12 results on '"Andrew J.S. Furness"'
Search Results
2. Dabrafenib plus trametinib versus anti-PD-1 monotherapy as adjuvant therapy in BRAF V600-mutant stage III melanoma after definitive surgery: a multicenter, retrospective cohort studyResearch in context
- Author
-
Xue Bai, Ahmed Shaheen, Charlotte Grieco, Paolo D. d’Arienzo, Florentia Mina, Juliane A. Czapla, Aleigha R. Lawless, Eleonora Bongiovanni, Umberto Santaniello, Helena Zappi, Dominika Dulak, Andrew Williamson, Rebecca Lee, Avinash Gupta, Caili Li, Lu Si, Martina Ubaldi, Naoya Yamazaki, Dai Ogata, Rebecca Johnson, Benjamin C. Park, Seungyeon Jung, Gabriele Madonna, Juliane Hochherz, Yoshiyasu Umeda, Yasuhiro Nakamura, Christoffer Gebhardt, Lucia Festino, Mariaelena Capone, Paolo Antonio Ascierto, Douglas B. Johnson, Serigne N. Lo, Georgina V. Long, Alexander M. Menzies, Kenjiro Namikawa, Mario Mandala, Jun Guo, Paul Lorigan, Yana G. Najjar, Andrew Haydon, Pietro Quaglino, Genevieve M. Boland, Ryan J. Sullivan, Andrew J.S. Furness, Ruth Plummer, and Keith T. Flaherty
- Subjects
Adjuvant therapy ,BRAF V600 mutation ,Melanoma ,PD-1 ,Dabrafenib/trametinib ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Both dabrafenib/trametinib (D/T) and anti-PD-1 monotherapy (PD-1) are approved adjuvant therapies for patients with stage III BRAF V600-mutant melanoma. However, there is still a lack of head-to-head comparative data. We aimed to describe efficacy and toxicity outcomes for these two standard therapies across melanoma centers. Methods: This multicenter, retrospective cohort study was conducted in 15 melanoma centers in Australia, China, Germany, Italy, Japan, UK, and US. We included adult patients with resected stage III BRAF V600-mutant melanoma who received either adjuvant D/T or PD-1 between Jul 2015 and Oct 2022. The primary endpoint was relapse-free survival (RFS). Secondary endpoints included overall survival (OS), recurrence pattern and toxicity. Findings: We included 598 patients with stage III BRAF V600-mutant melanoma who received either adjuvant D/T (n = 393 [66%]) or PD-1 (n = 205 [34%]) post definitive surgery between Jul 2015 and Oct 2022. At a median follow-up of 33 months (IQR 21–43), the median RFS was 51.0 months (95% CI 41.0-not reached [NR]) in the D/T group, significantly longer than PD-1 (44.8 months [95% CI 28.5-NR]) (univariate: HR 0.66, 95% CI 0.50–0.87, P = 0.003; multivariate: HR 0.58, 95% CI 0.39–0.86, P = 0.007), with comparable OS with PD-1 (multivariate, HR 0.90, 95% CI 0.48–1.70, P = 0.75). Similar findings were observed using a restricted-mean-survival-time model. Among those who experienced recurrence, the proportion of distant metastases was higher in the D/T cohort. D/T had a higher incidence of treatment modification due to adverse events (AEs) than PD-1, but fewer persistent AEs. Interpretation: In patients with stage III BRAF V600-mutant melanoma post definitive surgery, D/T yielded better RFS than PD-1, with higher transient but lower persistent toxicity, and comparable OS. D/T seems to provide a better outcome compared with PD-1, but a longer follow-up and ideally a large prospective trial are needed. Funding: Dr. Xue Bai was supported by the Beijing Hospitals Authority Youth Programme (QMS20211101) for her efforts devoted to this study. Dr. Keith T. Flaherty was funded by Adelson Medical Research Foundation for the efforts devoted to this study.
