Professor Samra Turajlic

Group Leader: Melanoma and Kidney Cancer

OrcID: 0000-0001-8846-136X

Email: [email protected]

Location: Chelsea

Dr Samra Turajlic (Profile Picture)

OrcID: 0000-0001-8846-136X

Email: [email protected]

Location: Chelsea

Biography

Samra Turajlic completed her undergraduate studies at Oxford University and her clinical training at UCL's medical school.  She gained a PhD in 2013 from the Institute of Cancer Research in the field of melanoma genetics and targeted therapy resistance. 

In 2014, she was awarded a Cancer Research UK Clinician Scientist Fellowship to study cancer evolution at the Francis Crick Institute. She completed her training in medical oncology in 2015 and was appointed a Consultant Medical Oncologist on the Skin and Urology Units at the Royal Marsden. She became an independent Group Leader at the Francis Crick Institute in 2019, and divides her time between the clinic and her lab.

Professor Turajlic is the Chief Investigator of translational studies into melanoma and kidney cancer, and her research goal is to develop an evolutionary understanding of cancer for patient benefit. She is a member of the NCRI Bladder and Renal Cancer Clinical Studies Group, ESMO Faculty member for genitourinary cancers; and Scientific Committee member for ESMO, Molecular Analysis for Personalised Therapy, Kidney Cancer Association, and EORTC-NCI-AACR Symposia.

Professor Turajlic is a Trustee of the Kidney Cancer Support Network and a Senior Editor at Macmillan Cancer Support. Professor Turajlic receives research funding from Cancer Research UK, the Rosetrees Trust, the Royal Marsden Cancer Charity, and the RMH/ICR Biomedical Research Centre, among others. She was conferred with the title of Professor in October 2022.

Types of Publications

Journal articles

Lim, K.H.J. Spain, L. Barker, C. Georgiou, A. Walls, G. Gore, M. Turajlic, S. Board, R. Larkin, J.M. Lorigan, P (2018) Contemporary outcomes from the use of regular imaging to detect relapse in high-risk cutaneous melanoma.. Show Abstract full text

<h4>Background</h4>Agreement on the utility of imaging follow-up in patients with high-risk melanoma is lacking. A UK consensus statement recommends a surveillance schedule of CT or positron-emission tomography-CT and MRI brain (every 6 months for 3 years, then annually in years 4 and 5) as well as clinical examination for high-risk resected Stages II and III cutaneous melanoma. Our aim was to assess patterns of relapse and whether imaging surveillance could be of clinical benefit.<h4>Patients and methods</h4>A retrospective study of patients enrolled between July 2013 and June 2015 from three UK tertiary cancer centres followed-up according to this protocol was undertaken. We evaluated time-to-recurrence (TTR), recurrence-free survival (RFS), method of detection and characteristics of recurrence, treatment received and overall survival (OS).<h4>Results</h4>A total of 173 patients were included. Most (79%) had treated Stages IIIB and IIIC disease. With a median follow-up of 23.3 months, 82 patients (47%) had relapsed. Median TTR was 10.1 months and median RFS was 21.2 months. The majority of recurrences (66%) were asymptomatic and detected by scheduled surveillance scan. Fifty-six (68%) patients recurred with Stage IV disease, with a median OS of 25.3 months; 26 (31.7%) patients had a locoregional recurrence, median OS not reached (P=0.016). Patients who underwent surgery at recurrence for either Stage III (27%) or IV (18%) disease did not reach their median OS. The median OS for the 33 patients (40%) who received systemic therapy was 12.9 months.<h4>Conclusion</h4>Imaging appears to reliably detect subclinical disease and identify patients suitable for surgery, conferring favourable outcomes. The short median TTR provides rationale to intensify imaging schedule in the first year of surveillance. The poor OS of patients treated with systemic therapy probably reflects the relatively inferior treatment options during this time and requires further evaluation in the current era.

Turajlic, S. Litchfield, K. Xu, H. Rosenthal, R. McGranahan, N. Reading, J.L. Wong, Y.N.S. Rowan, A. Kanu, N. Al Bakir, M. Chambers, T. Salgado, R. Savas, P. Loi, S. Birkbak, N.J. Sansregret, L. Gore, M. Larkin, J. Quezada, S.A. Swanton, C (2017) Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis.. Show Abstract full text

<h4>Background</h4>The focus of tumour-specific antigen analyses has been on single nucleotide variants (SNVs), with the contribution of small insertions and deletions (indels) less well characterised. We investigated whether the frameshift nature of indel mutations, which create novel open reading frames and a large quantity of mutagenic peptides highly distinct from self, might contribute to the immunogenic phenotype.<h4>Methods</h4>We analysed whole-exome sequencing data from 5777 solid tumours, spanning 19 cancer types from The Cancer Genome Atlas. We compared the proportion and number of indels across the cohort, with a subset of results replicated in two independent datasets. We assessed in-silico tumour-specific neoantigen predictions by mutation type with pan-cancer analysis, together with RNAseq profiling in renal clear cell carcinoma cases (n=392), to compare immune gene expression across patient subgroups. Associations between indel burden and treatment response were assessed across four checkpoint inhibitor datasets.<h4>Findings</h4>We observed renal cell carcinomas to have the highest proportion (0·12) and number of indel mutations across the pan-cancer cohort (p<2·2 × 10<sup>-16</sup>), more than double the median proportion of indel mutations in all other cancer types examined. Analysis of tumour-specific neoantigens showed that enrichment of indel mutations for high-affinity binders was three times that of non-synonymous SNV mutations. Furthermore, neoantigens derived from indel mutations were nine times enriched for mutant specific binding, as compared with non-synonymous SNV derived neoantigens. Immune gene expression analysis in the renal clear cell carcinoma cohort showed that the presence of mutant-specific neoantigens was associated with upregulation of antigen presentation genes, which correlated (r=0·78) with T-cell activation as measured by CD8-positive expression. Finally, analysis of checkpoint inhibitor response data revealed frameshift indel count to be significantly associated with checkpoint inhibitor response across three separate melanoma cohorts (p=4·7 × 10<sup>-4</sup>).<h4>Interpretation</h4>Renal cell carcinomas have the highest pan-cancer proportion and number of indel mutations. Evidence suggests indels are a highly immunogenic mutational class, which can trigger an increased abundance of neoantigens and greater mutant-binding specificity.<h4>Funding</h4>Cancer Research UK, UK National Institute for Health Research (NIHR) at the Royal Marsden Hospital National Health Service Foundation Trust, Institute of Cancer Research and University College London Hospitals Biomedical Research Centres, the UK Medical Research Council, the Rosetrees Trust, Novo Nordisk Foundation, the Prostate Cancer Foundation, the Breast Cancer Research Foundation, the European Research Council.

