Drayson, Akshay J Patel, and Babu Naidu
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,business.industry ,Respiratory infection ,non-small cell lung cancer (NSCLC) ,Odds ratio ,medicine.disease ,Culprit ,respiratory tract diseases ,Vaccination ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Cohort ,medicine ,Infection control ,Original Article ,030212 general & internal medicine ,Lung cancer ,business - Abstract
BACKGROUND: Infections remain a part of the natural course of cancer, and lung cancer patients often present with some form of respiratory infection that can lead to their ultimate demise. METHODS: Data was gathered concerning all unplanned hospital admissions (UHAs) to our centre from three separate patient cohorts; non-small cell lung cancer (NSCLC) patients (cohort 1), “other cancer” patients (breast, prostate, colon) (cohort 2) and all non-cancer patients (cohort 3). RESULTS: Across the three cohorts, there were 455, 1,190 and 54,158 individual patient UHAs to our centre respectively. Within the NSCLC cohort, 164 UHAs were as a direct result of pneumonia (36.0%), compared to 1.3% and 2.2% in the other two cohorts (P
- Published
- 2020
- Full Text
- View/download PDF
26. Use of an Integrated Pan-Cancer Oncology Enrichment Next-Generation Sequencing Assay to Measure Tumour Mutational Burden and Detect Clinically Actionable Variants
- Author
-
John M Findlay, Gerald Martin, Jean-Francois Laes, Matthew Smith, Gary Middleton, Andrew D Beggs, Valerie Pestinger, Toju Sillo, and Phillipe Taniere
- Subjects
Pharmacology ,Oncology ,FASTQ format ,medicine.medical_specialty ,Pan cancer ,Microsatellite instability ,General Medicine ,Biology ,medicine.disease ,Human genetics ,DNA sequencing ,Structural variation ,Internal medicine ,Genetics ,medicine ,Molecular Medicine ,Original Research Article ,Indel ,Gene - Abstract
Introduction The identification of tumour mutational burden (TMB) as a biomarker of response to programmed cell death protein 1 (PD-1) immunotherapy has necessitated the development of genomic assays to measure this. We carried out comprehensive molecular profiling of cancers using the Illumina TruSight Oncology 500 (TSO500) panel and compared these to whole-genome sequencing (WGS). Methods Cancer samples derived from formalin-fixed material were profiled on the TSO500 panel, sequenced on an Illumina NextSeq 500 instrument and processed through the TSO500 Docker pipeline. Either FASTQ files (PierianDx) or vcf files (OncoKDM) were processed to understand clinical actionability. Results In total, 108 samples (a mixture of colorectal, lung, oesophageal and control samples) were processed via the DNA panel. There was good correlation between TMB, single-nucleotide variants (SNVs), indels and copy-number variations as predicted by TSO500 and WGS (R2 > 0.9) and good reproducibility, with less than 5% variability between repeated controls. For the RNA panel, 13 samples were processed, with all known fusions observed via orthogonal techniques. For clinical actionability, 72 tier 1 variants and 297 tier 2 variants were detected, with clinical trials identified for all patients. Conclusions The TSO500 assay accurately measures TMB, microsatellite instability, SNVs, indels, copy-number/structural variation and gene fusions when compared to WGS and orthogonal technologies. Coupled with a clinical annotation pipeline, this provides a powerful methodology for identification of clinically actionable variants.
- Published
- 2020
27. The role of B lymphocytes in the immuno-biology of non-small-cell lung cancer
- Author
-
Alex G. Richter, Akshay J Patel, Gary Middleton, and Mark T. Drayson
- Subjects
Male ,Cancer Research ,Lung Neoplasms ,Immunology ,non-small cell lung cancer (NSCLC) ,Review ,Biology ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Carcinoma, Non-Small-Cell Lung ,Immunoglobulin ,medicine ,Tumour -infiltrating B lymphocyte (TIL-B) ,Humans ,Immunology and Allergy ,Lung cancer ,030304 developmental biology ,Non-small-cell lung cancer (NSCLC) ,B-Lymphocytes ,0303 health sciences ,B lymphocyte ,Cancer ,medicine.disease ,Oncology ,030220 oncology & carcinogenesis ,Humoral Immunity ,Humoral immunity ,Cancer research ,biology.protein ,Female ,Non small cell ,Antibody ,Carcinogenesis - Abstract
Tumour-infiltrating immune cells have been widely implicated to play a significant role in carcinogenesis, through both pro- or anti-tumour effects. The multi-faceted effects of lung cancer associated T lymphocytes have been extensively studied, and yet, the role of B lymphocytes remains an area less studied. In this review, we will describe the current understanding of the role of tumour-infiltrating B lymphocytes in NSCLC, discuss their prognostic significance, their functionality within the tumour microenvironment and ultimately how we might harness B-cell biology to develop B-cell therapeutic strategies in cancer.
- Published
- 2020
- Full Text
- View/download PDF
28. Namilumab or infliximab compared with standard of care in hospitalised patients with COVID-19 (CATALYST): a randomised, multicentre, multi-arm, multistage, open-label, adaptive, phase 2, proof-of-concept trial
- Author
-
Benjamin A Fisher, Tonny Veenith, Daniel Slade, Charlotte Gaskell, Matthew Rowland, Tony Whitehouse, James Scriven, Dhruv Parekh, Madhu S Balasubramaniam, Graham Cooke, Nick Morley, Zoe Gabriel, Matthew P Wise, Joanna Porter, Helen McShane, Ling-Pei Ho, Philip N Newsome, Anna Rowe, Rowena Sharpe, David R Thickett, Julian Bion, Simon Gates, Duncan Richards, Pamela Kearns, Bryan Williams, Rebecca Turner, Vincenzo Libri, Francis Mussai, Gary Middleton, Sarah Bowden, Mansoor Bangash, Fang Gao-Smith, Jaimin Patel, Elizabeth Sapey, Mark Thomas, Mark Coles, Peter Watkinson, Naj Rahman, Brian Angus, Alexander J. Mentzer, Alex Novak, Marc Feldman, Alex Richter, Sian Faustini, Camilla Bathurst, Joseph Van de Wiel, Susie Mee, Karen James, Bushra Rahman, Karen Turner, Adam Hill, Anthony Gordon, Christina Yap, Michael Matthay, Danny McAuley, Andrew Hall, Paul Dark, Andrew McMichael, Investigators, CATALYST, National Institute for Health Research, UK Research and Innovation, and NIHR
- Subjects
PNEUMONIA ,Pulmonary and Respiratory Medicine ,Adolescent ,BLOCKADE ,Respiratory System ,Antibodies, Monoclonal, Humanized ,COVID-19/drug therapy ,1117 Public Health and Health Services ,Critical Care Medicine ,General & Internal Medicine ,Humans ,TUMOR-NECROSIS-FACTOR ,Science & Technology ,SARS-CoV-2 ,COVID-19 ,CATALYST investigators ,1103 Clinical Sciences ,Bayes Theorem ,Standard of Care ,Articles ,COLONY-STIMULATING FACTOR ,Infliximab ,COVID-19 Drug Treatment ,Infliximab/therapeutic use ,Treatment Outcome ,Life Sciences & Biomedicine ,1199 Other Medical and Health Sciences - Abstract
Background: Dysregulated inflammation is associated with poor outcomes in COVID-19. We aimed to assess the efficacy of namilumab (a granulocyte-macrophage colony stimulating factor inhibitor) and infliximab (a tumour necrosis factor inhibitor) in hospitalised patients with COVID-19, to prioritise agents for phase 3 trials. Methods: In this randomised, multicentre, multi-arm, multistage, parallel-group, open-label, adaptive, phase 2, proof-of-concept trial (CATALYST), we recruited patients (aged ≥16 years) admitted to hospital with COVID-19 pneumonia and C-reactive protein (CRP) concentrations of 40 mg/L or greater, at nine hospitals in the UK. Participants were randomly assigned with equal probability to usual care or usual care plus a single intravenous dose of namilumab (150 mg) or infliximab (5 mg/kg). Randomisation was stratified by care location within the hospital (ward vs intensive care unit [ICU]). Patients and investigators were not masked to treatment allocation. The primary endpoint was improvement in inflammation, measured by CRP concentration over time, analysed using Bayesian multilevel models. This trial is now complete and is registered with ISRCTN, 40580903. Findings: Between June 15, 2020, and Feb 18, 2021, we screened 299 patients and 146 were enrolled and randomly assigned to usual care (n=54), namilumab (n=57), or infliximab (n=35). For the primary outcome, 45 patients in the usual care group were compared with 52 in the namilumab group, and 29 in the usual care group were compared with 28 in the infliximab group. The probabilities that the interventions were superior to usual care alone in reducing CRP concentration over time were 97% for namilumab and 15% for infliximab; the point estimates for treatment–time interactions were –0·09 (95% CI –0·19 to 0·00) for namilumab and 0·06 (–0·05 to 0·17) for infliximab. 134 adverse events occurred in 30 (55%) of 55 patients in the namilumab group compared with 145 in 29 (54%) of 54 in the usual care group. 102 adverse events occurred in 20 (69%) of 29 patients in the infliximab group compared with 112 in 17 (50%) of 34 in the usual care group. Death occurred in six (11%) patients in the namilumab group compared with ten (19%) in the usual care group, and in four (14%) in the infliximab group compared with five (15%) in the usual care group. Interpretation: Namilumab, but not infliximab, showed proof-of-concept evidence for reduction in inflammation—as measured by CRP concentration—in hospitalised patients with COVID-19 pneumonia. Namilumab should be prioritised for further investigation in COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