- Published
- 2023
- Full Text
- View/download PDF
3. The spectrum of cardiovascular complications related to immune-checkpoint inhibitor treatment
- Author
-
Maria Sol Andres, Sivatharshini Ramalingam, Stuart D. Rosen, John Baksi, Rajdeep Khattar, Yulia Kirichenko, Kate Young, Nadia Yousaf, Alicia Okines, Robert Huddart, Kevin Harrington, Andrew J.S. Furness, Samra Turajlic, Lisa Pickering, Sanjay Popat, James Larkin, and Alexander R. Lyon
- Subjects
Cancer survivorship ,Immunotherapy ,Myocarditis ,Cardiomyopathy ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The full range of cardiovascular complications related to the use of Immune checkpoint inhibitors (ICI) is not fully understood. We aim to describe the spectrum of cardiovascular adverse events (cvAEs) by presenting our real-world experience of the diagnosis and management of these complications. Methods Two thousand six hundred and forty-seven (2647) patients were started on ICI treatment between 2014 and 2020. Data from 110 patients referred to the cardio-oncology service with a suspected cvAE was collected prospectively and analysed. Results Eighty-nine patients (3.4%) were confirmed to have cvAEs while on ICI therapy. Myocarditis was the most frequent event (33/89), followed by tachyarrhythmia (27/89), non-inflammatory left ventricular dysfunction (NILVD) (15/89) and pericarditis (7/89). Results from myocarditis and non-inflammatory left ventricular dysfunction cohorts were compared. Myocarditis and NILVD showed significant differences in respect toof troponin elevation, cardiac magnetic resonance abnormalities and ventricular function. Dual ICI therapy and other immune related adverse events were more frequently associated with myocarditis than NILVD. There was a significant difference in the median time from starting ICI treatment to presentation with myocarditis versus NILVD (12 vs 26 weeks p = 0.049). Through early recognition of myocarditis, prompt treatment with steroids and interruption of ICI, there were no cardiovascular in-hospital deaths. NILVD did not require steroid treatment and ICI could be restarted safely. Conclusions The full spectrum of cardiovascular complications in patients with immune checkpoint inhibitors is much broader than initially described. Myocarditis remains the most frequent cvAE related to ICI treatment. A novel type of myocardial injury was observed and defined as Atrial tachyarrhythmias and NILVD were also frequent in this cohort. NILVD has a This differs fromdifferent presentation from ICI-related myocarditis, mainly usually presenting afterby the lack of inflammatory features on CMR and biomarkers and a later presentation in time.
- Published
- 2022
- Full Text
- View/download PDF
4. Ipilimumab plus nivolumab in patients with symptomatic melanoma brain metastasis requiring corticosteroids
- Author
-
Simona Manacorda, Maria De Toro Carmena, Ciara Malone, Ha Mo Linh Le, Andrew J.S. Furness, James Larkin, and Andreas M. Schmitt
- Subjects
Cancer Research ,Oncology - Published
- 2023
5. Sotrovimab restores neutralization against current Omicron subvariants in patients with blood cancer
- Author
-