Abbosh, C. Birkbak, N.J. Wilson, G.A. Jamal-Hanjani, M. Constantin, T. Salari, R. Le Quesne, J. Moore, D.A. Veeriah, S. Rosenthal, R. Marafioti, T. Kirkizlar, E. Watkins, T.B.K. McGranahan, N. Ward, S. Martinson, L. Riley, J. Fraioli, F. Al Bakir, M. Grönroos, E. Zambrana, F. Endozo, R. Bi, W.L. Fennessy, F.M. Sponer, N. Johnson, D. Laycock, J. Shafi, S. Czyzewska-Khan, J. Rowan, A. Chambers, T. Matthews, N. Turajlic, S. Hiley, C. Lee, S.M. Forster, M.D. Ahmad, T. Falzon, M. Borg, E. Lawrence, D. Hayward, M. Kolvekar, S. Panagiotopoulos, N. Janes, S.M. Thakrar, R. Ahmed, A. Blackhall, F. Summers, Y. Hafez, D. Naik, A. Ganguly, A. Kareht, S. Shah, R. Joseph, L. Marie Quinn, A. Crosbie, P.A. Naidu, B. Middleton, G. Langman, G. Trotter, S. Nicolson, M. Remmen, H. Kerr, K. Chetty, M. Gomersall, L. Fennell, D.A. Nakas, A. Rathinam, S. Anand, G. Khan, S. Russell, P. Ezhil, V. Ismail, B. Irvin-Sellers, M. Prakash, V. Lester, J.F. Kornaszewska, M. Attanoos, R. Adams, H. Davies, H. Oukrif, D. Akarca, A.U. Hartley, J.A. Lowe, H.L. Lock, S. Iles, N. Bell, H. Ngai, Y. Elgar, G. Szallasi, Z. Schwarz, R.F. Herrero, J. Stewart, A. Quezada, S.A. Peggs, K.S. Van Loo, P. Dive, C. Lin, C.J. Rabinowitz, M. Aerts, H.J.W.L. Hackshaw, A. Shaw, J.A. Zimmermann, B.G. TRACERx consortium, . PEACE consortium, . Swanton, C (2017) Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution.. Show Abstract full text

The early detection of relapse following primary surgery for non-small-cell lung cancer and the characterization of emerging subclones, which seed metastatic sites, might offer new therapeutic approaches for limiting tumour recurrence. The ability to track the evolutionary dynamics of early-stage lung cancer non-invasively in circulating tumour DNA (ctDNA) has not yet been demonstrated. Here we use a tumour-specific phylogenetic approach to profile the ctDNA of the first 100 TRACERx (Tracking Non-Small-Cell Lung Cancer Evolution Through Therapy (Rx)) study participants, including one patient who was also recruited to the PEACE (Posthumous Evaluation of Advanced Cancer Environment) post-mortem study. We identify independent predictors of ctDNA release and analyse the tumour-volume detection limit. Through blinded profiling of postoperative plasma, we observe evidence of adjuvant chemotherapy resistance and identify patients who are very likely to experience recurrence of their lung cancer. Finally, we show that phylogenetic ctDNA profiling tracks the subclonal nature of lung cancer relapse and metastasis, providing a new approach for ctDNA-driven therapeutic studies.

Jamal-Hanjani, M. Wilson, G.A. McGranahan, N. Birkbak, N.J. Watkins, T.B.K. Veeriah, S. Shafi, S. Johnson, D.H. Mitter, R. Rosenthal, R. Salm, M. Horswell, S. Escudero, M. Matthews, N. Rowan, A. Chambers, T. Moore, D.A. Turajlic, S. Xu, H. Lee, S.-.M. Forster, M.D. Ahmad, T. Hiley, C.T. Abbosh, C. Falzon, M. Borg, E. Marafioti, T. Lawrence, D. Hayward, M. Kolvekar, S. Panagiotopoulos, N. Janes, S.M. Thakrar, R. Ahmed, A. Blackhall, F. Summers, Y. Shah, R. Joseph, L. Quinn, A.M. Crosbie, P.A. Naidu, B. Middleton, G. Langman, G. Trotter, S. Nicolson, M. Remmen, H. Kerr, K. Chetty, M. Gomersall, L. Fennell, D.A. Nakas, A. Rathinam, S. Anand, G. Khan, S. Russell, P. Ezhil, V. Ismail, B. Irvin-Sellers, M. Prakash, V. Lester, J.F. Kornaszewska, M. Attanoos, R. Adams, H. Davies, H. Dentro, S. Taniere, P. O'Sullivan, B. Lowe, H.L. Hartley, J.A. Iles, N. Bell, H. Ngai, Y. Shaw, J.A. Herrero, J. Szallasi, Z. Schwarz, R.F. Stewart, A. Quezada, S.A. Le Quesne, J. Van Loo, P. Dive, C. Hackshaw, A. Swanton, C. TRACERx Consortium, (2017) Tracking the Evolution of Non-Small-Cell Lung Cancer.. Show Abstract full text

<h4>Background</h4>Among patients with non-small-cell lung cancer (NSCLC), data on intratumor heterogeneity and cancer genome evolution have been limited to small retrospective cohorts. We wanted to prospectively investigate intratumor heterogeneity in relation to clinical outcome and to determine the clonal nature of driver events and evolutionary processes in early-stage NSCLC.<h4>Methods</h4>In this prospective cohort study, we performed multiregion whole-exome sequencing on 100 early-stage NSCLC tumors that had been resected before systemic therapy. We sequenced and analyzed 327 tumor regions to define evolutionary histories, obtain a census of clonal and subclonal events, and assess the relationship between intratumor heterogeneity and recurrence-free survival.<h4>Results</h4>We observed widespread intratumor heterogeneity for both somatic copy-number alterations and mutations. Driver mutations in EGFR, MET, BRAF, and TP53 were almost always clonal. However, heterogeneous driver alterations that occurred later in evolution were found in more than 75% of the tumors and were common in PIK3CA and NF1 and in genes that are involved in chromatin modification and DNA damage response and repair. Genome doubling and ongoing dynamic chromosomal instability were associated with intratumor heterogeneity and resulted in parallel evolution of driver somatic copy-number alterations, including amplifications in CDK4, FOXA1, and BCL11A. Elevated copy-number heterogeneity was associated with an increased risk of recurrence or death (hazard ratio, 4.9; P=4.4×10<sup>-4</sup>), which remained significant in multivariate analysis.<h4>Conclusions</h4>Intratumor heterogeneity mediated through chromosome instability was associated with an increased risk of recurrence or death, a finding that supports the potential value of chromosome instability as a prognostic predictor. (Funded by Cancer Research UK and others; TRACERx ClinicalTrials.gov number, NCT01888601 .).