29. Vaccine effectiveness against COVID-19 breakthrough infections in patients with cancer (UKCCEP): a population-based test-negative case-control study
- Author
-
Lennard Y W Lee, Thomas Starkey, Maria C Ionescu, Martin Little, Michael Tilby, Arvind R Tripathy, Hayley S Mckenzie, Youssra Al-Hajji, Matthew Barnard, Liza Benny, Alexander Burnett, Emma L Cattell, Jackie Charman, James J Clark, Sam Khan, Qamar Ghafoor, George Illsley, Catherine Harper-Wynne, Rosie J Hattersley, Alvin J X Lee, Pauline C Leonard, Justin K H Liu, Matthew Pang, Jennifer S Pascoe, James R Platt, Vanessa A Potter, Amelia Randle, Anne S Rigg, Tim M Robinson, Tom W Roques, René L Roux, Stefan Rozmanowski, Mark H Tuthill, Isabella Watts, Sarah Williams, Tim Iveson, Siow Ming Lee, Gary Middleton, Mark Middleton, Andrew Protheroe, Matthew W Fittall, Tom Fowler, Peter Johnson, Emma Kinloch, Emily Lam, Gillian Murphy, Malcolm Rhodes, and Kate Robinson
- Subjects
Adult ,COVID-19 Vaccines ,Oncology ,Adolescent ,SARS-CoV-2 ,Case-Control Studies ,Neoplasms ,COVID-19 ,Humans ,Vaccine Efficacy ,Viral Vaccines - Abstract
Background People with cancer are at increased risk of hospitalisation and death following infection with SARS-CoV-2.Therefore, we aimed to conduct one of the first evaluations of vaccine effectiveness against breakthroughSARS-CoV-2 infections in patients with cancer at a population level.Methods In this population-based test-negative case-control study of the UK Coronavirus Cancer Evaluation Project(UKCCEP), we extracted data from the UKCCEP registry on all S ARS-CoV-2 PCR test results (from the SecondGeneration Surveillance System), vaccination records (from the National Immunisation Management Service),patient demographics, and cancer records from England, UK, from Dec 8, 2020, to Oct 15, 2021. Adults (aged≥18 years) with cancer in the UKCCEP registry were identified via Public Health England’s Rapid Cancer RegistrationDataset between Jan 1, 2018, and April 30, 2021, and comprised the cancer cohort. We constructed a control populationcohort from adults with PCR tests in the UKCCEP registry who were not contained within the Rapid CancerRegistration Dataset. The coprimary endpoints were overall vaccine effectiveness ag ainst breakthrough infectionsafter the second dose (positive PCR COVID-19 test) and vacci ne effectiveness against breakthrough infections at3–6 months after the second dose in the cancer cohort and control population.Findings The cancer cohort comprised 377 194 individuals, of whom 42882 had breakthrough SARS-CoV-2 infections.The control population consisted of 28 010 955 in dividuals, of wh om 5 748 708 had SARS- CoV-2 breakthroughinfections. Overall vaccine effectiveness was 69·8% (95% CI 69·8–69·9) in the control population and 65·5%(65·1–65·9) in the cancer cohort. Vaccine effectiveness at3–6 months was lower in the cancer cohort (47·0%, 46·3–47·6)than in the control population (61·4%, 61·4–61·5).Interpretation COVID-19 vaccination is effective for individuals with cancer, conferring varying levels of protectionagainst breakthrough infections. However, vaccine effectiveness is lower in patients with cancer than in the generalpopulation. COVID-19 vaccination for patients with cancer should be used in conjunction with non-pharmacologicalstrategies and community-based antiviral treatment programmes to reduce the risk that COVID-19 poses to patientswith cancer
- Published
- 2022
- Full Text
- View/download PDF
30. The Gut Microbiome, Microsatellite Status and the Response to Immunotherapy in Colorectal Cancer
- Author
-
Toritseju O. Sillo, Andrew D. Beggs, Gary Middleton, and Akinfemi Akingboye
- Subjects
Inorganic Chemistry ,Organic Chemistry ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
There is increasing evidence in a range of cancer types that the microbiome plays a direct role in modulating the anti-cancer immune response both at the gut level and systemically. Differences in the gut microbiota have been shown to correlate with differences in immunotherapy responses in a range of non-gastrointestinal tract cancers. DNA mismatch repair-deficient (dMMR) colorectal cancer (CRC) is radically different to DNA mismatch repair-proficient (pMMR) CRC in clinical phenotype and in its very good responses to immunotherapy. While this has usually been thought to be due to the high mutational burden in dMMR CRC, the gut microbiome is radically different in dMMR and pMMR CRC in terms of both composition and diversity. It is probable that differences in the gut microbiota contribute to the varied responses to immunotherapy in dMMR versus pMMR CRC. Targeting the microbiome offers a way to boost the response and increase the selection of patients who might benefit from this therapy. This paper reviews the available literature on the role of the microbiome in the response to immunotherapy in dMMR and pMMR CRC, explores the potential causal relationship and discusses future directions for study in this exciting and rapidly changing field.
- Published
- 2023
- Full Text
- View/download PDF
31. Association of SARS-CoV-2 Spike Protein Antibody Vaccine Response With Infection Severity in Patients With Cancer
- Author
-
Lennard Y W, Lee, Michael, Tilby, Thomas, Starkey, Maria C, Ionescu, Alex, Burnett, Rosie, Hattersley, Sam, Khan, Martin, Little, Justin K H, Liu, James R, Platt, Arvind, Tripathy, Isabella, Watts, Sophie Therese, Williams, Nathan, Appanna, Youssra, Al-Hajji, Matthew, Barnard, Liza, Benny, Andrew, Buckley, Emma, Cattell, Vinton, Cheng, James, Clark, Leonie, Eastlake, Kate, Gerrand, Qamar, Ghafoor, Simon, Grumett, Catherine, Harper-Wynne, Rachel, Kahn, Alvin J X, Lee, Anna, Lydon, Hayley, McKenzie, Hari, Panneerselvam, Jennifer, Pascoe, Grisma, Patel, Vijay, Patel, Vanessa, Potter, Amelia, Randle, Anne S, Rigg, Tim, Robinson, Rebecca, Roylance, Tom, Roques, Stefan, Rozmanowski, René L, Roux, Ketan, Shah, Martin, Sintler, Harriet, Taylor, Tania, Tillett, Mark, Tuthill, Sarah, Williams, Andrew, Beggs, Tim, Iveson, Siow Ming, Lee, Gary, Middleton, Mark, Middleton, Andrew S, Protheroe, Matthew W, Fittall, Tom, Fowler, Peter, Johnson, and Remarez, Sheehan
- Subjects
Cancer Research ,Oncology - Abstract
ImportanceAccurate identification of patient groups with the lowest level of protection following COVID-19 vaccination is important to better target resources and interventions for the most vulnerable populations. It is not known whether SARS-CoV-2 antibody testing has clinical utility for high-risk groups, such as people with cancer.ObjectiveTo evaluate whether spike protein antibody vaccine response (COV-S) following COVID-19 vaccination is associated with the risk of SARS-CoV-2 breakthrough infection or hospitalization among patients with cancer.Design, Setting, and ParticipantsThis was a population-based cross-sectional study of patients with cancer from the UK as part of the National COVID Cancer Antibody Survey. Adults with a known or reported cancer diagnosis who had completed their primary SARS-CoV-2 vaccination schedule were included. This analysis ran from September 1, 2021, to March 4, 2022, a period covering the expansion of the UK’s third-dose vaccination booster program.InterventionsAnti–SARS-CoV-2 COV-S antibody test (Elecsys; Roche).Main Outcomes and MeasuresOdds of SARS-CoV-2 breakthrough infection and COVID-19 hospitalization.ResultsThe evaluation comprised 4249 antibody test results from 3555 patients with cancer and 294 230 test results from 225 272 individuals in the noncancer population. The overall cohort of 228 827 individuals (patients with cancer and the noncancer population) comprised 298 479 antibody tests. The median age of the cohort was in the age band of 40 and 49 years and included 182 741 test results (61.22%) from women and 115 737 (38.78%) from men. There were 279 721 tests (93.72%) taken by individuals identifying as White or White British. Patients with cancer were more likely to have undetectable anti-S antibody responses than the general population (199 of 4249 test results [4.68%] vs 376 of 294 230 [0.13%]; P P P Conclusions and RelevanceThe findings of this cross-sectional study suggest that COV-S antibody testing allows the identification of patients with cancer who have the lowest level of antibody-derived protection from COVID-19. This study supports larger evaluations of SARS-CoV-2 antibody testing. Prevention of SARS-CoV-2 transmission to patients with cancer should be prioritized to minimize impact on cancer treatments and maximize quality of life for individuals with cancer during the ongoing pandemic.