Mary Y. Wu, Scott T.C. Shepherd, Annika Fendler, Edward J. Carr, Lewis Au, Ruth Harvey, Giulia Dowgier, Agnieszka Hobbs, Lou S. Herman, Martina Ragno, Lorin Adams, Andreas M. Schmitt, Zayd Tippu, Benjamin Shum, Sheima Farag, Aljosja Rogiers, Nicola O’Reilly, Philip Bawumia, Callie Smith, Eleanor Carlyle, Kim Edmonds, Lyra Del Rosario, Karla Lingard, Mary Mangwende, Lucy Holt, Hamid Ahmod, Justine Korteweg, Tara Foley, Taja Barber, Stephanie Hepworth, Andrea Emslie-Henry, Niamh Caulfield-Lynch, Fiona Byrne, Daqi Deng, Bryan Williams, Michael Brown, Simon Caidan, Mike Gavrielides, James I. MacRae, Gavin Kelly, Kema Peat, Denise Kelly, Aida Murra, Kayleigh Kelly, Molly O’Flaherty, Sanjay Popat, Nadia Yousaf, Shaman Jhanji, Kate Tatham, David Cunningham, Nicholas Van As, Kate Young, Andrew J.S. Furness, Lisa Pickering, Rupert Beale, Charles Swanton, Sonia Gandhi, Steve Gamblin, David L.V. Bauer, George Kassiotis, Michael Howell, Susanna Walker, Emma Nicholson, James Larkin, Emma C. Wall, and Samra Turajlic
- Subjects
Cancer Research ,Oncology - Published
- 2023
6. Data from Late-Stage Metastatic Melanoma Emerges through a Diversity of Evolutionary Pathways
- Author
-
Samra Turajlic, Mariam Jamal-Hanjani, Charles Swanton, Kevin Litchfield, James Larkin, Anna Green, Ula Mahadeva, Ruby Stewart, Lisa Pickering, Andrew J.S. Furness, Kate Young, Emma L. Nye, Gordon W.H. Stamp, Ian Proctor, Mary Falzon, David A. Moore, Miriam Mitchison, Elaine Borg, Cristina Naceur-Lombardelli, Jun Murai, Maise Al Bakir, Nicholas McGranahan, Ariana Huebner, Hang Xu, Aljosja Rogiers, Robert Mason, Joanna Lynch, Husayn Ahmed Pallikonda, Camille L. Gerard, Max Emmerich, Anne-Laure Cattin, Molly O'Flaherty, Charlotte Lewis, Justine Korteweg, Aida Murra, Jennifer Biano, Denise Kelly, Lauren Terry, Mary Mangwende, Sarah Vaughan, Sarah Sarker, Kayleigh Kelly, Kema Peat, Lauren Grostate, Karla Lingard, Zayd Tippu, Andreas M. Schmitt, Charlotte Spencer, Diana C.J. Spierings, Rene Wardenaar, Hilda van den Bos, Floris Foijer, Jaime Nobbs, Peta Hughes, Christina Messiou, Alexandra Renn, Nikki Hunter, Eleanor Carlyle, Kim Edmonds, Lewis Au, Elisa Piperni, Maria Goicoechea, Fiona Byrne, Benjamin Shum, Scott T.C. Shepherd, Desiree Schnidrig, Andrew Rowan, Irene Lobon, Alexander Coulton, and Lavinia Spain
- Abstract
Understanding the evolutionary pathways to metastasis and resistance to immune-checkpoint inhibitors (ICI) in melanoma is critical for improving outcomes. Here, we present the most comprehensive intrapatient metastatic melanoma dataset assembled to date as part of the Posthumous Evaluation of Advanced Cancer Environment (PEACE) research autopsy program, including 222 exome sequencing, 493 panel-sequenced, 161 RNA sequencing, and 22 single-cell whole-genome sequencing samples from 14 ICI-treated patients. We observed frequent whole-genome doubling and widespread loss of heterozygosity, often involving antigen-presentation machinery. We found KIT extrachromosomal DNA may have contributed to the lack of response to KIT inhibitors of a KIT-driven melanoma. At the lesion-level, MYC amplifications were enriched in ICI nonresponders. Single-cell sequencing revealed polyclonal seeding of metastases originating from clones with different ploidy in one patient. Finally, we observed that brain metastases that diverged early in molecular evolution emerge late in disease. Overall, our study illustrates the diverse evolutionary landscape of advanced melanoma.Significance:Despite treatment advances, melanoma remains a deadly disease at stage IV. Through research autopsy and dense sampling of metastases combined with extensive multiomic profiling, our study elucidates the many mechanisms that melanomas use to evade treatment and the immune system, whether through mutations, widespread copy-number alterations, or extrachromosomal DNA.