Spain, L. Walls, G. Julve, M. O'Meara, K. Schmid, T. Kalaitzaki, E. Turajlic, S. Gore, M. Rees, J. Larkin, J (2017) Neurotoxicity from immune-checkpoint inhibition in the treatment of melanoma: a single centre experience and review of the literature.. Show Abstract full text

<h4>Background</h4>Treatment with immune checkpoint inhibitors (ICPi) has greatly improved survival for patients with advanced melanoma in recent years. Anti-CTLA-4 and anti-PD1 antibodies have been approved following large Phase III trials. Immune-related neurological toxicity of varying severity has been reported in the literature. The cumulative incidence of neurotoxicity among ipilimumab, nivolumab and pembrolizumab is reported as <1% in published clinical trials. We aimed to identify the incidence of neurotoxicity in our institution across anti-CTLA4 and anti-PD-1 antibodies, including the combination of ipilimumab with nivolumab. We also review the existing literature and propose an investigation and management algorithm.<h4>Methods</h4>All patients with advanced melanoma treated with ipilimumab, nivolumab, pembrolizumab or the combination of ipilimumab and nivolumab (ipi + nivo), managed at the Royal Marsden Hospital between September 2010 and December 2015, including patients on (published) clinical trials were included. Medical records for each patient were reviewed and information on neurotoxicity recorded. A systematic search strategy was performed to collate existing reports of neurological toxicity.<h4>Results</h4>In total, 413 immunotherapy treatment episodes in 352 patients were included, with median follow-up of 26.7 months. Ten cases of neurotoxicity were recorded, affecting 2.8% of patients overall, ranging from grade 1 to 4, affecting both central and peripheral nervous systems. A rate of 14% was noted with ipi + nivo. Three of five patients commenced on corticosteroids responded to these. Six patients had made a full recovery at the time of reporting. A favorable radiological response was found in 7 of the 10 cases. Unusual presentations are described in detail.<h4>Conclusions</h4>Neurological toxicity is not uncommon, and may be more frequent in patients treated with combination ipi + nivo. Patterns of presentation and response to treatment are varied. A prompt and considered approach is required to optimize outcomes in this group of patients.

Arce Vargas, F. Furness, A.J.S. Solomon, I. Joshi, K. Mekkaoui, L. Lesko, M.H. Miranda Rota, E. Dahan, R. Georgiou, A. Sledzinska, A. Ben Aissa, A. Franz, D. Werner Sunderland, M. Wong, Y.N.S. Henry, J.Y. O'Brien, T. Nicol, D. Challacombe, B. Beers, S.A. Melanoma TRACERx Consortium, . Renal TRACERx Consortium, . Lung TRACERx Consortium, . Turajlic, S. Gore, M. Larkin, J. Swanton, C. Chester, K.A. Pule, M. Ravetch, J.V. Marafioti, T. Peggs, K.S. Quezada, S.A (2017) Fc-Optimized Anti-CD25 Depletes Tumor-Infiltrating Regulatory T Cells and Synergizes with PD-1 Blockade to Eradicate Established Tumors.. Show Abstract full text

CD25 is expressed at high levels on regulatory T (Treg) cells and was initially proposed as a target for cancer immunotherapy. However, anti-CD25 antibodies have displayed limited activity against established tumors. We demonstrated that CD25 expression is largely restricted to tumor-infiltrating Treg cells in mice and humans. While existing anti-CD25 antibodies were observed to deplete Treg cells in the periphery, upregulation of the inhibitory Fc gamma receptor (FcγR) IIb at the tumor site prevented intra-tumoral Treg cell depletion, which may underlie the lack of anti-tumor activity previously observed in pre-clinical models. Use of an anti-CD25 antibody with enhanced binding to activating FcγRs led to effective depletion of tumor-infiltrating Treg cells, increased effector to Treg cell ratios, and improved control of established tumors. Combination with anti-programmed cell death protein-1 antibodies promoted complete tumor rejection, demonstrating the relevance of CD25 as a therapeutic target and promising substrate for future combination approaches in immune-oncology.

Spain, L. Walls, G. Messiou, C. Turajlic, S. Gore, M. Larkin, J (2017) Efficacy and toxicity of rechallenge with combination immune checkpoint blockade in metastatic melanoma: a case series.. Show Abstract full text

<h4>Background</h4>The efficacy and potential toxicity of rechallenge with combination ipilimumab and nivolumab has not been described. Retreatment of patients with immune checkpoint inhibitors in the setting of prior significant toxicity lacks evidence-based guidance.<h4>Methods</h4>We present the first three, consecutive patients who received re-treatment with combination ipilimumab and nivolumab for metastatic melanoma managed at our institution.<h4>Results</h4>Rechallenge with combination ipilimumab and nivolumab in the setting of prior grade 3 toxicity with initial combination therapy is feasible, and responses are seen. We highlight the fact that grade 3 toxicity is likely to recur, but if so, can be manageable.<h4>Conclusions</h4>Retreatment with ipi + nivo may be considered an option in carefully selected, well-informed patients. More research is required to delineate the benefits and risks with this approach.

Turajlic, S. Larkin, J (2016) Systemic treatment of advanced papillary renal cell carcinoma: Where next?.
Spain, L. Higgins, R. Gopalakrishnan, K. Turajlic, S. Gore, M. Larkin, J (2016) Acute renal allograft rejection after immune checkpoint inhibitor therapy for metastatic melanoma.. Show Abstract full text

Immune checkpoint inhibitors such as ipilimumab and nivolumab improve survival in patients with advanced melanoma and are increasingly available to clinicians for use in the clinic. Their safety in organ transplant recipients is not well defined but published case reports describing treatment with ipilimumab have not been complicated by graft rejection. No cases of anti-programmed cell death protein 1 administration are reported in this group. We describe a case of acute graft rejection in a kidney transplant recipient after treatment with nivolumab, after progression on ipilimumab. Potential factors increasing the risk of graft rejection in this case are discussed, in particular the contribution of nivolumab.

Turajlic, S. Larkin, J. Swanton, C (2015) SnapShot: Renal Cell Carcinoma.. Show Abstract full text

This SnapShot summarizes current knowledge about the key features in mutational landscape, major pathways, and tumor evolution and heterogeneity in renal cell carcinoma, as well as the most recent advances in therapeutic development. To view this SnapShot, open or download the PDF.

Gulati, S. Turajlic, S. Larkin, J. Bates, P.A. Swanton, C (2015) Relapse models for clear cell renal carcinoma..
Turajlic, S. Larkin, J. Swanton, C (2015) Academically led clinical trials: challenges and opportunities..
Soultati, A. Stares, M. Swanton, C. Larkin, J. Turajlic, S (2015) How should clinicians address intratumour heterogeneity in clear cell renal cell carcinoma?. Show Abstract full text

<h4>Purpose of review</h4>Despite the availability of multiple targeted therapies, the 5-year survival rate of patients with metastatic clear cell renal cell carcinoma (ccRCC) rarely exceeds 10%. Recent insights into the mutational landscape and evolutionary dynamics of ccRCC have offered up a plausible explanation for these outcomes. The purpose of this review is to link the research findings to potential changes in clinical practice.<h4>Recent findings</h4>Intratumour heterogeneity (ITH) dominates the evolutionary landscape in ccRCC at the genetic, transcriptomic and proteomic level. Spatial and temporal separation of tumour subclones within the primary tumour as well as between primary and metastatic sites has been demonstrated at single nucleotide resolution. In the cases analysed to date, approximately two-thirds of somatic mutations are not shared between multiple biopsies from the same primary tumour. Very few of the key disease-driving events are shared across all primary tumour regions (with the exception of VHL and loss of chromosome 3p), whereas the majority are restricted to one or more tumour regions (TP53, SETD2, BAP1, PTEN, mTOR, PIK3CA and KDM5C).<h4>Summary</h4>ITH must be considered in the management of ccRCC with respect to diagnostic procedures, prognostic and predictive biomarkers and drug development.