- Published
- 2023
- Full Text
- View/download PDF
32. Key findings from the UKCCMP cohort of 877 patients with haematological malignancy and COVID-19: disease control as an important factor relative to recent chemotherapy or anti-CD20 therapy
- Author
-
David Wrench, Gary Middleton, Gordon Cook, Abigail Gault, Jonathan Carmichael, John Ashcroft, Rachel Kerr, Helen Curley, Austin G. Kulasekararaj, Neil Rabin, Helen S. Oram, Jack Illingworth, Roland Arnold, Ruth Pettengell, Tom Newsom-Davies, Jane Apperly, Michael Tilby, Graham P. Collins, Oliver Tomkins, Karin Purshouse, Stephen Booth, Mohammed Altohami, Jean-Baptiste Cazier, Lucy Cook, Andrew J. Innes, Naomi A. Campton, James Aries, Sam Moody, Tania Tillet, Lennard Y. W. Lee, Sajjan Mittal, Claire Palles, Saoirse Dolly, Leena Mukherjee, and Csilla Várnai
- Subjects
Adult ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Context (language use) ,Antineoplastic Agents, Immunological ,COVID‐19 ,Risk Factors ,Internal medicine ,medicine ,haematological malignancies ,Humans ,Prospective Studies ,1102 Cardiorespiratory Medicine and Haematology ,Chemotherapy ,Science & Technology ,Leukemia ,business.industry ,MORTALITY ,UKCCMP team ,Cancer ,COVID-19 ,Odds ratio ,Hematology ,medicine.disease ,Antigens, CD20 ,Research Papers ,CANCER ,Confidence interval ,SEVERITY ,Hematologic Neoplasms ,Cohort ,Observational study ,cancer treatments ,Female ,business ,Multiple Myeloma ,Life Sciences & Biomedicine ,Progressive disease ,Research Paper - Abstract
Summary Patients with haematological malignancies have a high risk of severe infection and death from SARS‐CoV‐2. In this prospective observational study, we investigated the impact of cancer type, disease activity, and treatment in 877 unvaccinated UK patients with SARS‐CoV‐2 infection and active haematological cancer. The primary end‐point was all‐cause mortality. In a multivariate analysis adjusted for age, sex and comorbidities, the highest mortality was in patients with acute leukaemia [odds ratio (OR) = 1·73, 95% confidence interval (CI) 1·1–2·72, P = 0·017] and myeloma (OR 1·3, 95% CI 0·96–1·76, P = 0·08). Having uncontrolled cancer (newly diagnosed awaiting treatment as well as relapsed or progressive disease) was associated with increased mortality risk (OR = 2·45, 95% CI 1·09–5·5, P = 0·03), as was receiving second or beyond line of treatment (OR = 1·7, 95% CI 1·08–2·67, P = 0·023). We found no association between recent cytotoxic chemotherapy or anti‐CD19/anti‐CD20 treatment and increased risk of death within the limitations of the cohort size. Therefore, disease control is an important factor predicting mortality in the context of SARS‐CoV‐2 infection alongside the possible risks of therapies such as cytotoxic treatment or anti‐CD19/anti‐CD20 treatments.
- Published
- 2021
33. A state-of-the-art review of stratified medicine in cancer: towards a future precision medicine strategy in cancer
- Author
-
Fabrice Andre, Charles Swanton, H.L. Robbins, and Gary Middleton
- Subjects
Response rate (survey) ,medicine.medical_specialty ,Clinical Trials, Phase I as Topic ,Genotype ,Mechanism (biology) ,business.industry ,medicine.medical_treatment ,Cancer ,Context (language use) ,Hematology ,Genomics ,medicine.disease ,Precision medicine ,Minimal residual disease ,Targeted therapy ,Oncology ,Neoplasms ,medicine ,Biomarker (medicine) ,Humans ,Precision Medicine ,business ,Intensive care medicine - Abstract
Background Building on the success of targeted therapy in certain well-defined cancer genotypes, three platform studies – NCI-MATCH, LUNG-MAP and The National Lung Matrix Trial (NLMT) have attempted to discover new genotype-matched therapies for people with cancer. Patients and methods We review the outputs from these platform studies. This review led us to propose a series of recommendations and considerations that we hope will inform future precision medicine programmes in cancer Results The three studies collectively screened over 13,000 patients. Across 37 genotype matched cohorts there have been 66/875 responders, an overall response rate of 7.5%. Targeting copy number gain yielded 5/199 responses across 9 biomarker:drug matched cohorts, a response rate of 2.5%. Conclusions The majority of these studies used single-agent targeted therapies. Whilst pre-clinical data can suggest rational combination treatment to reverse adaptive resistance or block parallel activated pathways there is an essential need for accurate modelling of the toxicity:activity trade-off of combinations. Agent selection is often sub-optimal; dose expansion should only be performed with agents with clear clinical proof of mechanism and the high target selectivity. Targeting copy number change has been disappointing; it is crucial to define the drivers on shared amplicons that include the targeted aberration. Maximising outcomes with currently available targeted therapies requires moving towards a more contextualised stratified medicine acknowledging the criticality of the genomic, transcriptional and immunological context on which the targeted aberration is inscribed. Genomic complexity and instability is likely to be a leading cause of targeted therapy failure in genomically complex cancers. Pre-clinical models must be developed that more accurately capture the genomic complexity of human disease. The degree of attrition of studies performed after standard of care therapy suggest that serious efforts be made to develop a suite of precision medicine studies in the minimal residual disease setting.
- Published
- 2021
34. Tetraspanin 6 is a regulator of carcinogenesis in colorectal cancer
- Author
-
Gary Middleton, Matthew Pugh, Andrew D Beggs, Maha Ibrahim, Sundaresan Rajesh, Balázs Győrffy, Chris Tselepis, Regina Andrijes, Rahul K Hejmadi, Michael Overduin, Vera Novitskaya, and Fedor Berditchevski
- Subjects
TGF alpha ,Medical Sciences ,Tetraspanins ,Colorectal cancer ,EGFR ,Cetuximab ,colorectal cancer ,Apoptosis ,Biology ,medicine.disease_cause ,Mice ,Antineoplastic Agents, Immunological ,Tetraspanin ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,Cell Proliferation ,Mice, Knockout ,Multidisciplinary ,Predictive marker ,Biological Sciences ,Prognosis ,medicine.disease ,APCmin ,digestive system diseases ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Survival Rate ,tetraspanin ,Cancer research ,biology.protein ,KRAS ,Colorectal Neoplasms ,Carcinogenesis ,medicine.drug - Abstract
Significance Tetraspanin protein (Tspan6) is a member of the tetraspanin family. Using a combination of in vitro and in vivo assays, we demonstrate that Tspan6 functions as a tumor suppressor in colorectal cancer (CRC) by attenuating the epidermal growth factor receptor (EGFR)–based signaling axis. Tspan6 forms a tripartite complex with transmembrane form of TGF-α and an adaptor protein syntenin-1 and negatively regulates secretion of TGF-α. The expression of Tspan6 is frequently decreased in CRC, and this correlates with poor survival. Importantly, the expression of Tspan6 in CRC correlated independently of tumor molecular profile with better patient responses to Cetuximab, an EGFR-targeted therapy. These results identify Tspan6 as a regulator of CRC development and a potential predictive biomarker for EGFR-targeted therapies., Early stages of colorectal cancer (CRC) development are characterized by a complex rewiring of transcriptional networks resulting in changes in the expression of multiple genes. Here, we demonstrate that the deletion of a poorly studied tetraspanin protein Tspan6 in Apcmin/+ mice, a well-established model for premalignant CRC, resulted in increased incidence of adenoma formation and tumor size. We demonstrate that the effect of Tspan6 deletion results in the activation of EGF-dependent signaling pathways through increased production of the transmembrane form of TGF-α (tmTGF-α) associated with extracellular vesicles. This pathway is modulated by an adaptor protein syntenin-1, which physically links Tspan6 and tmTGF-α. In support of this, the expression of Tspan6 is frequently decreased or lost in CRC, and this correlates with poor survival. Furthermore, the analysis of samples from the epidermal growth factor receptor (EGFR)–targeting clinical trial (COIN trial) has shown that the expression of Tspan6 in CRC correlated with better patient responses to EGFR-targeted therapy involving Cetuximab. Importantly, Tspan6-positive patients with tumors in the proximal colon (right-sided) and those with KRAS mutations had a better response to Cetuximab than the patients that expressed low Tspan6 levels. These results identify Tspan6 as a regulator of CRC development and a potential predictive marker for EGFR-targeted therapies in CRC beyond RAS pathway mutations.
