Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Manoj Raghavan is active.

Publication


Featured researches published by Manoj Raghavan.


Blood | 2012

Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML)

Oliver Goodyear; Michael Dennis; Nadira Y. Jilani; Justin Loke; Shamyla Siddique; Gordon Ryan; Jane Nunnick; Rahela Khanum; Manoj Raghavan; Mark Cook; John A. Snowden; Mike Griffiths; Nigel H. Russell; John A. Liu Yin; Charles Crawley; Gordon Cook; Paresh Vyas; Paul Moss; Ram Malladi; Charles Craddock

Strategies that augment a GVL effect without increasing the risk of GVHD are required to improve the outcome after allogeneic stem cell transplantation (SCT). Azacitidine (AZA) up-regulates the expression of tumor Ags on leukemic blasts in vitro and expands the numbers of immunomodulatory T regulatory cells (Tregs) in animal models. Reasoning that AZA might selectively augment a GVL effect, we studied the immunologic sequelae of AZA administration after allogeneic SCT. Twenty-seven patients who had undergone a reduced intensity allogeneic transplantation for acute myeloid leukemia were treated with monthly courses of AZA, and CD8(+) T-cell responses to candidate tumor Ags and circulating Tregs were measured. AZA after transplantation was well tolerated, and its administration was associated with a low incidence of GVHD. Administration of AZA increased the number of Tregs within the first 3 months after transplantation compared with a control population (P = .0127). AZA administration also induced a cytotoxic CD8(+) T-cell response to several tumor Ags, including melanoma-associated Ag 1, B melanoma antigen 1, and Wilm tumor Ag 1. These data support the further examination of AZA after transplantation as a mechanism of augmenting a GVL effect without a concomitant increase in GVHD.


Cell Reports | 2015

Chronic FLT3-ITD Signaling in Acute Myeloid Leukemia Is Connected to a Specific Chromatin Signature

Pierre Cauchy; Sally R. James; Joaquin Zacarias-Cabeza; Anetta Ptasinska; Maria Rosaria Imperato; Salam A. Assi; Jason Piper; Martina Canestraro; Maarten Hoogenkamp; Manoj Raghavan; Justin Loke; Susanna Akiki; Samuel Clokie; Stephen J. Richards; David R. Westhead; Michael Griffiths; Sascha Ott; Constanze Bonifer; Peter N. Cockerill

Summary Acute myeloid leukemia (AML) is characterized by recurrent mutations that affect the epigenetic regulatory machinery and signaling molecules, leading to a block in hematopoietic differentiation. Constitutive signaling from mutated growth factor receptors is a major driver of leukemic growth, but how aberrant signaling affects the epigenome in AML is less understood. Furthermore, AML cells undergo extensive clonal evolution, and the mutations in signaling genes are often secondary events. To elucidate how chronic growth factor signaling alters the transcriptional network in AML, we performed a system-wide multi-omics study of primary cells from patients suffering from AML with internal tandem duplications in the FLT3 transmembrane domain (FLT3-ITD). This strategy revealed cooperation between the MAP kinase (MAPK) inducible transcription factor AP-1 and RUNX1 as a major driver of a common, FLT3-ITD-specific gene expression and chromatin signature, demonstrating a major impact of MAPK signaling pathways in shaping the epigenome of FLT3-ITD AML.


Leukemia Research | 2013

Post-transplant T cell chimerism predicts graft versus host disease but not disease relapse in patients undergoing an alemtuzumab based reduced intensity conditioned allogeneic transplant

E. Nikolousis; S. Robinson; Sandeep Nagra; C. Brookes; F. Kinsella; Sudhir Tauro; S. Jeffries; M. Griffiths; Premini Mahendra; Mark Cook; S. Paneesha; R. Lovell; B. Kishore; S. Chaganti; Ram Malladi; Manoj Raghavan; Paul Moss; Donald Milligan; Charles Craddock

In this multicentre retrospective study we have studied the impact of T cell chimerism on the outcome of 133 patients undergoing an alemtuzumab based reduced intensity conditioning allograft (RIC). The median age of the patients was 50 years (range 42-55 years). 77 patients were transplanted using an HLA identical sibling donor while 56 patients received a fully matched volunteer unrelated donor graft. 64 patients had a lymphoid malignancy and 69 were transplanted for a myeloid malignancy. 38 patients (29%) relapsed with no significant difference in risk of relapse between patients developing full donor and mixed donor chimerism in the T-cell compartment on D+90 and D+180 post transplant. Day 90 full donor T cell chimerism correlated with an increased incidence of acute GVHD according to NIH criteria (p=0.0004) and the subsequent development of chronic GVHD. Consistent with previous observations, our results confirmed a correlation between the establishment of T cell full donor chimerism and acute GVHD in T deplete RIC allografts. However our study failed to identify any correlation between T cell chimerism and relapse risk and challenge the use of pre-emptive donor lymphocyte infusions (DLI) in patients with mixed T cell chimerism transplanted using an alemtuzumab based RIC regimen.