- Published
- 2023
7. Supplementary Figures S1-S25 from Late-Stage Metastatic Melanoma Emerges through a Diversity of Evolutionary Pathways
- Author
-
Samra Turajlic, Mariam Jamal-Hanjani, Charles Swanton, Kevin Litchfield, James Larkin, Anna Green, Ula Mahadeva, Ruby Stewart, Lisa Pickering, Andrew J.S. Furness, Kate Young, Emma L. Nye, Gordon W.H. Stamp, Ian Proctor, Mary Falzon, David A. Moore, Miriam Mitchison, Elaine Borg, Cristina Naceur-Lombardelli, Jun Murai, Maise Al Bakir, Nicholas McGranahan, Ariana Huebner, Hang Xu, Aljosja Rogiers, Robert Mason, Joanna Lynch, Husayn Ahmed Pallikonda, Camille L. Gerard, Max Emmerich, Anne-Laure Cattin, Molly O'Flaherty, Charlotte Lewis, Justine Korteweg, Aida Murra, Jennifer Biano, Denise Kelly, Lauren Terry, Mary Mangwende, Sarah Vaughan, Sarah Sarker, Kayleigh Kelly, Kema Peat, Lauren Grostate, Karla Lingard, Zayd Tippu, Andreas M. Schmitt, Charlotte Spencer, Diana C.J. Spierings, Rene Wardenaar, Hilda van den Bos, Floris Foijer, Jaime Nobbs, Peta Hughes, Christina Messiou, Alexandra Renn, Nikki Hunter, Eleanor Carlyle, Kim Edmonds, Lewis Au, Elisa Piperni, Maria Goicoechea, Fiona Byrne, Benjamin Shum, Scott T.C. Shepherd, Desiree Schnidrig, Andrew Rowan, Irene Lobon, Alexander Coulton, and Lavinia Spain
- Abstract
Supplementary figure 1: Cohort overview. Number of samples sequenced with whole exome, panel or whole RNA sequencing. Supplementary figure 2: Phylogeny and WGD events in CRUKP1047. Supplementary figure 3: Ploidy and SCNA burden. Supplementary figure 4: Overview of each case. Supplementary figure 5: MEDICC2 copy number sample trees. Supplementary figure 6: MEDICC tree of all exome samples demonstrating that samples cluster together by patient, and not by melanoma subtype. Supplementary figure 7: SCNA frequency of cutaneous (a), acral (b) and melanoma of unknown primary (MUP, c). Supplementary figure 8: Correlation between liver copy number distance to other sites and time of emergence after stage IV diagnosis. Supplementary figure 9: Examination of tumour heterogeneity of alterations to antigen-presentation machinery genes, with site and patient annotation. Supplementary figure 10: Boxplots indicating the proportion of losses in the cohort for each segment. Supplementary figure 11: Balance of expression between nonsynonymous mutations that were not predicted to be neoantigens and clonal predicted neoantigens. Supplementary figure 12: Barplot of TIL infiltration score frequencies, determined by pathologist assessment of histology, across all samples. Supplementary figure 13: Number of samples per patient that are classified as either none-low in terms of TILs or moderate-heavy. Supplementary figure 14: Histogram of purity for samples with RNA-seq data. Supplementary figure 15: TME deconvolution. Supplementary figure 16: The effect of metastatic site on transcriptional profiles. Supplementary figure 17: Association of PHF3 copy number with expression. Supplementary figure 18: Overview of gene fusions identified in RNA-seq data. Supplementary figure 19: Comparison of ploidy estimates from panel sequencing data, exome data and FISH. Supplementary figure 20: Comparison of ploidy estimates in panel, exome, FISH and single cell data. Supplementary figure 21: FACs sort plot for CRUKP2567 diaphragmatic metastasis. Supplementary figure 22: Ploidy and wGII values from single cell sequencing of FACS-sorted tumour cells. Supplementary figure 23: Copy number profiles on chromosome 5 for bulk samples from primary and DI_2_R2, a diaphragmatic metastasis. Supplementary figure 24: Histogram of purity of samples for which RNA-seq was performed faceted by patient. Supplementary figure 25: Histogram of purity of samples for which RNA-seq was performed faceted by tissue site.