- Published
- 2021
- Full Text
- View/download PDF
35. Development and Validation of a Combined Hypoxia and Immune Prognostic Classifier for Head and Neck Cancer
- Author
-
Neeraj Lal, Riyue Bao, Sandra Ventorin Von Zeidler, Arun Khattri, Helen R Valentine, Albert N. Menezes, Jean-Baptiste Cazier, Hisham Mehanna, Tanguy Y. Seiwert, Maha Ibrahim, Steven P. Lee, Jonathan J Deeks, Lucinda Archer, Daniel A. Tennant, Andrew D Beggs, Christopher Bagnall, Margaret Hartley, Nikolaos Batis, Kalu U.E. Ogbureke, Baksho Kaul, Jill Brooks, Gary Middleton, Jennifer L. Bryant, Catharine M L West, Gordon B. Ryan, Rachel Spruce, and Benjamin E. Willcox
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,In silico ,B7-H1 Antigen ,Targeted therapy ,Cohort Studies ,Young Adult ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Internal medicine ,Biomarkers, Tumor ,Humans ,Medicine ,Multiplex ,Hypoxia ,Survival rate ,Aged ,Retrospective Studies ,Aged, 80 and over ,Regulation of gene expression ,business.industry ,Gene Expression Profiling ,Head and neck cancer ,Middle Aged ,Gene signature ,Prognosis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Survival Rate ,030104 developmental biology ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Female ,business - Abstract
Purpose: Intratumoral hypoxia and immunity have been correlated with patient outcome in various tumor settings. However, these factors are not currently considered for treatment selection in head and neck cancer (HNC) due to lack of validated biomarkers. Here we sought to develop a hypoxia-immune classifier with potential application in patient prognostication and prediction of response to targeted therapy. Experimental Design: A 54-gene hypoxia-immune signature was constructed on the basis of literature review. Gene expression was analyzed in silico using the The Cancer Genome Atlas (TCGA) HNC dataset (n = 275) and validated using two independent cohorts (n = 130 and 123). IHC was used to investigate the utility of a simplified protein signature. The spatial distribution of hypoxia and immune markers was examined using multiplex immunofluorescence staining. Results: Unsupervised hierarchical clustering of TCGA dataset (development cohort) identified three patient subgroups with distinct hypoxia-immune phenotypes and survival profiles: hypoxialow/immunehigh, hypoxiahigh/immunelow, and mixed, with 5-year overall survival (OS) rates of 71%, 51%, and 49%, respectively (P = 0.0015). The prognostic relevance of the hypoxia-immune gene signature was replicated in two independent validation cohorts. Only PD-L1 and intratumoral CD3 protein expression were associated with improved OS on multivariate analysis. Hypoxialow/immunehigh and hypoxiahigh/immunelow tumors were overrepresented in “inflamed” and “immune-desert” microenvironmental profiles, respectively. Multiplex staining demonstrated an inverse correlation between CA-IX expression and prevalence of intratumoral CD3+ T cells (r = −0.5464; P = 0.0377), further corroborating the transcription-based classification. Conclusions: We developed and validated a hypoxia-immune prognostic transcriptional classifier, which may have clinical application to guide the use of hypoxia modification and targeted immunotherapies for the treatment of HNC.
- Published
- 2019
- Full Text
- View/download PDF
36. MDSC targeting with Gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancersResearch in Context
- Author
-
Livingstone Fultang, Silvia Panetti, Margaret Ng, Paul Collins, Suzanne Graef, Nagy Rizkalla, Sarah Booth, Richard Lenton, Boris Noyvert, Claire Shannon-Lowe, Gary Middleton, Francis Mussai, and Carmela De Santo
- Subjects
lcsh:R5-920 ,lcsh:R ,lcsh:Medicine ,lcsh:Medicine (General) - Abstract
Background: Targeting of MDSCs is a major clinical challenge in the era of immunotherapy. Antibodies which deplete MDSCs in murine models can reactivate T cell responses. In humans such approaches have not developed due to difficulties in identifying targets amenable to clinical translation. Methods: RNA-sequencing of M-MDSCs and G-MDSCs from cancer patients was undertaken. Flow cytometry and immunohistochemistry of blood and tumours determined MDSC CD33 expression. MDSCs were treated with Gemtuzumab ozogamicin and internalisation kinetics, and cell death mechanisms determined by flow cytometry, confocal microscopy and electron microscopy. Effects on T cell proliferation and CAR-T cell anti-tumour cytotoxicity were identified in the presence of Gemtuzumab ozogamicin. Findings: RNA-sequencing of human M-MDSCs and G-MDSCs identified transcriptomic differences, but that CD33 is a common surface marker. Flow cytometry indicated CD33 expression is higher on M-MDSCs, and CD33+ MDSCs are found in the blood and tumours regardless of cancer subtype. Treatment of human MDSCs leads to Gemtuzumab ozogamicin internalisation, increased p-ATM, and cell death; restoring T cell proliferation. Anti-GD2-/mesothelin-/EGFRvIII-CAR-T cell activity is enhanced in combination with the anti-MDSC effects of Gemtuzumab ozogamicin. Interpretation: The study identifies that M-MDSCs and G-MDSCs are transcriptomically different but CD33 is a therapeutic target on peripheral and infiltrating MDSCs across cancer subtypes. The immunotoxin Gemtuzumab ozogamicin can deplete MDSCs providing a translational approach to reactivate T cell and CAR-T cell responses against multiple cancers. In the rare conditions of HLH/MAS gemtuzumab ozogamicin provides a novel anti-myeloid strategy. Fund: This work was supported by Cancer Research UK, CCLG, Treating Children with Cancer, and the alumni and donors to the University of Birmingham. Keywords: MDSC, Gemtuzumab, CD33, Cancer, CAR-T
- Published
- 2019
37. MDSC targeting with Gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancers
- Author
-
Gary Middleton, Nagy Rizkalla, Richard Lenton, Margaret H.L. Ng, Carmela De Santo, Livingstone Fultang, Boris Noyvert, Suzanne Graef, Claire Shannon-Lowe, Sarah Booth, Francis Mussai, Paul Collins, and Silvia Panetti
- Subjects
0301 basic medicine ,Research paper ,Gemtuzumab ozogamicin ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,MDSC ,Cell ,CD33 ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Immunophenotyping ,Flow cytometry ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Immunotoxin ,Neoplasms ,Medicine and Health Sciences ,medicine ,Humans ,Cancer ,medicine.diagnostic_test ,business.industry ,Gene Expression Profiling ,Myeloid-Derived Suppressor Cells ,Biology and Life Sciences ,General Medicine ,Immunotherapy ,medicine.disease ,Immunohistochemistry ,Gemtuzumab ,CAR-T ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Transcriptome ,business ,Biomarkers ,medicine.drug - Abstract
Background: Targeting of MDSCs is a major clinical challenge in the era of immunotherapy. Antibodies which deplete MDSCs in murine models can reactivate T cell responses. In humans such approaches have not developed due to difficulties in identifying targets amenable to clinical translation. Methods: RNA-sequencing of M-MDSCs and G-MDSCs from cancer patients was undertaken. Flow cytometry and immunohistochemistry of blood and tumours determined MDSC CD33 expression. MDSCs were treated with Gemtuzumab ozogamicin and internalisation kinetics, and cell death mechanisms determined by flow cytometry, confocal microscopy and electron microscopy. Effects on T cell proliferation and CAR-T cell anti-tumour cytotoxicity were identified in the presence of Gemtuzumab ozogamicin. Findings: RNA-sequencing of human M-MDSCs and G-MDSCs identified transcriptomic differences, but that CD33 is a common surface marker. Flow cytometry indicated CD33 expression is higher on M-MDSCs, and CD33+ MDSCs are found in the blood and tumours regardless of cancer subtype. Treatment of human MDSCs leads to Gemtuzumab ozogamicin internalisation, increased p-ATM, and cell death; restoring T cell proliferation. Anti-GD2-/mesothelin-/EGFRvIII-CAR-T cell activity is enhanced in combination with the anti-MDSC effects of Gemtuzumab ozogamicin. Interpretation: The study identifies that M-MDSCs and G-MDSCs are transcriptomically different but CD33 is a therapeutic target on peripheral and infiltrating MDSCs across cancer subtypes. The immunotoxin Gemtuzumab ozogamicin can deplete MDSCs providing a translational approach to reactivate T cell and CAR-T cell responses against multiple cancers. In the rare conditions of HLH/MAS gemtuzumab ozogamicin provides a novel anti-myeloid strategy. Fund: This work was supported by Cancer Research UK, CCLG, Treating Children with Cancer, and the alumni and donors to the University of Birmingham. (c) 2019 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
- Published
- 2019
- Full Text
- View/download PDF
38. TGF-β orchestrates the phenotype and function of monocytic myeloid-derived suppressor cells in colorectal cancer
- Author
-
Gary Middleton, Francis Mussai, Carmela De Santo, Luciana Gneo, Rahul K. Hejmadi, and Nagy Rizkalla
- Subjects
Cancer Research ,medicine.medical_treatment ,T cell ,CD14 ,Immunology ,Monocytes ,Transforming Growth Factor beta ,medicine ,Tumor Microenvironment ,Immunology and Allergy ,Humans ,biology ,Chemistry ,Monocyte ,Myeloid-Derived Suppressor Cells ,Nitric oxide synthase 2 ,HLA-DR Antigens ,Interleukin-10 ,Interleukin 10 ,Cytokine ,medicine.anatomical_structure ,Phenotype ,Oncology ,biology.protein ,Myeloid-derived Suppressor Cell ,Cancer research ,Immunohistochemistry ,Colorectal Neoplasms - Abstract
BackgroundMonocytic myeloid-derived suppressor cells (M-MDSCs) are significantly expanded in the blood of colorectal cancer (CRC) patients. However, their presence and underlying mechanisms in the tumour microenvironment of CRC have not been examined in detail.MethodsTumour tissues and peripheral blood from CRC patients were analysed for the presence of M-MDSCs. The mechanisms of suppression were analysed by blocking pathways by which MDSCs abrogate T cell proliferation. Co-culture of CRC cells with monocytes were performed with and without cytokine blocking antibodies to determine the mechanism by which CRC cells polarise monocytes. Multi-spectral IHC was used to demonstrate the intra-tumoral location of M-MDSCs.ResultsTumour tissues and blood of CRC patients contain M-MDSCs which inhibit T cell proliferation. Whilst inhibition of arginase and nitric oxide synthase 2 fail to rescue T cell proliferation, blockade of IL-10 released by these HLA-DR−cells abrogates the suppresivity of M-MDSCs. Tumour conditioned media (TCM) significantly reduces HLA-DR expression, increases IL-10 release from monocytes and causes them to become suppressive. TGF-β is highly expressed in the TCM and accumulates in the plasma. TGF-β reduces HLA-DR expression and drives monocyte immunosuppressivity. The invasive margin of CRC is enriched in CD14+HLA-DR−cells in close proximity to T cells.ConclusionsOur study demonstrates the cross-talk between CRC cells, M-MDSCs and T cells. Characterisation of CRC M-MDSCs point to therapeutic avenues to target these cells in addition to TGF-β blockade.