Furney, S.J. Turajlic, S. Stamp, G. Thomas, J.M. Hayes, A. Strauss, D. Gavrielides, M. Xing, W. Gore, M. Larkin, J. Marais, R (2014) The mutational burden of acral melanoma revealed by whole-genome sequencing and comparative analysis.. Show Abstract full text

Acral melanoma is a subtype of melanoma with distinct epidemiological, clinical and mutational profiles. To define the genomic alterations in acral melanoma, we conducted whole-genome sequencing and SNP array analysis of five metastatic tumours and their matched normal genomes. We identified the somatic mutations, copy number alterations and structural variants in these tumours and combined our data with published studies to identify recurrently mutated genes likely to be the drivers of acral melanomagenesis. We compared and contrasted the genomic landscapes of acral, mucosal, uveal and common cutaneous melanoma to reveal the distinctive mutational characteristics of each subtype.

Sanchez-Laorden, B. Viros, A. Girotti, M.R. Pedersen, M. Saturno, G. Zambon, A. Niculescu-Duvaz, D. Turajlic, S. Hayes, A. Gore, M. Larkin, J. Lorigan, P. Cook, M. Springer, C. Marais, R (2014) BRAF inhibitors induce metastasis in RAS mutant or inhibitor-resistant melanoma cells by reactivating MEK and ERK signaling.. Show Abstract full text

Melanoma is a highly metastatic and lethal form of skin cancer. The protein kinase BRAF is mutated in about 40% of melanomas, and BRAF inhibitors improve progression-free and overall survival in these patients. However, after a relatively short period of disease control, most patients develop resistance because of reactivation of the RAF-ERK (extracellular signal-regulated kinase) pathway, mediated in many cases by mutations in RAS. We found that BRAF inhibition induces invasion and metastasis in RAS mutant melanoma cells through a mechanism mediated by the reactivation of the MEK (mitogen-activated protein kinase kinase)-ERK pathway, increased expression and secretion of interleukin 8, and induction of protease-dependent invasion. These events were accompanied by a cell morphology switch from predominantly rounded to predominantly elongated cells. We also observed similar responses in BRAF inhibitor-resistant melanoma cells. These data show that BRAF inhibitors can induce melanoma cell invasion and metastasis in tumors that develop resistance to these drugs.

Turajlic, S. Furney, S.J. Stamp, G. Rana, S. Ricken, G. Oduko, Y. Saturno, G. Springer, C. Hayes, A. Gore, M. Larkin, J. Marais, R (2014) Whole-genome sequencing reveals complex mechanisms of intrinsic resistance to BRAF inhibition.. Show Abstract full text

<h4>Background</h4>BRAF is mutated in ∼42% of human melanomas (COSMIC. http://www.sanger.ac.uk/genetics/CGP/cosmic/) and pharmacological BRAF inhibitors such as vemurafenib and dabrafenib achieve dramatic responses in patients whose tumours harbour BRAF(V600) mutations. Objective responses occur in ∼50% of patients and disease stabilisation in a further ∼30%, but ∼20% of patients present primary or innate resistance and do not respond. Here, we investigated the underlying cause of treatment failure in a patient with BRAF mutant melanoma who presented primary resistance.<h4>Methods</h4>We carried out whole-genome sequencing and single nucleotide polymorphism (SNP) array analysis of five metastatic tumours from the patient. We validated mechanisms of resistance in a cell line derived from the patient's tumour.<h4>Results</h4>We observed that the majority of the single-nucleotide variants identified were shared across all tumour sites, but also saw site-specific copy-number alterations in discrete cell populations at different sites. We found that two ubiquitous mutations mediated resistance to BRAF inhibition in these tumours. A mutation in GNAQ sustained mitogen-activated protein kinase (MAPK) signalling, whereas a mutation in PTEN activated the PI3 K/AKT pathway. Inhibition of both pathways synergised to block the growth of the cells.<h4>Conclusions</h4>Our analyses show that the five metastases arose from a common progenitor and acquired additional alterations after disease dissemination. We demonstrate that a distinct combination of mutations mediated primary resistance to BRAF inhibition in this patient. These mutations were present in all five tumours and in a tumour sample taken before BRAF inhibitor treatment was administered. Inhibition of both pathways was required to block tumour cell growth, suggesting that combined targeting of these pathways could have been a valid therapeutic approach for this patient.

Furney, S.J. Pedersen, M. Gentien, D. Dumont, A.G. Rapinat, A. Desjardins, L. Turajlic, S. Piperno-Neumann, S. de la Grange, P. Roman-Roman, S. Stern, M.-.H. Marais, R (2013) SF3B1 mutations are associated with alternative splicing in uveal melanoma.. Show Abstract full text

<h4>Unlabelled</h4>Uveal melanoma, the most common eye malignancy, causes severe visual morbidity and is fatal in approximately 50% of patients. Primary uveal melanoma can be cured by surgery or radiotherapy, but the metastatic disease is treatment refractory. To understand comprehensively uveal melanoma genetics, we conducted single-nucleotide polymorphism arrays and whole-genome sequencing on 12 primary uveal melanomas. We observed only approximately 2,000 predicted somatic single-nucleotide variants per tumor and low levels of aneuploidy. We did not observe an ultraviolet radiation DNA damage signature, but identified SF3B1 mutations in three samples and a further 15 mutations in an extension cohort of 105 samples. SF3B1 mutations were associated with good prognosis and were rarely coincident with BAP1 mutations. SF3B1 encodes a component of the spliceosome, and RNA sequencing revealed that SF3B1 mutations were associated with differential alternative splicing of protein coding genes, including ABCC5 and UQCC, and of the long noncoding RNA CRNDE.<h4>Significance</h4>Our data show that despite its dismal prognosis, uveal melanoma is a relatively simple genetic disease characterized by recurrent chromosomal losses and gains and a low mutational burden. We show that SF3B1 is recurrently mutated in uveal melanoma, and the mutations are associated with aberrant alternative splicing.

