Anna Schuh
John Radcliffe Hospital
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Anna Schuh.
The New England Journal of Medicine | 2014
John C. Byrd; Jennifer R. Brown; Susan O'Brien; Jaqueline C. Barrientos; Neil E. Kay; Nashitha Reddy; Steven Coutre; Constantine S. Tam; Stephen P. Mulligan; Ulrich Jaeger; S Devereux; Paul M. Barr; Richard R. Furman; Thomas J. Kipps; Florence Cymbalista; Christopher Pocock; Patrick Thornton; Federico Caligaris-Cappio; Tadeusz Robak; J. Delgado; Stephen J. Schuster; Marco Montillo; Anna Schuh; S. de Vos; Devinder Gill; Adrian Bloor; Claire Dearden; Carol Moreno; J. J. Jones; Alvina D. Chu
BACKGROUNDnIn patients with chronic lymphoid leukemia (CLL) or small lymphocytic lymphoma (SLL), a short duration of response to therapy or adverse cytogenetic abnormalities are associated with a poor outcome. We evaluated the efficacy of ibrutinib, a covalent inhibitor of Brutons tyrosine kinase, in patients at risk for a poor outcome.nnnMETHODSnIn this multicenter, open-label, phase 3 study, we randomly assigned 391 patients with relapsed or refractory CLL or SLL to receive daily ibrutinib or the anti-CD20 antibody ofatumumab. The primary end point was the duration of progression-free survival, with the duration of overall survival and the overall response rate as secondary end points.nnnRESULTSnAt a median follow-up of 9.4 months, ibrutinib significantly improved progression-free survival; the median duration was not reached in the ibrutinib group (with a rate of progression-free survival of 88% at 6 months), as compared with a median of 8.1 months in the ofatumumab group (hazard ratio for progression or death in the ibrutinib group, 0.22; P<0.001). Ibrutinib also significantly improved overall survival (hazard ratio for death, 0.43; P=0.005). At 12 months, the overall survival rate was 90% in the ibrutinib group and 81% in the ofatumumab group. The overall response rate was significantly higher in the ibrutinib group than in the ofatumumab group (42.6% vs. 4.1%, P<0.001). An additional 20% of ibrutinib-treated patients had a partial response with lymphocytosis. Similar effects were observed regardless of whether patients had a chromosome 17p13.1 deletion or resistance to purine analogues. The most frequent nonhematologic adverse events were diarrhea, fatigue, pyrexia, and nausea in the ibrutinib group and fatigue, infusion-related reactions, and cough in the ofatumumab group.nnnCONCLUSIONSnIbrutinib, as compared with ofatumumab, significantly improved progression-free survival, overall survival, and response rate among patients with previously treated CLL or SLL. (Funded by Pharmacyclics and Janssen; RESONATE ClinicalTrials.gov number, NCT01578707.).
The Lancet | 2015
Peter Hillmen; Tadeusz Robak; Ann Janssens; K Govind Babu; Janusz Kloczko; Sebastian Grosicki; Michael Doubek; Panagiotis Panagiotidis; Eva Kimby; Anna Schuh; Andrew R. Pettitt; Thomas E. Boyd; Marco Montillo; Ira V. Gupta; Oliver Wright; Iestyn Dixon; J. Carey; Chai-Ni Chang; Steen Lisby; Astrid McKeown; Fritz Offner
BACKGROUNDnTreatment for patients with chronic lymphocytic leukaemia who are elderly or who have comorbidities is challenging because fludarabine-based chemoimmunotherapies are mostly not suitable. Chlorambucil remains the standard of care in many countries. We aimed to investigate whether the addition of ofatumumab to chlorambucil could lead to better clinical outcomes than does treatment with chlorambucil alone, while also being tolerable for patients who have few treatment options.nnnMETHODSnWe carried out a randomised, open-label, phase 3 trial for treatment-naive patients with chronic lymphocytic leukaemia in 109 centres in 16 countries. We included patients who had active disease needing treatment, but in whom fludarabine-based treatment was not possible. We randomly assigned patients (1:1) to receive oral chlorambucil (10 mg/m(2)) on days 1-7 of a 28 day treatment course or to receive chlorambucil by this schedule plus intravenous ofatumumab (cycle 1: 300 mg on day 1 and 1000 mg on day 8; subsequent cycles: 1000 mg on day 1) for three to 12 cycles. Assignment was done with a randomisation list that was computer generated at GlaxoSmithKline, and was stratified, in a block size of two, by age, disease stage, and performance status. The primary endpoint was progression-free survival in the intention-to-treat population and assessment was done by an independent review committee that was masked to group assignment. The study is registered with ClinicalTrials.gov, number NCT00748189.nnnFINDINGSnWe enrolled 447 patients, median age 69 years (range 35-92). Between Dec 22, 2008, and May 26, 2011, we randomly assigned 221 patients to chlorambucil plus ofatumumab and 226 patients to chlorambucil alone. Median progression-free survival was 22·4 months (95% CI 19·0-25·2) in the group assigned to chlorambucil plus ofatumumab compared with 13·1 months (10·6-13·8) in the group assigned to chlorambucil only (hazard ratio 0·57, 95% CI 0·45-0·72; p<0·0001). Grade 3 or greater adverse events were more common in the chlorambucil plus ofatumumab group (109 [50%] patients; vs 98 [43%] given chlorambucil alone), with neutropenia being the most common event (56 [26%] vs 32 [14%]). Grade 3 or greater infections had similar frequency in both groups. Grade 3 or greater infusion-related adverse events were reported in 22 (10%) patients given chlorambucil plus ofatumumab. Five (2%) patients died during treatment in each group.nnnINTERPRETATIONnAddition of ofatumumab to chlorambucil led to clinically important improvements with a manageable side-effect profile in treatment-naive patients with chronic lymphocytic leukaemia who were elderly or had comorbidities. Chlorambucil plus ofatumumab is therefore an important treatment option for these patients who cannot tolerate more intensive therapy.nnnFUNDINGnGlaxoSmithKline, Genmab A/S.
Leukemia | 2013
Charles Craddock; Lynn Quek; Nicolas Goardon; Sylvie Freeman; Shamyla Siddique; Manoj Raghavan; A. Aztberger; Anna Schuh; David Grimwade; Adam Ivey; Paul Virgo; Robert Kerrin Hills; T. McSkeane; Julie Arrazi; Steven Knapper; Cassandra Brookes; B Davies; A J Price; Kerry Wall; Michael Griffiths; J Cavenagh; Ravindra Majeti; Irving L. Weissman; Alan Kenneth Burnett; Paresh Vyas
Epigenetic therapies demonstrate significant clinical activity in acute myeloid leukemia (AML) and myelodysplasia (MDS) and constitute an important new class of therapeutic agents. However hematological responses are not durable and disease relapse appears inevitable. Experimentally, leukemic stem/progenitor cells (LSC) propagate disease in animal models of AML and it has been postulated that their relative chemo-resistance contributes to disease relapse. We serially measured LSC numbers in patients with high-risk AML and MDS treated with 5-azacitidine and sodium valproate (VAL–AZA). Fifteen out of seventy-nine patients achieved a complete remission (CR) or complete remission with incomplete blood count recovery (CRi) with VAL–AZA therapy. There was no significant reduction in the size of the LSC-containing population in non-responders. While the LSC-containing population was substantially reduced in all patients achieving a CR/CRi it was never eradicated and expansion of this population antedated morphological relapse. Similar studies were performed in seven patients with newly diagnosed AML treated with induction chemotherapy. Eradication of the LSC-containing population was observed in three patients all of whom achieved a durable CR in contrast to patients with resistant disease where LSC persistence was observed. LSC quantitation provides a novel biomarker of disease response and relapse in patients with AML treated with epigenetic therapies. New drugs that target this cellular population in vivo are required.
British Journal of Haematology | 2012
David Oscier; Claire Dearden; Efrem Eren; Christopher Fegan; George A. Follows; Peter Hillmen; Tim Illidge; Estella Matutes; Don Milligan; Andrew R. Pettitt; Anna Schuh; Jennifer Wimperis
Royal Bournemouth Hospital, Bournemouth, 2 Royal Marsden Hospital, London, 3 Southampton General Hospital, Southampton, Cardiff and Vale NHS Trust, Cardiff Cambridge University Hospitals NHS Foundation Trust, Cambridge UK; St. Jamess Institute of Oncology, Leeds, Christie Hospital NHS Trust, Manchester Royal Marsden Hospital London; Heart of England NHS Foundataion Trust, Birmingham; 10 Royal Liverpool University Hospital, Liverpool; Churchill Hospital, Headington, Oxford, Norfolk and Norwich University Hospital, Norwich
British Journal of Haematology | 2011
Letizia Foroni; Gill Wilson; Gareth Gerrard; Joanne Mason; David Grimwade; Helen E. White; David Gonzalez de Castro; Stephen Austin; Abida Awan; Emma Burt; Tim Clench; Joanna Farruggia; Jeremy Hancock; Alexandra Irvine; Aytug Kizilors; Stephen E. Langabeer; Benedict Milner; Guillermina Nickless; Anna Schuh; Anne Sproul; Lihui Wang; Caroline Wickham; Nicholas C.P. Cross
Molecular testing for the BCR‐ABL1 fusion gene by real time quantitative polymerase chain reaction (RT‐qPCR) is the most sensitive routine approach for monitoring the response to therapy of patients with chronic myeloid leukaemia. In the context of tyrosine kinase inhibitor (TKI) therapy, the technique is most appropriate for patients who have achieved complete cytogenetic remission and can be used to define specific therapeutic milestones. To achieve this effectively, standardization of the laboratory procedures and the interpretation of results are essential. We present here consensus best practice guidelines for RT‐qPCR testing, data interpretation and reporting that have been drawn up and agreed by a consortium of 21 testing laboratories in the United Kingdom and Ireland in accordance with the procedures of the UK Clinical Molecular Genetics Society.