- Published
- 2023
8. Supplementary Tables S1-S8 from Late-Stage Metastatic Melanoma Emerges through a Diversity of Evolutionary Pathways
- Author
-
Samra Turajlic, Mariam Jamal-Hanjani, Charles Swanton, Kevin Litchfield, James Larkin, Anna Green, Ula Mahadeva, Ruby Stewart, Lisa Pickering, Andrew J.S. Furness, Kate Young, Emma L. Nye, Gordon W.H. Stamp, Ian Proctor, Mary Falzon, David A. Moore, Miriam Mitchison, Elaine Borg, Cristina Naceur-Lombardelli, Jun Murai, Maise Al Bakir, Nicholas McGranahan, Ariana Huebner, Hang Xu, Aljosja Rogiers, Robert Mason, Joanna Lynch, Husayn Ahmed Pallikonda, Camille L. Gerard, Max Emmerich, Anne-Laure Cattin, Molly O'Flaherty, Charlotte Lewis, Justine Korteweg, Aida Murra, Jennifer Biano, Denise Kelly, Lauren Terry, Mary Mangwende, Sarah Vaughan, Sarah Sarker, Kayleigh Kelly, Kema Peat, Lauren Grostate, Karla Lingard, Zayd Tippu, Andreas M. Schmitt, Charlotte Spencer, Diana C.J. Spierings, Rene Wardenaar, Hilda van den Bos, Floris Foijer, Jaime Nobbs, Peta Hughes, Christina Messiou, Alexandra Renn, Nikki Hunter, Eleanor Carlyle, Kim Edmonds, Lewis Au, Elisa Piperni, Maria Goicoechea, Fiona Byrne, Benjamin Shum, Scott T.C. Shepherd, Desiree Schnidrig, Andrew Rowan, Irene Lobon, Alexander Coulton, and Lavinia Spain
- Abstract
S. Table 1: List of PEACE Consortium members. S. Table 2: Sample database used for the study for Panel, Exome and RNASeq samples. S. Table 3: Overview of lines of treatment given to each patient. S. Table 4: Lesions and patients included in the lesion-level response to ICI analysis. S. Table 5: Hallmark genesets significantly upregulated in normal tissue vs tumor tissue. Table shows p-values for tissue type, purity; T value for tissue type, purity; q-value for tissue type, purity (FDR corrected). S. Table 6: Hallmark genesets significantly upregulated in tumor tissue vs normal tissue. Table shows p-values for tissue type, purity; T value for tissue type, purity; q-value for tissue type, purity (FDR corrected). S. Table 7: Genes included in the target panel. S. Table 8: Tree topology comparisons between manually constructed trees and pairtree-constructed trees.