- Published
- 2021
39. Towards personalized treatment of smoking-related lung cancers
- Author
-
Gary Middleton
- Subjects
Oncology ,medicine.medical_specialty ,Multidisciplinary ,Lung ,business.industry ,Personalized treatment ,Cancer ,respiratory system ,medicine.disease ,respiratory tract diseases ,Clinical trial ,medicine.anatomical_structure ,Internal medicine ,Rare mutations ,medicine ,Lung tumours ,business - Abstract
An innovative approach to a phase II clinical trial aims to test the effects of multiple targeted treatments simultaneously in participants in the UK National Lung Matrix Trial. The design makes it possible to study small numbers of people with rare mutations in non-small-cell lung tumours. An adaptive trial design to test treatments for rare lung cancers.
- Published
- 2021
- Full Text
- View/download PDF
40. Determinants of anti-PD-1 response and resistance in clear cell renal cell carcinoma
- Author
-
Sanjay Popat, Lewis Au, Jan Attig, Catherine Horsfield, Hayley Bridger, Kitty Chan, Haixi Yan, David Moore, Lara-Rose Iredale, Salma Kadiri, Sebastian Brandner, Rebecca C. Fitzgerald, Bruce Tanchel, Maise Al-Bakir, Katey S. S. Enfield, Merche Jimenez-Linan, Andrew P. Robinson, Kim Edmonds, Stuart Horswell, Elena Provenzano, Andrew V. Biankin, Benny Chain, Scott Shepherd, Antonia Toncheva, Carlos Caldas, Gerald Langman, Fabio Gomes, I Puccio, Amy Kerr, Sharmistha Ghosh, Caroline Dive, James Larkin, Siow Ming Lee, Nicholas McGranahan, Peter Ellery, Charlotte Spencer, Dionysis Papadatos-Pastos, Charles Swanton, Maryam Razaq, Richard J. Gilbertson, Rachael Thompson, William Drake, Lyra Del Rosario, Debra Enting, Lisa Pickering, Crispin T. Hiley, David A Moore, Christian H. Ottensmeier, Ehsan Ghorani, Simon Chowdhury, Simon Tavaré, Sophie Ward, Gordon Stamp, Peter J. Parker, Sam M. Janes, Giorgia Trevisan, Mary Falzon, Ultan McDermott, Christopher Abbosh, Fiona Byrne, Kroopa Joshi, Kim Dhillon, George Kassiotis, James L. Reading, Heather Shaw, Tariq Enver, Dean A. Fennell, Jonathan Ledermann, Annika Fendler, Emma Beddowes, Peter Cockcroft, Mary Mangwende, Desiree Schnidrig, Ian Tomlinson, Mark Linch, Ben Challacombe, Vasiliki Michalarea, Yvonne Summers, Fiona H Blackhall, Robert Mason, Emma Nye, Robert E. Hynds, Debra H. Josephs, Mariana Werner Sunderland, Adrian Tookman, Emilia L. Lim, Paddy Stone, Cristina Naceur-Lombardelli, Bernard Olisemeke, Teresa Marafioti, Mat Carter, Grant D. Stewart, Sanjay Jogai, Richard Marais, Imran Uddin, Kevin Litchfield, Daniel Hochhauser, Alexander Polson, William Yang, Hang Xu, Peter Hill, Jonathon Olsburgh, Gordon Beattie, Justine Korteweg, Nnenna Kanu, Martin Forster, Andrew Tutt, Ben Shum, Elias Pintus, Alison Cluroe, Matt Krebs, Patricia Roxburgh, Caroline Stirling, Selvaraju Veeriah, Olivia Curtis, Marc Robert de Massy, Emine Hatipoglu, Tom Lund, Kai-Keen Shiu, Tina Mackay, Pablo D. Becker, Faye Gishen, Massimo Loda, Aida Murra, Karin A. Oien, Joanne Webb, Jose Lopez, Sarah Sarker, Adrienne M. Flanagan, Ula Mahadeva, Ian Proctor, Ruby Stewart, John Le Quesne, Elaine Borg, Archana Fernando, Babu Naidu, Andrew Rowan, Abby Sharp, Mairead McKenzie, Ayse Akarca, Anthony J. Chalmers, James Spicer, Gary Middleton, Hollie Bancroft, Jo Dransfield, Nicos Fotiadis, Charlotte Ferris, Ron Sinclair, Mary Varia, Peter Van Loo, Lavinia Spain, Lena Karapagniotou, Nikki Hunter, Roberto Salgado, Sarah Vaughan, Chi-wah Lok, Karen Harrison-Phipps, Hema Verma, Jacqui Shaw, Rodelaine Wilson, Zoe Rhodes, Anna Green, Reena Khiroya, Miriam Mitchison, Ashish Chandra, Colin Watts, Peter Colloby, Uzma Asghar, Laura Farrelly, Tim O'Brien, Stephan Beck, Steve Hazell, Tanya Ahmad, Martin Collard, John Bridgewater, James D. Brenton, Sarah Rudman, Eleanor Carlyle, Andrew C. Kidd, Lizi Manzano, Sergio A. Quezada, Sioban Fraser, Allan Hackshaw, Nadia Yousaf, Samra Turajlic, Henning Walczak, David Nicol, Mariam Jamal-Hanjani, Sarah Howlett, Andrew Furness, Simranpreet Summan, Kevin G. Blyth, S. Baijal, Gert Attard, Marcos Duran Vasquez, Mita Afroza Akther, Karla Lingard, Ben Deakin, Ariana Huebner, and David G. Harrison
- Subjects
Cancer Research ,Receptors, Antigen, T-Cell ,Biology ,CD8-Positive T-Lymphocytes ,Clinical Trials, Phase II as Topic ,Antigen ,Immunity ,Exome Sequencing ,medicine ,Tumor Microenvironment ,Humans ,Prospective Studies ,Spotlight ,Mode of action ,Receptor ,Carcinoma, Renal Cell ,Immune Checkpoint Inhibitors ,Sequence Analysis, RNA ,Gene Expression Profiling ,T-cell receptor ,Endogenous Retroviruses ,Genomics ,medicine.disease ,Kidney Neoplasms ,Clear cell renal cell carcinoma ,Nivolumab ,Oncology ,Drug Resistance, Neoplasm ,Cancer research ,Tumor Escape ,Single-Cell Analysis ,CD8 - Abstract
ADAPTeR is a prospective, phase II study of nivolumab (anti-PD-1) in 15 treatment-naive patients (115 multiregion tumor samples) with metastatic clear cell renal cell carcinoma (ccRCC) aiming to understand the mechanism underpinning therapeutic response. Genomic analyses show no correlation between tumor molecular features and response, whereas ccRCC-specific human endogenous retrovirus expression indirectly correlates with clinical response. T cell receptor (TCR) analysis reveals a significantly higher number of expanded TCR clones pre-treatment in responders suggesting pre-existing immunity. Maintenance of highly similar clusters of TCRs post-treatment predict response, suggesting ongoing antigen engagement and survival of families of T cells likely recognizing the same antigens. In responders, nivolumab-bound CD8+ T cells are expanded and express GZMK/B. Our data suggest nivolumab drives both maintenance and replacement of previously expanded T cell clones, but only maintenance correlates with response. We hypothesize that maintenance and boosting of a pre-existing response is a key element of anti-PD-1 mode of action.
- Published
- 2021
- Full Text
- View/download PDF
41. Beyond Ipilimumab: a review of immunotherapeutic approaches in clinical trials in melanoma
- Author
-
Gary Middleton
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Cell cycle checkpoint ,business.industry ,Melanoma ,Ipilimumab ,General Medicine ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
Summary In this first in a series of ‘Trials Watch’ articles, we briefly review a highly selected set of clinical trials that are currently recruiting or about to open to recruitment in melanoma, the disease first transformed by the introduction of immune checkpoint blockade inhibitors (ICI). We place equal emphasis on phase I/II studies investigating the activity of biologically compelling novel immunotherapeutics, and on randomised trials of ICI with and without novel agents, as these latter studies optimise the standard-of-care use of ICI, and determine whether novel agents become part of the approved therapeutic armamentarium. We do not consider here combination therapy with other checkpoint antagonists or agonists besides combination of anti-PD-1/PD-L1 monoclonal antibodies (mAbs) with anti-CTLA4 mAbs, as these will be reviewed in a subsequent article in this series. A glossary of agents to be discussed is found at the end of this article.