Cancer immunology research | 2017

Identification of Glycopeptides as Posttranslationally Modified Neoantigens in Leukemia

Stacy A. Malaker; Sarah Penny; Lora Steadman; Paisley T. Myers; Justin Loke; Manoj Raghavan; Dina L. Bai; Jeffrey Shabanowitz; Donald F. Hunt; Mark Cobbold

The identification of neoepitopes expressed by tumors will aid the effectiveness of antitumor therapies. Four classes of posttranslationally modified tumor neoantigens were identified on primary tumors. Healthy donors had detectable natural immunity to a subset of these. Leukemias are highly immunogenic, but they have a low mutational load, providing few mutated peptide targets. Thus, the identification of alternative neoantigens is a pressing need. Here, we identify 36 MHC class I–associated peptide antigens with O-linked β-N-acetylglucosamine (O-GlcNAc) modifications as candidate neoantigens, using three experimental approaches. Thirteen of these peptides were also detected with disaccharide units on the same residues and two contain either mono- and/or di-methylated arginine residues. A subset were linked with key cancer pathways, and these peptides were shared across all of the leukemia patient samples tested (5/5). Seven of the O-GlcNAc peptides were synthesized and five (71%) were shown to be associated with multifunctional memory T-cell responses in healthy donors. An O-GlcNAc-specific T-cell line specifically killed autologous cells pulsed with the modified peptide, but not the equivalent unmodified peptide. Therefore, these posttranslationally modified neoantigens provide logical targets for cancer immunotherapy. Cancer Immunol Res; 5(5); 376–84. ©2017 AACR.


Cancer Research | 2005

Association between Large-scale Genomic Homozygosity without Chromosomal Loss and Nonseminomatous Germ Cell Tumor Development

Yong-Jie Lu; Jinshu Yang; Elodie Noel; Spyros Skoulakis; Tracy Chaplin; Manoj Raghavan; Trisha Purkis; Alan McIntyre; Sakunthala C. Kudahetti; Mahmoud Naase; Daniel M. Berney; Janet Shipley; R.T.D. Oliver; Bryan D. Young

The genotype of a tumor determines its biology and clinical behavior. The genetic alterations associated with the unique embryonal morphology of nonseminomatous subtypes of testicular germ cell tumors remain to be established. Using single nucleotide polymorphism microarray analysis, we found in all of the 15 nonseminomas analyzed, large-scale chromosomal homozygosities, most of which were not associated with relative chromosome loss. This unusual genotype, distinguishing nonseminoma from seminomas and other human tumors, may be associated with the special embryonal development morphologic transition of this malignancy. Based on these genetic data, we hypothesized a new potential origin of nonseminomas through sperm fusion. Nonrandom involvement of certain chromosomes also suggests that genes on these chromosome regions may play an important role in nonseminoma development.


Clinical Cancer Research | 2017

Outcome of Azacitidine Therapy in Acute Myeloid Leukemia Is not Improved by Concurrent Vorinostat Therapy but Is Predicted by a Diagnostic Molecular Signature.

Charles Craddock; Aimee E. Houlton; Lynn Quek; Paul Ferguson; Emma Gbandi; Corran Roberts; M Metzner; Natalia Garcia-Martin; Alison Kennedy; Angela Hamblin; Manoj Raghavan; Sandeep Nagra; Louise Dudley; Keith Wheatley; Mary Frances McMullin; Srinivas Pillai; Richard Kelly; Shamyla Siddique; Michael Dennis; Jamie Cavenagh; Paresh Vyas