- Published
- 2023
9. Data from Marrow-Infiltrating Regulatory T Cells Correlate with the Presence of Dysfunctional CD4+PD-1+ Cells and Inferior Survival in Patients with Newly Diagnosed Multiple Myeloma
- Author
-
Kwee L. Yong, Sergio A. Quezada, Javier Herrero, Martin Pule, Manuel Rodriguez-Justo, Anthony Brooks, Dominic Patel, Oliver C. Cohen, Dunnya De-Silva, Huw Richards, Selina J. Chavda, Andrew J.S. Furness, Daria Galas-Filipowicz, Melody Chin, Lucia Conde, Jake Y. Henry, Ehsan Ghorani, Lydia Lee, and Nouf Alrasheed
- Abstract
Purpose:Immune dysregulation is described in multiple myeloma. While preclinical models suggest a role for altered T-cell immunity in disease progression, the contribution of immune dysfunction to clinical outcomes remains unclear. We aimed to characterize marrow-infiltrating T cells in newly diagnosed patients and explore associations with outcomes of first-line therapy.Experimental Design:We undertook detailed characterization of T cells from bone marrow (BM) samples, focusing on immune checkpoints and features of immune dysfunction, correlating with clinical features and progression-free survival.Results:We found that patients with multiple myeloma had greater abundance of BM regulatory T cells (Tregs) which, in turn, expressed higher levels of the activation marker CD25 compared with healthy donors. Patients with higher frequencies of Tregs had shorter PFS and a distinct Treg immune checkpoint profile (increased PD-1, LAG-3) compared with patients with lower frequencies of Tregs. Analysis of CD4 and CD8 effectors revealed that low CD4effector (CD4eff):Treg ratio and increased frequency of PD-1–expressing CD4eff cells were independent predictors of early relapse over and above conventional risk factors, such as genetic risk and depth of response. Ex vivo functional analysis and RNA sequencing revealed that CD4 and CD8 cells from patients with greater abundance of CD4effPD-1+ cells displayed transcriptional and secretory features of dysfunction.Conclusions:BM-infiltrating T-cell subsets, specifically Tregs and PD-1–expressing CD4 effectors, negatively influence clinical outcomes in newly diagnosed patients. Pending confirmation in larger cohorts and further mechanistic work, these immune parameters may inform new risk models, and present potential targets for immunotherapeutic strategies.
- Published
- 2023
10. Figure from Marrow-Infiltrating Regulatory T Cells Correlate with the Presence of Dysfunctional CD4+PD-1+ Cells and Inferior Survival in Patients with Newly Diagnosed Multiple Myeloma
- Author
-
Kwee L. Yong, Sergio A. Quezada, Javier Herrero, Martin Pule, Manuel Rodriguez-Justo, Anthony Brooks, Dominic Patel, Oliver C. Cohen, Dunnya De-Silva, Huw Richards, Selina J. Chavda, Andrew J.S. Furness, Daria Galas-Filipowicz, Melody Chin, Lucia Conde, Jake Y. Henry, Ehsan Ghorani, Lydia Lee, and Nouf Alrasheed
- Abstract
Supplementary tables and figures
- Published
- 2023
11. Functional immune responses against SARS-CoV-2 variants of concern after fourth COVID-19 vaccine dose or infection in patients with blood cancer
- Author
-
Annika Fendler, Scott T.C. Shepherd, Lewis Au, Mary Wu, Ruth Harvey, Katalin A. Wilkinson, Andreas M. Schmitt, Zayd Tippu, Benjamin Shum, Sheima Farag, Aljosja Rogiers, Eleanor Carlyle, Kim Edmonds, Lyra Del Rosario, Karla Lingard, Mary Mangwende, Lucy Holt, Hamid Ahmod, Justine Korteweg, Tara Foley, Taja Barber, Andrea Emslie-Henry, Niamh Caulfield-Lynch, Fiona Byrne, Daqi Deng, Svend Kjaer, Ok-Ryul Song, Christophe J. Queval, Caitlin Kavanagh, Emma C. Wall, Edward J. Carr, Simon Caidan, Mike Gavrielides, James I. MacRae, Gavin Kelly, Kema Peat, Denise Kelly, Aida Murra, Kayleigh Kelly, Molly O’Flaherty, Robyn L. Shea, Gail Gardner, Darren Murray, Sanjay Popat, Nadia Yousaf, Shaman Jhanji, Kate Tatham, David Cunningham, Nicholas Van As, Kate Young, Andrew J.S. Furness, Lisa Pickering, Rupert Beale, Charles Swanton, Sonia Gandhi, Steve Gamblin, David L.V. Bauer, George Kassiotis, Michael Howell, Emma Nicholson, Susanna Walker, Robert J. Wilkinson, James Larkin, and Samra Turajlic