Turajlic, S. Ali, Z. Yousaf, N. Larkin, J (2013) Phase I/II RAF kinase inhibitors in cancer therapy.. Show Abstract full text

<h4>Introduction</h4>Aberrant activation of RAF signalling is a frequent finding in human cancers. BRAF is the only RAF family member that is commonly mutated, whilst CRAF and ARAF play important roles in the signal transduction from mutant RAS. BRAF-specific inhibitors have been more effective in the treatment of BRAF-mutant melanoma than BRAF-mutant thyroid and colorectal cancers.<h4>Areas covered</h4>The review summarises the experience with RAF kinase inhibitors, including efficacy, modes of acquired resistance, and the mechanism behind the progression of pre-malignant RAS-mutant lesions observed with RAF kinase inhibitors. The authors review all the completed and ongoing Phase I or II clinical trials of RAF kinase inhibitors and discuss in detail the rationale behind the combinatorial approaches.<h4>Expert opinion</h4>The success of RAF kinase inhibitors has demonstrated the necessity of genotype-driven treatment selection for cancer patients. The spectrum of responses in different tumour types is explained by feedback events that are determined by cell lineage. Dissection of these events and the mechanisms of acquired resistance will determine the appropriate combination therapies. Ongoing characterisation of RAS-MAPK regulation in malignant cells may aid the development of novel agents that have greater potency for the inhibition of activated RAF kinase, and lesser propensity for promotion of RAS-mutant tumours.

Furney, S.J. Turajlic, S. Stamp, G. Nohadani, M. Carlisle, A. Thomas, J.M. Hayes, A. Strauss, D. Gore, M. van den Oord, J. Larkin, J. Marais, R (2013) Genome sequencing of mucosal melanomas reveals that they are driven by distinct mechanisms from cutaneous melanoma.. Show Abstract full text

Mucosal melanoma displays distinct clinical and epidemiological features compared to cutaneous melanoma. Here we used whole genome and whole exome sequencing to characterize the somatic alterations and mutation spectra in the genomes of ten mucosal melanomas. We observed somatic mutation rates that are considerably lower than occur in sun-exposed cutaneous melanoma, but comparable to the rates seen in cancers not associated with exposure to known mutagens. In particular, the mutation signatures are not indicative of ultraviolet light- or tobacco smoke-induced DNA damage. Genes previously reported as mutated in other cancers were also mutated in mucosal melanoma. Notably, there were substantially more copy number and structural variations in mucosal melanoma than have been reported in cutaneous melanoma. Thus, mucosal and cutaneous melanomas are distinct diseases with discrete genetic features. Our data suggest that different mechanisms underlie the genesis of these diseases and that structural variations play a more important role in mucosal than in cutaneous melanomagenesis.

Khattak, M. Fisher, R. Turajlic, S. Larkin, J (2013) Targeted therapy and immunotherapy in advanced melanoma: an evolving paradigm.. Show Abstract full text

Metastatic melanoma is one of the most challenging malignancies to treat and often has a poor outcome. Until recently, systemic treatment options were limited, with poor response rates and no survival advantage. However, the treatment of metastatic melanoma has been revolutionized by developments in targeted therapy and immunotherapy; the BRAF inhibitor, vemurafenib, and anticytotoxic T-lymphocyte antigen 4 antibody, ipilimumab, are the first agents to demonstrate a survival benefit. Despite the success of these treatments, most patients eventually progress, and research into response and resistance mechanisms, rationally designed combination therapies and evaluation of the role of these agents in the adjuvant setting is critically important.

Girotti, M.R. Pedersen, M. Sanchez-Laorden, B. Viros, A. Turajlic, S. Niculescu-Duvaz, D. Zambon, A. Sinclair, J. Hayes, A. Gore, M. Lorigan, P. Springer, C. Larkin, J. Jorgensen, C. Marais, R (2013) Inhibiting EGF receptor or SRC family kinase signaling overcomes BRAF inhibitor resistance in melanoma.. Show Abstract full text

<h4>Unlabelled</h4>We generated cell lines resistant to BRAF inhibitors and show that the EGF receptor (EGFR)-SRC family kinase (SFK)-STAT3 signaling pathway was upregulated in these cells. In addition to driving proliferation of resistant cells, this pathway also stimulated invasion and metastasis. EGFR inhibitors cooperated with BRAF inhibitors to block the growth of the resistant cells in vitro and in vivo, and monotherapy with the broad specificity tyrosine kinase inhibitor dasatinib blocked growth and metastasis in vivo. We analyzed tumors from patients with intrinsic or acquired resistance to vemurafenib and observed increased EGFR and SFK activity. Furthermore, dasatinib blocked the growth and metastasis of one of the resistant tumors in immunocompromised mice. Our data show that BRAF inhibitor-mediated activation of EGFR-SFK-STAT3 signaling can mediate resistance in patients with BRAF-mutant melanoma. We describe 2 treatments that seem to overcome this resistance and could deliver therapeutic efficacy in patients with drug-resistant BRAF-mutant melanoma.<h4>Significance</h4>Therapies that target the driver oncogenes in cancer can achieve remarkable responses if patients are stratified for treatment. However, as with conventional therapies, patients often develop acquired resistance to targeted therapies, and a proportion of patients are intrinsically resistant and fail to respond despite the presence of an appropriate driver oncogene mutation. We found that the EGFR/SFK pathway mediated resistance to vemurafenib in BRAF -mutant melanoma and that BRAF and EGFR or SFK inhibition blocked proliferation and invasion of these resistant tumors, providing potentially effective therapeutic options for these patients.

Furney, S.J. Turajlic, S. Fenwick, K. Lambros, M.B. MacKay, A. Ricken, G. Mitsopoulos, C. Kozarewa, I. Hakas, J. Zvelebil, M. Lord, C.J. Ashworth, A. Reis-Filho, J.S. Herlyn, M. Murata, H. Marais, R (2012) Genomic characterisation of acral melanoma cell lines.. Show Abstract full text

Acral melanoma is a rare melanoma subtype with distinct epidemiological, clinical and genetic features. To determine if acral melanoma cell lines are representative of this melanoma subtype, six lines were analysed by whole-exome sequencing and array comparative genomic hybridisation. We demonstrate that the cell lines display a mutation rate that is comparable to that of published primary and metastatic acral melanomas and observe a mutational signature suggestive of UV-induced mutagenesis in two of the cell lines. Mutations were identified in oncogenes and tumour suppressors previously linked to melanoma including BRAF, NRAS, KIT, PTEN and TP53, in cancer genes not previously linked to melanoma and in genes linked to DNA repair such as BRCA1 and BRCA2. Our findings provide strong circumstantial evidence to suggest that acral melanoma cell lines and acral tumours share genetic features in common and that these cells are therefore valuable tools to investigate the biology of this aggressive melanoma subtype. Data are available at: http://rock.icr.ac.uk/collaborations/Furney_et_al_2012/.