Purpose: Azacitidine (AZA) is a novel therapeutic option in older patients with acute myeloid leukemia (AML), but its rational utilization is compromised by the fact that neither the determinants of clinical response nor its mechanism of action are defined. Co-administration of histone deacetylase inhibitors, such as vorinostat (VOR), is reported to improve the clinical activity of AZA, but this has not been prospectively studied in patients with AML. Experimental Design: We compared outcomes in 259 adults with AML (n = 217) and MDS (n = 42) randomized to receive either AZA monotherapy (75 mg/m2 × 7 days every 28 days) or AZA combined with VOR 300 mg twice a day on days 3 to 9 orally. Next-generation sequencing was performed in 250 patients on 41 genes commonly mutated in AML. Serial immunophenotyping of progenitor cells was performed in 47 patients. Results: Co-administration of VOR did not increase the overall response rate (P = 0.84) or overall survival (OS; P = 0.32). Specifically, no benefit was identified in either de novo or relapsed AML. Mutations in the genes CDKN2A (P = 0.0001), IDH1 (P = 0.004), and TP53 (P = 0.003) were associated with reduced OS. Lymphoid multipotential progenitor populations were greatly expanded at diagnosis and although reduced in size in responding patients remained detectable throughout treatment. Conclusions: This study demonstrates no benefit of concurrent administration of VOR with AZA but identifies a mutational signature predictive of outcome after AZA-based therapy. The correlation between heterozygous loss of function CDKN2A mutations and decreased OS implicates induction of cell-cycle arrest as a mechanism by which AZA exerts its clinical activity. Clin Cancer Res; 23(21); 6430–40. ©2017 AACR.


Blood Advances | 2016

Mutational analysis of disease relapse in patients allografted for acute myeloid leukemia

Lynn Quek; Paul Ferguson; M Metzner; Ikhlaaq Ahmed; Alison Kennedy; Catherine Garnett; Sally Jeffries; Claudia Walter; Kim Piechocki; Adele Timbs; Robert Danby; Manoj Raghavan; Andrew Peniket; Mike Griffiths; Andrew Bacon; Janice Ward; Keith Wheatley; Paresh Vyas; Charles Craddock

Disease relapse is the major cause of treatment failure after allogeneic stem cell transplantation (allo-SCT) in acute myeloid leukemia (AML). To identify AML-associated genes prognostic of AML relapse post-allo-SCT, we resequenced 35 genes in 113 adults at diagnosis, 49 of whom relapsed. Two hundred sixty-two mutations were detected in 102/113 (90%) patients. An increased risk of relapse was observed in patients with mutations in WT1 (P = .018), DNMT3A (P = .045), FLT3 ITD (P = .071), and TP53 (P = .06), whereas mutations in IDH1 were associated with a reduced risk of disease relapse (P = .018). In 29 patients, we additionally compared mutational profiles in bone marrow at diagnosis and relapse to study changes in clonal structure at relapse. In 13/29 patients, mutational profiles altered at relapse. In 9 patients, mutations present at relapse were not detected at diagnosis. In 15 patients, additional available pre-allo-SCT samples demonstrated that mutations identified posttransplant but not at diagnosis were detectable immediately prior to transplant in 2 of 15 patients. Taken together, these observations, if confirmed in larger studies, have the potential to inform the design of novel strategies to reduce posttransplant relapse highlighting the potential importance of post-allo-SCT interventions with a broad antitumor specificity in contrast to targeted therapies based on mutational profile at diagnosis.


Cell Reports | 2017

RUNX1-ETO and RUNX1-EVI1 Differentially Reprogram the Chromatin Landscape in t(8;21) and t(3;21) AML

Justin Loke; Salam A. Assi; Maria Rosaria Imperato; Anetta Ptasinska; Pierre Cauchy; Yura Grabovska; Natalia Martinez Soria; Manoj Raghavan; H. Ruud Delwel; Peter N. Cockerill; Olaf Heidenreich; Constanze Bonifer

Summary Acute myeloid leukemia (AML) is a heterogeneous disease caused by mutations in transcriptional regulator genes, but how different mutant regulators shape the chromatin landscape is unclear. Here, we compared the transcriptional networks of two types of AML with chromosomal translocations of the RUNX1 locus that fuse the RUNX1 DNA-binding domain to different regulators, the t(8;21) expressing RUNX1-ETO and the t(3;21) expressing RUNX1-EVI1. Despite containing the same DNA-binding domain, the two fusion proteins display distinct binding patterns, show differences in gene expression and chromatin landscape, and are dependent on different transcription factors. RUNX1-EVI1 directs a stem cell-like transcriptional network reliant on GATA2, whereas that of RUNX1-ETO-expressing cells is more mature and depends on RUNX1. However, both types of AML are dependent on the continuous expression of the fusion proteins. Our data provide a molecular explanation for the differences in clinical prognosis for these types of AML.