- Subjects
Model organisms ,COVID-19 Vaccines ,Immunology ,T cells ,Infectious Disease ,variants of concern ,Antibodies, Viral ,Biochemistry & Proteomics ,General Biochemistry, Genetics and Molecular Biology ,Signalling & Oncogenes ,Ecology,Evolution & Ethology ,Neoplasms ,Humans ,neutralizing antibodies ,BNT162 Vaccine ,Computational & Systems Biology ,Chemical Biology & High Throughput ,Human Biology & Physiology ,SARS-CoV-2 ,blood cancer ,FOS: Clinical medicine ,Stem Cells ,Clinical Studies as Topic ,Genome Integrity & Repair ,Immunity ,Neurosciences ,COVID-19 ,Cell Biology ,Tumour Biology ,Antibodies, Neutralizing ,Metabolism ,Cell Cycle & Chromosomes ,Genetics & Genomics ,Developmental Biology ,Structural Biology & Biophysics - Abstract
Patients with blood cancer continue to have a greater risk of inadequate immune responses following three COVID-19 vaccine doses and risk of severe COVID-19 disease. In the context of the CAPTURE study (NCT03226886), we report immune responses in 80 patients with blood cancer who received a fourth dose of BNT162b2. We measured neutralizing antibody titers (NAbTs) using a live virus microneutralization assay against wild-type (WT), Delta, and Omicron BA.1 and BA.2 and T cell responses against WT and Omicron BA.1 using an activation-induced marker (AIM) assay. The proportion of patients with detectable NAb titers and T cell responses after the fourth vaccine dose increased compared with that after the third vaccine dose. Patients who received B cell-depleting therapies within the 12 months before vaccination have the greatest risk of not having detectable NAbT. In addition, we report immune responses in 57 patients with breakthrough infections after vaccination.
- Published
- 2022
- Full Text
- View/download PDF
12. Meta-analysis of tumor and T cell intrinsic mechanisms of sensitization to checkpoint inhibition
- Author
-
Kevin Litchfield, James L. Reading, Clare Puttick, Chris Abbosh, Robert Bentham, Thomas B. K. Watkins, Rachel Rosenthal, Dhruva Biswas, Emilia Lim, Maise AL-Bakir, Virginia Turati, José Afonso Guerra-Assunção, Lucia Conde, Andrew J.S. Furness, Sunil Kumar Saini, Sine R Hadrup, Javier Herrero, Andrew Rowan, Tariq Enver, Matthew D. Hellmann, James Larkin, Samra Turajlic, Sergio Quezada, Nicholas McGranahan, and Charles Swanton
- Abstract
Checkpoint inhibitors (CPIs) augment adaptive immunity. Systematic pan-tumor analyses may reveal the relative importance of tumour cell intrinsic and microenvironmental features underpinning CPI sensitization. Here we collated whole-exome and transcriptomic data for >1000 CPI-treated patients across eight tumor-types, utilizing standardized bioinformatics-workflows and clinical outcome-criteria to validate multivariate predictors of CPI-sensitization. Clonal-TMB was the strongest predictor of CPI response, followed by TMB and CXCL9 expression. Subclonal-TMB, somatic copy alteration burden and HLA-evolutionary divergence failed to attain significance. Discovery analysis identified two additional determinants of CPI-response supported by prior functional evidence: 9q34.3 (TRAF2) loss and CCND1 amplification, both independently validated in >1600 CPI-treated patients. We find evidence for collateral sensitivity, likely mediated through selection for CDKN2A-loss, with 9q34.3 loss as a passenger event leading to CPI-sensitization. Finally, scRNA sequencing of clonal neoantigen-reactive CD8-TILs, combined with bulk RNAseq analysis of CPI responding tumors, identified CCR5 and CXCL13 as T cell-intrinsic mediators of CPI-sensitisation.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.