Turajlic, S. Furney, S.J. Lambros, M.B. Mitsopoulos, C. Kozarewa, I. Geyer, F.C. Mackay, A. Hakas, J. Zvelebil, M. Lord, C.J. Ashworth, A. Thomas, M. Stamp, G. Larkin, J. Reis-Filho, J.S. Marais, R (2012) Whole genome sequencing of matched primary and metastatic acral melanomas.. Show Abstract full text

Next generation sequencing has enabled systematic discovery of mutational spectra in cancer samples. Here, we used whole genome sequencing to characterize somatic mutations and structural variation in a primary acral melanoma and its lymph node metastasis. Our data show that the somatic mutational rates in this acral melanoma sample pair were more comparable to the rates reported in cancer genomes not associated with mutagenic exposure than in the genome of a melanoma cell line or the transcriptome of melanoma short-term cultures. Despite the perception that acral skin is sun-protected, the dominant mutational signature in these samples is compatible with damage due to ultraviolet light exposure. A nonsense mutation in ERCC5 discovered in both the primary and metastatic tumors could also have contributed to the mutational signature through accumulation of unrepaired dipyrimidine lesions. However, evidence of transcription-coupled repair was suggested by the lower mutational rate in the transcribed regions and expressed genes. The primary and the metastasis are highly similar at the level of global gene copy number alterations, loss of heterozygosity and single nucleotide variation (SNV). Furthermore, the majority of the SNVs in the primary tumor were propagated in the metastasis and one nonsynonymous coding SNV and one splice site mutation appeared to arise de novo in the metastatic lesion.

Natrajan, R. Mackay, A. Lambros, M.B. Weigelt, B. Wilkerson, P.M. Manie, E. Grigoriadis, A. A'Hern, R. van der Groep, P. Kozarewa, I. Popova, T. Mariani, O. Turaljic, S. Furney, S.J. Marais, R. Rodruigues, D.-.N. Flora, A.C. Wai, P. Pawar, V. McDade, S. Carroll, J. Stoppa-Lyonnet, D. Green, A.R. Ellis, I.O. Swanton, C. van Diest, P. Delattre, O. Lord, C.J. Foulkes, W.D. Vincent-Salomon, A. Ashworth, A. Stern, M.H. Reis-Filho, J.S (2012) A whole-genome massively parallel sequencing analysis of BRCA1 mutant oestrogen receptor-negative and -positive breast cancers.. Show Abstract full text

BRCA1 encodes a tumour suppressor protein that plays pivotal roles in homologous recombination (HR) DNA repair, cell-cycle checkpoints, and transcriptional regulation. BRCA1 germline mutations confer a high risk of early-onset breast and ovarian cancer. In more than 80% of cases, tumours arising in BRCA1 germline mutation carriers are oestrogen receptor (ER)-negative; however, up to 15% are ER-positive. It has been suggested that BRCA1 ER-positive breast cancers constitute sporadic cancers arising in the context of a BRCA1 germline mutation rather than being causally related to BRCA1 loss-of-function. Whole-genome massively parallel sequencing of ER-positive and ER-negative BRCA1 breast cancers, and their respective germline DNAs, was used to characterize the genetic landscape of BRCA1 cancers at base-pair resolution. Only BRCA1 germline mutations, somatic loss of the wild-type allele, and TP53 somatic mutations were recurrently found in the index cases. BRCA1 breast cancers displayed a mutational signature consistent with that caused by lack of HR DNA repair in both ER-positive and ER-negative cases. Sequencing analysis of independent cohorts of hereditary BRCA1 and sporadic non-BRCA1 breast cancers for the presence of recurrent pathogenic mutations and/or homozygous deletions found in the index cases revealed that DAPK3, TMEM135, KIAA1797, PDE4D, and GATA4 are potential additional drivers of breast cancers. This study demonstrates that BRCA1 pathogenic germline mutations coupled with somatic loss of the wild-type allele are not sufficient for hereditary breast cancers to display an ER-negative phenotype, and has led to the identification of three potential novel breast cancer genes (ie DAPK3, TMEM135, and GATA4).

Litchfield, K. Turajlic, S. Swanton, C (2017) The GENIE Is Out of the Bottle: Landmark Cancer Genomics Dataset Released.. Show Abstract full text

<b/> In this issue of <i>Cancer Discovery</i>, an overview of the AACR Project GENIE, a landmark study in cancer genomics, is presented by The AACR Project GENIE Consortium. A summary of the goals and objectives of this ambitious program is provided, together with an analysis of the phase I cohort of 19,000 samples. <i>Cancer Discov; 7(8); 796-8. ©2017 AACR.</i><i>See related article by The AACR Project GENIE Consortium, p. 818</i>.

Turajlic, S. Swanton, C (2017) Implications of cancer evolution for drug development..
Turajlic, S. Swanton, C (2016) Metastasis as an evolutionary process.. Show Abstract full text

Therapeutic advances in oncology have not fully translated to the treatment of metastatic disease, which remains largely incurable. Metastatic subclones can emerge both early and late in the life of the primary tumor. A better understanding of the genetic evolution of metastatic disease has the potential to reveal differences in the therapeutic vulnerabilities of primary and metastatic tumors, shed light on the temporal patterns of and routes to metastatic colonization, and provide insight into the biology of the metastatic process. Here we review recent comparative studies of primary and metastatic tumors, including data suggesting that macroevolutionary shifts (the onset of chromosomal instability) contribute to the evolution of metastatic disease. We also discuss the practical challenges associated with these studies and how they might be overcome.

Turajlic, S. Swanton, C (2015) Gastrointestinal cancer: Tracking tumour evolution through liquid biopsy..
Turajlic, S. McGranahan, N. Swanton, C (2015) Inferring mutational timing and reconstructing tumour evolutionary histories.. Show Abstract full text

Cancer evolution can be considered within a Darwinian framework. Both micro and macro-evolutionary theories can be applied to understand tumour progression and treatment failure. Owing to cancers' complexity and heterogeneity the rules of tumour evolution, such as the role of selection, remain incompletely understood. The timing of mutational events during tumour evolution presents diagnostic, prognostic and therapeutic opportunities. Here we review the current sampling and computational approaches for inferring mutational timing and the evidence from next generation sequencing-informed data on mutational timing across all tumour types. We discuss how this knowledge can be used to illuminate the genes and pathways that drive cancer initiation and relapse; and to support drug development and clinical trial design.

Perez, A. Turajlic, S. Szyszko, T. O'Doherty, M. Calonje, E. Harries, M. Acland, K (2010) Generalized melanosis and melanuria in a patient with metastatic melanoma..
TRACERx Renal consortium, (2017) TRACERx Renal: tracking renal cancer evolution through therapy..
Larkin, J.M. Turajlic, S. Nathan, P.D. Lorigan, P. Stamp, G. Gonzalez de Castro, D. Martin, N. Griffiths, J. Edmonds, K. Sarker, S. James, M.G. A'Hern, R. Coombes, G. Snowdon, C. Bliss, J.M. Gore, M.E. Marais, R (2011) A phase II trial of nilotinib in the treatment of patients with KIT mutated advanced acral and mucosal melanoma (NICAM).. Show Abstract full text

TPS229 Background: Recent studies have indicated that KIT mutations occur in approximately 20% of acral and mucosal melanomas (Curtin, al. 2006; Beadling, et al. 2008). Several case reports have shown that KIT mutated melanomas can respond to treatment with KIT inhibitors (Hodi et al. 2008; Lutzky et al. 2008; Quintas-Cardama et al. 2008; Handolias et al. 2010; Itoh, et al. 2010; Satzger et al. 2010; Terheyden et al. 2010; Woodman and Davies 2010) suggesting that such tumours critically depend on upregulated KIT signalling. Nilotinib, which potently inhibits the tyrosine kinase activity of BCR-ABL and KIT, has been evaluated in patients with advanced KIT mutated GIST and has shown clinical efficacy at well tolerated doses (Demetri et al. 2009; Montemurro et al. 2009; Schlemmer et al. 2010). METHODS: NICAM is a single arm 2-stage open label phase II study of nilotinib in KIT mutated advanced mucosal or acral melanoma. Patients with KIT mutations known to confer resistance to nilotinib are excluded. The primary objective in this feasibility study is to evaluate the efficacy of nilotinib in KIT mutated advanced mucosal and acral melanoma with a primary endpoint of progression free survival at 6 months. Secondary endpoints include response rate at 12 weeks; overall survival; toxicity of treatment; correlation between the response to nilotinib and KIT genotype, KIT gene copy number and KIT expression. In addition, the following will be explored: 1. Suppression of phosphorylation of KIT and downstream pathways on day 15 tumour biopsies; 2. Possible mechanisms of resistance by analysis of biopsies obtained at disease progression; 3. The use of circulating tumour DNA for non-invasive KIT mutational testing; 4. Genetic lesions that potentially cooperate with oncogenic KIT in melanoma (using Next Generation Sequencing). All patients are treated until disease progression or unacceptable toxicity. If 2 or more of the first 9 patients are progression free at 6 months, patient entry will continue into the second stage until a total of 24 patients have been recruited. To date, 45 patients have been registered and screened for KIT mutation and 5 patients have entered the study.

Arce Vargas, F. Furness, A.J.S. Litchfield, K. Joshi, K. Rosenthal, R. Ghorani, E. Solomon, I. Lesko, M.H. Ruef, N. Roddie, C. Henry, J.Y. Spain, L. Ben Aissa, A. Georgiou, A. Wong, Y.N.S. Smith, M. Strauss, D. Hayes, A. Nicol, D. O'Brien, T. Mårtensson, L. Ljungars, A. Teige, I. Frendéus, B. TRACERx Melanoma, . TRACERx Renal, . TRACERx Lung consortia, . Pule, M. Marafioti, T. Gore, M. Larkin, J. Turajlic, S. Swanton, C. Peggs, K.S. Quezada, S.A (2018) Fc Effector Function Contributes to the Activity of Human Anti-CTLA-4 Antibodies.. Show Abstract full text

With the use of a mouse model expressing human Fc-gamma receptors (FcγRs), we demonstrated that antibodies with isotypes equivalent to ipilimumab and tremelimumab mediate intra-tumoral regulatory T (Treg) cell depletion in vivo, increasing the CD8<sup>+</sup> to Treg cell ratio and promoting tumor rejection. Antibodies with improved FcγR binding profiles drove superior anti-tumor responses and survival. In patients with advanced melanoma, response to ipilimumab was associated with the CD16a-V158F high affinity polymorphism. Such activity only appeared relevant in the context of inflamed tumors, explaining the modest response rates observed in the clinical setting. Our data suggest that the activity of anti-CTLA-4 in inflamed tumors may be improved through enhancement of FcγR binding, whereas poorly infiltrated tumors will likely require combination approaches.

Turajlic, S. Xu, H. Litchfield, K. Rowan, A. Horswell, S. Chambers, T. O'Brien, T. Lopez, J.I. Watkins, T.B.K. Nicol, D. Stares, M. Challacombe, B. Hazell, S. Chandra, A. Mitchell, T.J. Au, L. Eichler-Jonsson, C. Jabbar, F. Soultati, A. Chowdhury, S. Rudman, S. Lynch, J. Fernando, A. Stamp, G. Nye, E. Stewart, A. Xing, W. Smith, J.C. Escudero, M. Huffman, A. Matthews, N. Elgar, G. Phillimore, B. Costa, M. Begum, S. Ward, S. Salm, M. Boeing, S. Fisher, R. Spain, L. Navas, C. Grönroos, E. Hobor, S. Sharma, S. Aurangzeb, I. Lall, S. Polson, A. Varia, M. Horsfield, C. Fotiadis, N. Pickering, L. Schwarz, R.F. Silva, B. Herrero, J. Luscombe, N.M. Jamal-Hanjani, M. Rosenthal, R. Birkbak, N.J. Wilson, G.A. Pipek, O. Ribli, D. Krzystanek, M. Csabai, I. Szallasi, Z. Gore, M. McGranahan, N. Van Loo, P. Campbell, P. Larkin, J. Swanton, C. TRACERx Renal Consortium, (2018) Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal.. Show Abstract full text

The evolutionary features of clear-cell renal cell carcinoma (ccRCC) have not been systematically studied to date. We analyzed 1,206 primary tumor regions from 101 patients recruited into the multi-center prospective study, TRACERx Renal. We observe up to 30 driver events per tumor and show that subclonal diversification is associated with known prognostic parameters. By resolving the patterns of driver event ordering, co-occurrence, and mutual exclusivity at clone level, we show the deterministic nature of clonal evolution. ccRCC can be grouped into seven evolutionary subtypes, ranging from tumors characterized by early fixation of multiple mutational and copy number drivers and rapid metastases to highly branched tumors with >10 subclonal drivers and extensive parallel evolution associated with attenuated progression. We identify genetic diversity and chromosomal complexity as determinants of patient outcome. Our insights reconcile the variable clinical behavior of ccRCC and suggest evolutionary potential as a biomarker for both intervention and surveillance.