- Published
- 2020
- Full Text
- View/download PDF
42. NK cells in pancreatic cancer demonstrate impaired cytotoxicity and a regulatory IL-10 phenotype
- Author
-
Brinder S. Mahon, Rachel M. Brown, Sandra Margielewska-Davies, F. Marcon, Keith J. Roberts, Hayden Pearce, Gary Middleton, Samantha Nicol, Jianmin Zuo, Mustafa Farhat, and Paul Moss
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,NK cells ,immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Pancreatic cancer ,Immunology and Allergy ,Medicine ,Cytotoxic T cell ,Humans ,RC254-282 ,cancer Immunology ,Cancer immunology ,Original Research ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,PDAC ,Immunotherapy ,RC581-607 ,medicine.disease ,NKG2D ,Interleukin-10 ,Killer Cells, Natural ,Pancreatic Neoplasms ,Interleukin 10 ,030104 developmental biology ,Cytokine ,Phenotype ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunologic diseases. Allergy ,business ,Research Article ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most common tumor subtypes and remains associated with very poor survival. T cell infiltration into tumor tissue is associated with improved clinical outcome but little is known regarding the potential role of NK cells in disease control. Here we analyze the phenotype and function of NK cells in the blood and tumor tissue from patients with PDAC. Peripheral NK cells are present in normal numbers but display a CD16hiCD57hi phenotype with marked downregulation of NKG2D. Importantly, these cells demonstrate reduced cytotoxic activity and low levels of IFN-γ expression but instead produce high levels of intracellular IL-10, an immunoregulatory cytokine found at increased levels in the blood of PDAC patients. In contrast, NK cells are largely excluded from tumor tissue where they display strong downregulation of both CD16 and CD57, a phenotype that was recapitulated in primary NK cells following co-culture with PDAC organoids. Moreover, expression of activatory proteins, including DNAM-1 and NKP30, was markedly suppressed and the DNAM-1 ligand PVR was strongly expressed on tumor cells. As such, in situ and peripheral NK cells display differential features in patients with PDAC and indicate local and systemic mechanisms by which the tumor can evade immune control. These findings offer a number of potential options for NK-based immunotherapy in the management of patients with PDAC.
- Published
- 2020
43. Extended RAS Analysis of the Phase III EPIC Trial: Irinotecan + Cetuximab Versus Irinotecan as Second-Line Treatment for Patients with Metastatic Colorectal Cancer
- Author
-
Howard A. Burris, Gary Middleton, Alberto Sobrero, Cathy Eng, Wen-Feng Chen, Werner Scheithauer, Heinz-Josef Lenz, Johannes Nippgen, and Regina Esser
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,medicine.medical_treatment ,Population ,Cetuximab ,Irinotecan ,Disease-Free Survival ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Quality of life ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Gastrointestinal Cancer ,medicine ,Humans ,030212 general & internal medicine ,education ,Adverse effect ,neoplasms ,Retrospective Studies ,education.field_of_study ,Chemotherapy ,business.industry ,Metastatic colorectal cancer ,medicine.disease ,digestive system diseases ,Oxaliplatin ,030220 oncology & carcinogenesis ,Quality of Life ,Camptothecin ,Fluorouracil ,business ,Colorectal Neoplasms ,EPIC ,medicine.drug ,RAS - Abstract
Background The multicenter, open‐label, randomized, phase III EPIC study (EMR 062202‐025) investigated cetuximab plus irinotecan versus irinotecan in patients with epidermal growth factor receptor–detectable metastatic colorectal cancer (mCRC) that progressed on first‐line fluoropyrimidine‐ and oxaliplatin‐based chemotherapy; we report the outcomes of patients with RAS‐wild‐type (wt) disease. Materials and Methods Available DNA samples from RAS‐unselected patients (n = 1,164 of 1,298 [89.7%]) were reanalyzed for RAS mutations using beads, emulsion, amplification, and magnetics. Baseline characteristics, efficacy, safety, and poststudy therapy were assessed. RAS‐wt status was defined as a mutated RAS allele frequency of ≤5%, with all relevant alleles being analyzable. Results Baseline characteristics were comparable between the groups (n = 452 patients with RAS‐wt mCRC; cetuximab plus irinotecan n = 231, irinotecan n = 221) and between the RAS‐wt and RAS‐unselected populations. In the cetuximab plus irinotecan versus irinotecan arms, median overall survival was 12.3 versus 12.0 months, median progression‐free survival (PFS) was 5.4 versus 2.6 months, and objective response rate (ORR) was 29.4% versus 5.0%, respectively. Quality of life (QoL) was improved in the cetuximab plus irinotecan arm. Serious adverse events occurred in 45.4% (cetuximab plus irinotecan) and 42.4% (irinotecan) of patients. In total, 47.1% of patients in the irinotecan arm received subsequent cetuximab therapy. Conclusion PFS, ORR, and QoL were improved with cetuximab plus irinotecan as a second‐line treatment in patients with RAS‐wt mCRC, confirming that cetuximab‐based therapy is suitable in this population. Almost half of patients in the irinotecan arm received poststudy cetuximab, masking a potential overall survival benefit of cetuximab addition. Implications for Practice Cetuximab is approved for the treatment of RAS–wild‐type metastatic colorectal cancer (mCRC). In this retrospective analysis of the phase III EPIC study (cetuximab plus irinotecan vs. irinotecan alone as second‐line treatment in patients with RAS‐unselected mCRC), the subgroup of patients with RAS–wild‐type mCRC who received cetuximab plus irinotecan had improved progression‐free survival, objective response rate, and quality of life compared with the RAS‐unselected population. These findings suggest that cetuximab‐based therapy is a suitable second‐line treatment for patients with RAS–wild‐type mCRC., This article analyzes outcomes of patients with RAS‐wild‐type metastatic colorectal cancer who received cetuximab plus irinotecan versus irinotecan alone in the EPIC trial.
- Published
- 2020
44. Immune checkpoint blockade: releasing the breaks or a protective barrier to COVID-19 severe acute respiratory syndrome?
- Author
-
Alvin J.X. Lee, Thomas Starkey, Vinton Cheng, Gary Middleton, D.J. Hughes, Lennard Y W Lee, Karin Purshouse, Luke Freeman-Mills, Oliver J Pickles, and Anna Olsson-Brown
- Subjects
ARDS ,Cancer Research ,medicine.medical_treatment ,Clinical Decision-Making ,Pneumonia, Viral ,Cancer immunotherapy ,B7-H1 Antigen ,Virus ,Betacoronavirus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,medicine ,Humans ,CTLA-4 Antigen ,Respiratory system ,Pandemics ,Cancer ,030304 developmental biology ,Immunosuppression Therapy ,Oncologists ,Respiratory Distress Syndrome ,0303 health sciences ,Interleukin-6 ,SARS-CoV-2 ,business.industry ,Incidence ,Comment ,COVID-19 ,Immunotherapy ,medicine.disease ,Immune checkpoint ,Blockade ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Coronavirus Infections ,business ,Immunosuppressive Agents - Abstract
SummaryThe rapid emergence of COVID-19 has sent shockwaves through healthcare systems globally, with cancer patients at increased risk. The interplay of the virus and host immune system has been implicated in the development of ARDS. Immunotherapy agents have the potential to adversely potentiate this phenomenon, requiring careful real-world observation.
- Published
- 2020
45. Representative Sequencing: Unbiased Sampling of Solid Tumor Tissue
- Author
-
Richard Marais, Andrew Furness, Thomas B.K. Watkins, Maise Al-Bakir, Dionysis Papadatos-Pastos, Maryam Razaq, Mairead McKenzie, Lisa Pickering, Peter Ellery, Jonathan Ledermann, Andrew V. Biankin, Richard J. Gilbertson, Peter Cockcroft, Mary Mangwende, Sophie Ward, Mary Falzon, Cristina Naceur-Lombardelli, Carlos Caldas, Karla Pearce, Jacqui Shaw, Salma Kadiri, Lars Dyrskjøt, Mark Linch, Daniel Burgess, Nelson Alexander, Christian H. Ottensmeier, Kevin G. Blyth, Lisa L. Gallegos, Rodelaine Wilson, Hayley Bridger, Fiona H Blackhall, Peter J. Parker, Charles Swanton, Allan Hackshaw, Gert Attard, Gordon Stamp, Dean A. Fennell, Debra H. Josephs, Andrew P. Robinson, Kim Edmonds, Tina Mackay, Mita Afroza Akther, Miriam Mitchison, Sam M. Janes, Giorgia Trevisan, Adrienne M. Flanagan, Alison Cluroe, Henning Walczak, Stuart Horswell, Lena Karapagniotou, Simon Tavaré, Sarah Sarker, Teresa Marafioti, Matt Krebs, Anna Green, Philippe Lamy, Reena Khiroya, Samantha M. Hill, Andrew Tutt, Ashish Chandra, Selvaraju Veeriah, Faye Gishen, Lisa Thompson, Sarah Vaughan, Stacey Stanislaw, Kevin Litchfield, Colin Watts, Caroline Dive, Zayd Tippu, Ron Sinclair, Merche Jimenez-Linan, Lavinia Spain, Lizi Manzano, Zoe Rhodes, Caroline Stirling, Elena Provenzano, Andrew Rowan, Katey S. S. Enfield, Vasiliki Michalarea, Nahid Sheikh, Antonia Toncheva, Charlotte Ferris, James Spicer, Kai-Keen Shiu, Aida Murra, Gerald Langman, Scott Thomas Colville Shepherd, Nicholas McGranahan, Fabio Gomes, Daniel Hochhauser, Hang Xu, Sergio A. Quezada, James Larkin, Siow Ming Lee, Chi-wah Lok, Massimo Loda, Gary Middleton, Hollie Bancroft, Jo Dransfield, David Moore, Jennifer Thomas, Rebecca C. Fitzgerald, Peter Colloby, Annika Fendler, Emma Beddowes, Ultan McDermott, Christopher Abbosh, Uzma Asghar, Martin Forster, John Le Quesne, Katherine F. Leith, Paddy Stone, Bernard Olisemeke, Bruce Tanchel, Laura Farrelly, Grant D. Stewart, Justine Korteweg, Sanjay Jogai, Nnenna Kanu, Desiree Schnidrig, Heidi Rosenbaum, Elaine Borg, Mat Carter, Anthony J. Chalmers, Andrew C. Kidd, Alexandre Harlé, Ula Mahadeva, Crispin T. Hiley, Fiona Byrne, Heather Shaw, Mariam Jamal-Hanjani, Karin A. Oien, Ian Proctor, Joanne Webb, Sioban Fraser, Sarah Howlett, Ruby Stewart, Peter Van Loo, Nadia Yousaf, Tanya Ahmad, Martin Collard, Sanjay Popat, James D. Brenton, Sarah Rudman, Lewis Au, David G. Harrison, Elias Pintus, Patricia Roxburgh, Olivia Curtis, Ben Deakin, Ariana Huebner, Debra Enting, Simranpreet Summan, S. Baijal, Iver Nordentoft, Carol Jones, Haixi Yan, Sebastian Brandner, Tariq Enver, Lara-Rose Iredale, Yvonne Summers, Babu Naidu, Abby Sharp, Stephen Hazell, Aoune Barhoumi, Emma Nye, Robert E. Hynds, Sharmistha Ghosh, Emilia L. Lim, Ben Shum, Nikki Hunter, John Bridgewater, Eleanor Carlyle, Stephan Beck, Nicolai Juul Birkbak, Steve Hazell, Samra Turajlic, Amy Kerr, Ian Tomlinson, Adrian Tookman, Litchfield, Kevin [0000-0002-3725-0914], Birkbak, Nicolai J [0000-0003-1613-9587], Nordentoft, Iver [0000-0003-4856-4086], Dyrskjøt, Lars [0000-0001-7061-9851], Apollo - University of Cambridge Repository, Division of genetics and epidemiology, The institute of cancer research [London], The Wellcome Trust Centre for Human Genetics [Oxford], University of Oxford [Oxford], Centre de Recherche en Automatique de Nancy (CRAN), Université de Lorraine (UL)-Centre National de la Recherche Scientifique (CNRS), The Royal Marsden, University of Arizona, University of Western Ontario, Physic and Astronomy Department, University of Western Ontario (UWO), Center for Biological Sequence Analysis [Lyngby], Technical University of Denmark [Lyngby] (DTU), Laboratoire d'Astrophysique de Marseille (LAM), Aix Marseille Université (AMU)-Institut national des sciences de l'Univers (INSU - CNRS)-Centre National d'Études Spatiales [Toulouse] (CNES)-Centre National de la Recherche Scientifique (CNRS), Aarhus University Hospital, Cancer Research UK Lung Cancer Centre of Excellence [Londres, Royaume-Uni], University College of London [London] (UCL), Comprehensive Cancer Center, UCL Cancer Institute [University College London], The Wellcome Trust Sanger Institute [Cambridge], Research Department of Pathology, Innovation North - Faculty of Information and Technology, Leeds Metropolitan University, Department of histopathology, University College Hospital, Mammalian Genetics Laboratory, Centre for Research on Adaptive Nanostructures and Nanodevices, Trinity College Dublin, Cancer Research UK London Research Institute, Guy's and St Thomas' Hospital [London], Breakthrough Breast Cancer Centre, London Institute of Cancer, Cancer Research UK Cambridge Research Institute and Department of Oncology, University of Cambridge, University of Cambridge [UK] (CAM), Cancer Division [Sydney, Australia] (The Kinghorn Cancer Centre), Garvan Institute of Medical Research [Sydney, Australia], Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, University of Leicester, Experimental Cancer Medicine Centre, University of Southampton-Faculty of Medicine, Moses, Wittemyer, Harrison and Woodruff, P.C., University of Oxford, Danmarks Tekniske Universitet = Technical University of Denmark (DTU), and Garvan Institute of medical research
- Subjects
0301 basic medicine ,tumor sequencing ,medicine.medical_specialty ,Sampling protocol ,tumor mutational burden ,tumor sampling ,Lung Neoplasms ,molecular profiling ,Biopsy ,homogenization ,Tissue sample ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Biomarkers, Tumor ,Humans ,Solid tumor ,lcsh:QH301-705.5 ,Representative sampling ,medicine.diagnostic_test ,Manchester Cancer Research Centre ,ResearchInstitutes_Networks_Beacons/mcrc ,Clonal structure ,biomarkers ,High-Throughput Nucleotide Sequencing ,Tumor tissue ,tumor hetereogeneity ,Tumor Burden ,030104 developmental biology ,lcsh:Biology (General) ,representative sampling ,Urinary Bladder Neoplasms ,Mutation ,Radiology ,030217 neurology & neurosurgery - Abstract
International audience; Although thousands of solid tumors have been sequenced to date, a fundamental under-sampling bias isinherent in current methodologies. This is caused by a tissue sample input of fixed dimensions (e.g., 6 mmbiopsy), which becomes grossly under-powered as tumor volume scales. Here, we demonstrate representative sequencing (Rep-Seq) as a new method to achieve unbiased tumor tissue sampling. Rep-Seq uses fixed residual tumor material, which is homogenized and subjected to next-generation sequencing. Analysis of intratumor tumor mutation burden (TMB) variability shows a high level of misclassification using current single-biopsy methods, with 20% of lung and 52% of bladder tumors having at least one biopsy with high TMB butlow clonal TMB overall. Misclassification rates by contrast are reduced to 2% (lung) and 4% (bladder) when a more representative sampling methodology is used. Rep-Seq offers an improved sampling protocol for tumor profiling, with significant potential for improved clinical utility and more accurate deconvolution of clonal structure.
- Published
- 2020
- Full Text
- View/download PDF
46. The UK Coronavirus Cancer Monitoring Project: protecting patients with cancer in the era of COVID-19
- Author
-
Sam Benkwitz-Beford, Ashley Wilder Smith, Jean-Baptiste Cazier, Helen Curley, Naomi Longworth, Simon Branford, Iris Anil, Archana Sharma-Oates, D.J. Hughes, Andrew Edmondson, Naomi A. Campton, Shivan Sivakumar, Alvin J.X. Lee, Thomas Starkey, Vinton Cheng, Jamie J. D’Costa, Anshita Goel, Karin Purshouse, Claire Palles, Gary Middleton, Simon Hartley, Simon Thompson, Anna Olsson-Brown, Rachel Kerr, Christopher P. Middleton, Roland Arnold, Lennard Y W Lee, Carol Sandys, Csilla Várnai, and Piangfan Naksukpaiboon
- Subjects
medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Pneumonia, Viral ,medicine.disease_cause ,Vulnerable Populations ,Patient safety ,Betacoronavirus ,Neoplasms ,Pandemic ,Medicine ,Humans ,Intensive care medicine ,Pandemics ,Coronavirus ,biology ,business.industry ,SARS-CoV-2 ,Cancer ,COVID-19 ,medicine.disease ,biology.organism_classification ,United Kingdom ,Oncology ,Patient Safety ,business ,Coronavirus Infections - Published
- 2020
47. Use of an integrated pan-cancer oncology enrichment NGS assay to measure tumour mutational burden and detect clinically actionable variants
- Author
-
Gerald Martin, Andrew D Beggs, Valerie Pestinger, John M Findlay, Gary Middleton, Matthew D. Smith, Jean-Francois Laes, Phillipe Taniere, and Toju Sillo
- Subjects
Whole genome sequencing ,Oncology ,FASTQ format ,Structural variation ,medicine.medical_specialty ,Pan cancer ,Internal medicine ,medicine ,Formalin fixed ,Biology ,Indel ,Gene - Abstract
IntroductionThe identification of tumour mutational burden (TMB) as a biomarker of response to PD-1 immunotherapy has necessitated the development of genomic assays to measure this. We carried out comprehensive molecular profiling of cancers using the Illumina TruSight Oncology panel (TSO500) and compared to whole genome sequencing.MethodsCancer samples derived from formalin fixed material were profiled on the TSO500 panel, sequenced on an Illumina NextSeq 500 instrument and processed through the TSO500 Docker Pipeline. Either FASTQ files (PierianDx) or VCF files (OncoKDM) were processed to understand clinical actionabilityResultsIn total, 108 samples (a mixture of colorectal, lung, oesophageal and control samples) were processed via the DNA panel. There was good correlation between TMB, SNV, indels and CNV as predicted by TSO500 and WGS (R2>0.9) and good reproducibility, with less than 5% variability between repeated controls. For the RNA panel, 13 samples were processed, with all known fusions observed via orthogonal techniques detected. For clinical actionability 72 Tier 1 variants and 297 Tier 2 variants were identified with clinical trials identified for all patients.ConclusionsThe TruSight Oncology 500 assay accurately measures TMB, MSI, single nucleotide variants, indels, copy number/structural variation and gene fusions when compared to whole genome sequencing and orthogonal technologies. Coupled with a clinical annotation pipeline this provides a powerful methodology for identification of clinically actionable variants.