Leukemia | 2012
Samantha J. L. Knight; Christopher Yau; Ruth Clifford; Adele Timbs; E Sadighi Akha; Helene Dreau; Adam Burns; C Ciria; David Oscier; Andrew R. Pettitt; S Dutton; Christopher Holmes; Jenny C. Taylor; J-B Cazier; Anna Schuh
Genome-wide array approaches and sequencing analyses are powerful tools for identifying genetic aberrations in cancers, including leukemias and lymphomas. However, the clinical and biological significance of such aberrations and their subclonal distribution are poorly understood. Here, we present the first genome-wide array based study of pre-treatment and relapse samples from patients with B-cell chronic lymphocytic leukemia (B-CLL) that uses the computational statistical tool OncoSNP. We show that quantification of the proportion of copy number alterations (CNAs) and copy neutral loss of heterozygosity regions (cnLOHs) in each sample is feasible. Furthermore, we (i) reveal complex changes in the subclonal architecture of paired samples at relapse compared with pre-treatment, (ii) provide evidence supporting an association between increased genomic complexity and poor clinical outcome (iii) report previously undefined, recurrent CNA/cnLOH regions that expand or newly occur at relapse and therefore might harbor candidate driver genes of relapse and/or chemotherapy resistance. Our findings are likely to impact on future therapeutic strategies aimed towards selecting effective and individually tailored targeted therapies.Leukemia advance online publication, 14 February 2012; doi:10.1038/leu.2012.13
Clinical Biochemistry | 2009
Shirley Henderson; Adele Timbs; Janice McCarthy; Alice Gallienne; Margaretha Van Mourik; Gillian S. Masters; Alison May; Mohamed S. Khalil; Anna Schuh; John Old
OBJECTIVESnThe aim of this study was to update the incidence data of beta thalassaemia mutations in various populations and compare it to the spectrum of mutations in the United Kingdom (UK) population in order to determine the impact of immigration.nnnDESIGN AND METHODSnPublished data for the beta-thalassaemia mutation spectrum and allele frequencies for 60 other countries was updated and collated into regional tables. The beta-thalassaemia mutations in the UK population have been characterised in 1712 unrelated carriers referred for antenatal screening. Similarly, the alpha-thalassaemia mutations in the UK population have been characterised in 2500 possible alpha-thalassaemia carriers.nnnRESULTSnA total of 68 different beta-thalassaemia mutations were identified in couples requiring screening for antenatal diagnosis in the UK population. Of these mutations, 59 were found in immigrants to the UK, from all major ethnic groups with a high incidence of haemoglobinopathies. A total of 40 different alpha-thalassaemia mutations were characterised in the UK population. Ten deletion mutations were identified, including all the Southeast Asian and Mediterranean alpha(0)-thalassaemia mutations. In addition, 30 non-deletion alpha(+)-thalassaemia mutations were discovered, accounting for 46% of the worldwide known non-deletion mutations.nnnCONCLUSIONSnThe impact of immigration has resulted in the UK population having a higher number of beta-thalassaemia mutations and alpha-thalassaemia mutations than any of the 60 other countries with a published spectrum of mutations, including both endemic countries and the non-endemic countries of Northern Europe. The racial heterogeneity of the immigrant population in a non-endemic country significantly increases the spectrum of haemoglobinopathy mutations and their combinations found in individuals, making the provision of a molecular diagnostic prenatal diagnosis service more challenging.