Turajlic, S. Xu, H. Litchfield, K. Rowan, A. Chambers, T. Lopez, J.I. Nicol, D. O'Brien, T. Larkin, J. Horswell, S. Stares, M. Au, L. Jamal-Hanjani, M. Challacombe, B. Chandra, A. Hazell, S. Eichler-Jonsson, C. Soultati, A. Chowdhury, S. Rudman, S. Lynch, J. Fernando, A. Stamp, G. Nye, E. Jabbar, F. Spain, L. Lall, S. Guarch, R. Falzon, M. Proctor, I. Pickering, L. Gore, M. Watkins, T.B.K. Ward, S. Stewart, A. DiNatale, R. Becerra, M.F. Reznik, E. Hsieh, J.J. Richmond, T.A. Mayhew, G.F. Hill, S.M. McNally, C.D. Jones, C. Rosenbaum, H. Stanislaw, S. Burgess, D.L. Alexander, N.R. Swanton, C. PEACE, . TRACERx Renal Consortium, (2018) Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal.. Show Abstract full text

Clear-cell renal cell carcinoma (ccRCC) exhibits a broad range of metastatic phenotypes that have not been systematically studied to date. Here, we analyzed 575 primary and 335 metastatic biopsies across 100 patients with metastatic ccRCC, including two cases sampledat post-mortem. Metastatic competence was afforded by chromosome complexity, and we identify 9p loss as a highly selected event driving metastasis and ccRCC-related mortality (p = 0.0014). Distinct patterns of metastatic dissemination were observed, including rapid progression to multiple tissue sites seeded by primary tumors of monoclonal structure. By contrast, we observed attenuated progression in cases characterized by high primary tumor heterogeneity, with metastatic competence acquired gradually and initial progression to solitary metastasis. Finally, we observed early divergence of primitive ancestral clones and protracted latency of up to two decades as a feature of pancreatic metastases.

Mitchell, T.J. Turajlic, S. Rowan, A. Nicol, D. Farmery, J.H.R. O'Brien, T. Martincorena, I. Tarpey, P. Angelopoulos, N. Yates, L.R. Butler, A.P. Raine, K. Stewart, G.D. Challacombe, B. Fernando, A. Lopez, J.I. Hazell, S. Chandra, A. Chowdhury, S. Rudman, S. Soultati, A. Stamp, G. Fotiadis, N. Pickering, L. Au, L. Spain, L. Lynch, J. Stares, M. Teague, J. Maura, F. Wedge, D.C. Horswell, S. Chambers, T. Litchfield, K. Xu, H. Stewart, A. Elaidi, R. Oudard, S. McGranahan, N. Csabai, I. Gore, M. Futreal, P.A. Larkin, J. Lynch, A.G. Szallasi, Z. Swanton, C. Campbell, P.J. TRACERx Renal Consortium, (2018) Timing the Landmark Events in the Evolution of Clear Cell Renal Cell Cancer: TRACERx Renal.. Show Abstract full text

Clear cell renal cell carcinoma (ccRCC) is characterized by near-universal loss of the short arm of chromosome 3, deleting several tumor suppressor genes. We analyzed whole genomes from 95 biopsies across 33 patients with clear cell renal cell carcinoma. We find hotspots of point mutations in the 5' UTR of TERT, targeting a MYC-MAX-MAD1 repressor associated with telomere lengthening. The most common structural abnormality generates simultaneous 3p loss and 5q gain (36% patients), typically through chromothripsis. This event occurs in childhood or adolescence, generally as the initiating event that precedes emergence of the tumor's most recent common ancestor by years to decades. Similar genomic changes drive inherited ccRCC. Modeling differences in age incidence between inherited and sporadic cancers suggests that the number of cells with 3p loss capable of initiating sporadic tumors is no more than a few hundred. Early development of ccRCC follows well-defined evolutionary trajectories, offering opportunity for early intervention.

Flynn, M. Pickering, L. Larkin, J. Turajlic, S (2018) Immune-checkpoint inhibitors in melanoma and kidney cancer: from sequencing to rational selection.. Show Abstract full text

Immune-checkpoint inhibitors (ICPIs), including antibodies against cytotoxic T-lymphocyte associated antigen 4 and programmed cell death protein 1, have been shown to induce durable complete responses in a proportion of patients in the first-line and refractory setting in advanced melanoma and renal cell carcinoma. In fact, there are several lines of both targeted agents and ICPI that are now feasible treatment options. However, survival in the metastatic setting continues to be poor and there remains a need for improved therapeutic approaches. In order to enhance patient selection for the most appropriate next line of therapy, better predictive biomarkers of responsiveness will need to be developed in tandem with technologies to identify mechanisms of ICPI resistance. Adaptive, biomarker-driven trials will drive this evolution. The combination of ICPI with specific chemotherapies, targeted therapies and other immuno-oncology (IO) drugs in order to circumvent ICPI resistance and enhance efficacy is discussed. Recent data support the role for both targeted therapies and ICPI in the adjuvant setting of melanoma and targeted therapies in the adjuvant setting for renal cell carcinoma, which may influence the consideration of treatment on subsequent relapse. Approaches to select the optimal treatment sequences for these patients will need to be refined.

Turajlic, S. Swanton, C. Boshoff, C (2018) Kidney cancer: The next decade.. Show Abstract full text

Chris Boshoff, Senior Vice President of Immuno-Oncology, Translational and Early Development at Pfizer, and colleagues Samra Turajlic and Charles Swanton from the Francis Crick Institute and University College London give us their personal point of view on new insights and future therapeutic approaches for renal cancer.

Tio, M. Rai, R. Ezeoke, O.M. McQuade, J.L. Zimmer, L. Khoo, C. Park, J.J. Spain, L. Turajlic, S. Ardolino, L. Yip, D. Goldinger, S.M. Cohen, J.V. Millward, M. Atkinson, V. Kane, A.Y. Ascierto, P.A. Garbe, C. Gutzmer, R. Johnson, D.B. Rizvi, H.A. Joshua, A.M. Hellmann, M.D. Long, G.V. Menzies, A.M (2018) Anti-PD-1/PD-L1 immunotherapy in patients with solid organ transplant, HIV or hepatitis B/C infection.. Show Abstract full text

<h4>Background</h4>Anti-programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) immunotherapy is now routinely used to treat several cancers. Clinical trials have excluded several populations, including patients with solid organ transplant, HIV infection and hepatitis B/C infection. We examined the safety outcomes of these populations treated with anti-PD-1/PD-L1 treatment in a multicentre retrospective study.<h4>Methods</h4>Patients from 16 centres with advanced cancer and solid organ transplant, HIV infection or hepatitis B/C infection were included. Demographic, tumour, treatment, toxicity and outcome data were recorded.<h4>Results</h4>Forty-six patients were included for analysis, with a median age of 60 years, and the majority of patients diagnosed with melanoma (72%). Among six patients with solid organ transplants, two graft rejections occurred, with one resulting in death, whereas two patients achieved partial responses. There were four responses in 12 patients with HIV infection. In 14 patients with hepatitis B, there were three responses, and similarly, there were three responses in 14 patients with hepatitis C. There was no unexpected toxicity in any viral infection group or an increase in viral load.<h4>Conclusion</h4>Patients with HIV or hepatitis B/C infections treated with anti-PD-1/PD-L1 immunotherapy may respond to treatment without increased toxicity. Given the risk of graft rejection in solid organ transplant patients and also the potential for response, the role of anti-PD-1/PD-L1 immunotherapy needs to be carefully considered.