- Published
- 2020
- Full Text
- View/download PDF
48. The National Lung Matrix Trial of personalized therapy in lung cancer
- Author
-
Martin Forster, Joshua Savage, Dee Wherton, Yvonne Summers, Emilia L. Lim, P. Jain, James Spicer, Noelle O'Rourke, Alison Brewster, Thomas B.K. Watkins, Peter Fletcher, Tara C. Mills, Charles Swanton, David Gilligan, Sanjay Popat, Melanie Mackean, Maria Antonietta Cerone, Alastair Greystoke, Judith Cave, Lucinda Billingham, Manita Mehmi, Rowena Sharpe, Elizabeth Toy, Gary Middleton, Amanda Farley, Adam Dangoor, and Timothy A. Yap
- Subjects
0301 basic medicine ,Oncology ,Genetic Markers ,medicine.medical_specialty ,Lung Neoplasms ,Genotype ,MEDLINE ,Tobacco smoke ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Clinical Protocols ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Smoke ,Carcinoma ,Medicine ,Humans ,Molecular Targeted Therapy ,Precision Medicine ,Lung cancer ,Adaptive clinical trial ,Clinical Trials as Topic ,Multidisciplinary ,business.industry ,Patient Selection ,Smoking ,High-Throughput Nucleotide Sequencing ,Bayes Theorem ,Oncogenes ,medicine.disease ,Precision medicine ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,Triage ,business ,Cohort study - Abstract
The majority of targeted therapies for non-small-cell lung cancer (NSCLC) are directed against oncogenic drivers that are more prevalent in patients with light exposure to tobacco smoke1–3. As this group represents around 20% of all patients with lung cancer, the discovery of stratified medicine options for tobacco-associated NSCLC is a high priority. Umbrella trials seek to streamline the investigation of genotype-based treatments by screening tumours for multiple genomic alterations and triaging patients to one of several genotype-matched therapeutic agents. Here we report the current outcomes of 19 drug–biomarker cohorts from the ongoing National Lung Matrix Trial, the largest umbrella trial in NSCLC. We use next-generation sequencing to match patients to appropriate targeted therapies on the basis of their tumour genotype. The Bayesian trial design enables outcome data from open cohorts that are still recruiting to be reported alongside data from closed cohorts. Of the 5,467 patients that were screened, 2,007 were molecularly eligible for entry into the trial, and 302 entered the trial to receive genotype-matched therapy—including 14 that re-registered to the trial for a sequential trial drug. Despite pre-clinical data supporting the drug–biomarker combinations, current evidence shows that a limited number of combinations demonstrate clinically relevant benefits, which remain concentrated in patients with lung cancers that are associated with minimal exposure to tobacco smoke. Current outcomes are reported from the ongoing National Lung Matrix Trial, an umbrella trial for the treatment of non-small-cell lung cancer in which patients are triaged according to their tumour genotype and matched with targeted therapeutic agents.
- Published
- 2020
49. Interplay between whole-genome doubling and the accumulation of deleterious alterations in cancer evolution
- Author
-
Javier Herrero, Gary Middleton, John Edwards, Richard Kevin Stone, Dominic Rothwell, Fiona Blackhall, Lynsey Priest, Marco Scarci, Caroline Dive, Mickael Escudero, Babu Naidu, Christian Ottensmeier, Michael Shackcloth, Kevin Litchfield, Nicolai Birkbak, Istvan Csabai, Neal Navani, Helen Lowe, JAMES READING, Paulo De Sousa, Francesca Chemi, Nicholas McGranahan, Diana Johnson, Hugo Aerts, John Le Quesne, Benny Chain, Stuart Horswell, and Robert Hynds
- Subjects
Genetics ,0303 health sciences ,Mutation ,Cancer ,Genomics ,Biology ,medicine.disease_cause ,medicine.disease ,Genome ,Human genetics ,Loss of heterozygosity ,03 medical and health sciences ,Negative selection ,0302 clinical medicine ,Gene duplication ,medicine ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Whole-genome doubling (WGD) is a prevalent event in cancer, involving a doubling of the entire chromosome complement. However, despite its prevalence and prognostic relevance, the evolutionary selection pressures for WGD in cancer have not been investigated. Here, we combine evolutionary simulations with an analysis of cancer sequencing data to explore WGD during cancer evolution. Simulations suggest that WGD can be selected to mitigate the irreversible, ratchet-like, accumulation of deleterious somatic alterations, provided that they occur at a sufficiently high rate. Consistent with this, we observe an enrichment for WGD in tumor types with extensive loss of heterozygosity, including lung squamous cell carcinoma and triple-negative breast cancers, and we find evidence for negative selection against homozygous loss of essential genes before, but not after, WGD. Finally, we demonstrate that loss of heterozygosity and temporal dissection of mutations can be exploited to identify novel tumor suppressor genes and to obtain a deeper characterization of known cancer genes.
- Published
- 2020
- Full Text
- View/download PDF
50. Genomic loss of heterozygosity and survival in the REAL3 trial
- Author
-
Catherine Cafferkey, Elizabeth C Smyth, Naureen Starling, David Watkins, Minh Nam Nguyen, Wasat Mansoor, Alicia Okines, Ian Chau, David Cunningham, Andrea Loehr, Jonathan Wadsley, Thomas Harding, Andrew Wotherspoon, Gary Middleton, Ruwaida Begum, Tom Crosby, Clare Peckitt, Mitch Raponi, T. Waddell, and Sheela Rao
- Subjects
0301 basic medicine ,oesophageal cancer ,medicine.medical_specialty ,endocrine system diseases ,Population ,chemotherapy ,Loss of heterozygosity ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Medicine ,Differential survival ,Progression-free survival ,Rucaparib ,education ,neoplasms ,education.field_of_study ,business.industry ,gastric cancer ,Cancer ,medicine.disease ,University hospital ,stomatognathic diseases ,030104 developmental biology ,Oncology ,chemistry ,homologous recombination deficiency ,030220 oncology & carcinogenesis ,loss of heterozygosity ,business ,Homologous Recombination Deficiency ,Research Paper - Abstract
// Elizabeth C. Smyth 1, 2, * , Catherine Cafferkey 1, * , Andrea Loehr 3 , Tom Waddell 1, 4 , Ruwaida Begum 1 , Clare Peckitt 5 , Thomas C. Harding 3 , Minh Nguyen 3 , Alicia F. Okines 1 , Mitch Raponi 3 , Sheela Rao 1 , David Watkins 1 , Naureen Starling 1 , Gary W. Middleton 6 , Jonathan Wadsley 7 , Wasat Mansoor 4 , Tom Crosby 8 , Andrew Wotherspoon 9 , Ian Chau 1 and David Cunningham 1 1 Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom 2 Current affiliation: Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom 3 Clovis Oncology, San Francisco, CA, United States of America 4 Current affiliation: Department of Medical Oncology, Christie Hospital, Manchester, United Kingdom 5 Department of Clinical Research & Development, Royal Marsden Hospital, London & Sutton, United Kingdom 6 Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom 7 Department of Medical Oncology, Weston Park Hospital, Sheffield, United Kingdom 8 Department of Clinical Oncology, Velindre Hospital, Cardiff, Wales, United Kingdom 9 Department of Histopathology, Royal Marsden Hospital, London & Surrey, United Kingdom * These authors have contributed equally to this work Correspondence to: David Cunningham, email: david.cunningham@rmh.nhs.uk Keywords: gastric cancer; oesophageal cancer; chemotherapy; homologous recombination deficiency; loss of heterozygosity Received: August 01, 2018 Accepted: October 06, 2018 Published: November 30, 2018 ABSTRACT Background: Homologous recombination deficiency (HRD) measured using a genomic signature for loss of heterozygosity (LOH) predicts benefit from rucaparib in ovarian cancer. We hypothesized that some oesophagogastric cancers will have high-LOH which would be prognostic in patients treated with platinum chemotherapy. Methods: Diagnostic biopsy DNA from patients treated in the REAL3 trial was sequenced using the Foundation Medicine T5 next-generation sequencing (NGS) assay. An algorithm quantified the percentage of interrogable genome with LOH. Multidimensional optimization was performed to identify a cut-off dichotomizing the population into LOH-high and low groups associated with differential survival outcomes. Results: Of 158 available samples, 117 were successfully sequenced; LOH was derived for 74 of these. A cut-off of 21% genomic LOH defined an LOH-high subgroup (n=10, 14% of population) who had median overall survival (OS) of 18.3 months (m) versus 11m for the LOH-low group (HR 0.55 95% CI 0.19-0.97, p= 0.10). Progression free survival (PFS) for LOH-high and LOH-low groups was 10.7m and 7.3m (HR 0.61 (95% CI 0.21 – 1.09, p=0.09). Sensitivity analysis censoring operated patients (n=4), demonstrated OS of 18.3m vs. 10.2m (HR 0.43, 95% CI (0.20-0.92), p=0.02; PFS was 10.5m vs. 7.2m (HR 0.55, (95% CI 0.26-1.17), p=0.09 for LOH-high and LOH-low. Conclusion: HRD assessment using an algorithmically derived LOH signature on a standard NGS panel identifies oesophagogastric cancer patients with high LOH who have prolonged survival when treated with platinum chemotherapy. Validation work will determine the signature’s predictive value in patients treated with a PARP inhibitor and with platinum chemotherapy.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.