Journal of Clinical Immunology | 2014
Fatima Dhalla; Mary Lucas; Anna Schuh; Malini V. Bhole; Rashmi Jain; Smita Y. Patel; Siraj Misbah; Helen Chapel
Hypogammaglobulinemia is a common finding in chronic lymphocytic leukemia (CLL). Its incidence increases with disease duration and stage such that it is present in up to 85xa0% of patients at some point in their disease course. It is therefore important to monitor patients for the development of an antibody deficiency. However, not all patients with antibody deficiency secondary to CLL are symptomatic with bacterial infections. In addition patients are susceptible to viral, fungal and opportunistic infections as a result of iatrogenic immunosuppression and through a variety of disease-related mechanisms, which affect cellular immunity and phagocytes. Published guidelines suggest that patients with a history of recurrent bacterial infections and a documented failure of antibody production should be treated with antibiotic prophylaxis in the first instance, with replacement immunoglobulin reserved for those who continue to suffer with significant bacterial infections. Here we present a review of the existing literature in order to provide a practical approach, based on best available evidence, to the investigation, monitoring and treatment of patients with antibody failure secondary to CLL; and we highlight areas in which further studies are needed.
Blood | 2013
Adam Mead; Onima Chowdhury; Christian Pecquet; Alexandra Dusa; Petter S. Woll; Deborah Atkinson; Adam Burns; Joannah Score; Michelle Rugless; Ruth Clifford; Simon Moule; Nicola Bienz; Paresh Vyas; Nicholas C.P. Cross; Rosemary E. Gale; Shirley Henderson; Stefan N. Constantinescu; Anna Schuh; Sten Eirik W. Jacobsen
The association between somatic JAK2 mutation and myeloproliferative neoplasms (MPNs) is now well established. However, because JAK2 mutations are associated with heterogeneous clinical phenotypes and often occur as secondary genetic events, some aspects of JAK2 mutation biology remain to be understood. We recently described a germline JAK2V617I mutation in a family with hereditary thrombocytosis and herein characterize the hematopoietic and signaling impact of JAK2V617I. Through targeted sequencing of MPN-associated mutations, exome sequencing, and clonality analysis, we demonstrate that JAK2V617I is likely to be the sole driver mutation in JAK2V617I-positive individuals with thrombocytosis. Phenotypic hematopoietic stem cells (HSCs) were increased in the blood and bone marrow of JAK2V617I-positive individuals and were sustained at higher levels than controls after xenotransplantation. In signaling and transcriptional assays, JAK2V617I demonstrated more activity than wild-type JAK2 but substantially less than JAK2V617F. After cytokine stimulation, JAK2V617I resulted in markedly increased downstream signaling compared with wild-type JAK2 and comparable with JAK2V617F. These findings demonstrate that JAK2V617I induces sufficient cytokine hyperresponsiveness in the absence of other molecular events to induce a homogeneous MPN-like phenotype. We also provide evidence that the JAK2V617I mutation may expand the HSC pool, providing insights into both JAK2 mutation biology and MPN disease pathogenesis.
Haematologica | 2015
Carol Moreno; Marco Montillo; P Panayiotidis; Maria Dimou; Adrian Bloor; Jehan Dupuis; Anna Schuh; Stefan Norin; Christian H. Geisler; Peter Hillmen; Michael Doubek; Marek Trněný; Petra Obrtlikova; Luca Laurenti; Stephan Stilgenbauer; Lukas Smolej; Paolo Ghia; Florence Cymbalista; Ulrich Jaeger; Kostas Stamatopoulos; Niki Stavroyianni; Patrick Carrington; Hamadi Zouabi; Véronique Leblond; Juan C. Gomez-Garcia; Martin Rubio; Roberto Marasca; Gerardo Musuraca; Luigi Rigacci; Lucia Farina
We report the largest retrospective, phase IV non-interventional, observational study of ofatumumab therapy in heavily pre-treated patients with poor-prognosis chronic lymphocytic leukemia. Total number of patients was 103; median age was 65 years (range 39–85). Median number of prior lines of therapy was 4 (range 1–13), including, in most cases, rituximab-, fludarabine- and alemtuzumab-based regimens; 13 patients had been allografted. Of 113 adverse events, 28 (29%) were considered to be directly related to ofatumumab. Grade 3–4 toxicities included neutropenia (10%), thrombocytopenia (5%), anemia (3%), pneumonia (17%), and fever (3%). Two heavily pre-treated patients developed progressive multifocal leukoencephalopathy. On an intention-to-treat analysis, the overall response rate was 22% (3 complete response, 1 incomplete complete response). Median progression-free and overall survival times were 5 and 11 months, respectively. This study confirms in a daily-life setting the feasibility and acceptable toxicity of ofatumumab treatment in advanced chronic lymphocytic leukemia. The complete response rate, however, was low. Therefore, treatment with ofatumumab should be moved to earlier phases of the disease. Ideally, this should be done in combination with other agents, as recently approved for ofatumumab plus chlorambucil as front-line treatment for patients unfit for fludarabine. This study is registered at clinicaltrials.gov identifier:01453062.