Cancer Research | 2018

MYBL2 Supports DNA Double Strand Break Repair in Hematopoietic Stem Cells

Rachel Bayley; Daniel Blakemore; Laila Cancian; Stephanie Dumon; Giacomo Volpe; Carl Ward; Ruba Almaghrabi; Jidnyasa Gujar; Natasha Reeve; Manoj Raghavan; Martin R. Higgs; Grant S. Stewart; Eva Petermann; Paloma García

Myelodysplastic syndromes (MDS) are a heterogeneous group of diseases characterized by blood cytopenias that occur as a result of somatic mutations in hematopoietic stem cells (HSC). MDS leads to ineffective hematopoiesis, and as many as 30% of patients progress to acute myeloid leukemia (AML). The mechanisms by which mutations accumulate in HSC during aging remain poorly understood. Here we identify a novel role for MYBL2 in DNA double-strand break (DSB) repair in HSC. In patients with MDS, low MYBL2 levels associated with and preceded transcriptional deregulation of DNA repair genes. Stem/progenitor cells from these patients display dysfunctional DSB repair kinetics after exposure to ionizing radiation (IR). Haploinsufficiency of Mybl2 in mice also led to a defect in the repair of DSBs induced by IR in HSC and was characterized by unsustained phosphorylation of the ATM substrate KAP1 and telomere fragility. Our study identifies MYBL2 as a crucial regulator of DSB repair and identifies MYBL2 expression levels as a potential biomarker to predict cellular response to genotoxic treatments in MDS and to identify patients with defects in DNA repair. Such patients with worse prognosis may require a different therapeutic regimen to prevent progression to AML.Significance: These findings suggest MYBL2 levels may be used as a biological biomarker to determine the DNA repair capacity of hematopoietic stem cells from patients with MDS and as a clinical biomarker to inform decisions regarding patient selection for treatments that target DNA repair.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/20/5767/F1.large.jpg Cancer Res; 78(20); 5767-79. ©2018 AACR.


The Lancet | 2017

Identification of common and distinct epigenetic reprogramming properties of RUNX1 fusion proteins in acute myeloid leukaemia

Justin Loke; Salam A. Assi; Maria Rosaria Imperato; Anetta Ptasinska; Pierre Cauchy; Manoj Raghavan; Ruud Delwel; Peter N. Cockerill; Olaf Heidenreich; Constanze Bonifer

Abstract Background Regulation of gene expression by transcription factors such as RUNX1 is crucial for haemopoiesis. The most common RUNX1 translocation resulting in acute myeloid leukaemia is t(8;21), which forms RUNX1–ETO; a second translocation—t(3;21)—results in RUNX1–EVI-1. Although, these two fusion proteins have the same DNA binding domain, the prognoses of patients with these translocations differ greatly. Whether different RUNX1 fusion proteins deregulate the same genes is unknown. We sought to understand the differences in the epigenome that underlie these prognostic differences. Methods DNase-seq maps regions of open chromatin that represent active gene regulatory elements. By using this technique with RNA-seq, we were able to describe the epigenetic landscape in CD34+ purified, primary material from patients with RUNX1–EVI-1 and RUNX1–ETO leukaemias, and in healthy controls. We used ChIP-seq to map the binding sites of both normal RUNX1 and the fusion proteins. We integrated these analyses to determine transcription factor networks that characterise each type of leukaemia. Findings RUNX1–EVI-1, but not RUNX1–ETO, directly regulated a unique subset of genes required for stem-cell function. We found that these differences in binding sites of the two fusion proteins were associated with differences in the transcription factor complexes that collaborate with them, and were directly related to the differences in the epigenetic landscape of each leukaemia. RUNX1–EVI-1 knockdown restored differentiation of t(3;21) cells and this finding was associated with upregulation of genes crucial for myeloid differentiation, including C/EBPα. We showed that C/EBPα was necessary and sufficient for the response of t(3;21) cells to RUNX1–EVI-1 knockdown and that C/EBPα was commonly deregulated in both leukaemias. Interpretation The differences in the clinical outcomes of each RUNX1 mutant leukaemia was reflected by differences in their epigenetic landscape. This finding was driven by differences in the transcription factor networks in each type of leukaemia. Despite these differences, both leukaemias were dependent on downregulating C/EBP, thereby providing a common therapeutic route for both RUNX1–ETO and RUNX1–EVI-1 leukaemia. Funding Kay Kendall Leukaemia Fund, Bloodwise.

Collaboration


Dive into the Manoj Raghavan's collaboration.

Top Co-Authors

Avatar

Charles Craddock

Queen Elizabeth Hospital Birmingham

View shared research outputs
Top Co-Authors

Avatar

Bryan D. Young

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tracy Chaplin

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Justin Loke

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul Ferguson

Queen Elizabeth Hospital Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ram Malladi

University of Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge