Dexter, T.
Taiwo, T.
Dearden, C.
Chan, L.Y.
Taussig, D.
El-Sharkawi, D.
Dunlop, A.
Iyengar, S.
(2024). Correlation of T-cell receptor constant beta-chain 1 by flow cytometry with molecular T-cell receptor clonality for the investigation of T-cell lymphoproliferation. Br j haematol,
Vol.204
(4),
pp. 1554-1556.
Pietka, G.
De Lord, C.
Matthias, G.
Cheung, B.
Atwal, S.
Furtado, M.
Cullis, J.
Grey-Davies, L.
Narayanan, S.
McGregor, A.
Kilner, M.
Bosworth, J.
McMullin, M.F.
Coats, T.
Parcharidou, A.
Cavenagh, J.
Byrne, J.
Iyengar, S.
Mohammed, K.
Cross, N.
Hubank, M.
Ribeiro, S.
Khorashad, J.
Wren, D.
O'Connor, S.
Taussig, D.
(2024). Capture-based targeted sequencing using a T-cell control in myeloid malignancies and idiopathic cytopenias. Br j haematol,
Vol.204
(4),
pp. 1325-1334.
show abstract
We report on a study of next-generation sequencing in 257 patients undergoing investigations for cytopenias. We sequenced bone marrow aspirates using a target enrichment panel comprising 82 genes and used T cells from paired blood as a control. One hundred and sixty patients had idiopathic cytopenias, 81 had myeloid malignancies and 16 had lymphoid malignancies or other diagnoses. Forty-seven of the 160 patients with idiopathic cytopenias had evidence of somatic pathogenic variants consistent with clonal cytopenias. Only 39 genes of the 82 tested were mutated in the 241 patients with either idiopathic cytopenias or myeloid neoplasms. We confirm that T cells can be used as a control to distinguish between germline and somatic variants. The use of paired analysis with a T-cell control significantly reduced the time molecular scientists spent reporting compared to unpaired analysis. We identified somatic variants of uncertain significance (VUS) in a higher proportion (24%) of patients with myeloid malignancies or clonal cytopenias compared to less than 2% of patients with non-clonal cytopenias. This suggests that somatic VUS are indicators of a clonal process. Lastly, we show that blood depleted of lymphocytes can be used in place of bone marrow as a source of material for sequencing..
Jimenez-Chillon, C.
Othman, J.
Taussig, D.
Jimenez-Vicente, C.
Martinez-Roca, A.
Tiong, I.S.
Jain, M.
Aries, J.
Cakmak, S.
Knapper, S.
Kristensen, D.T.
Murthy, V.
Galani, J.Z.
Kallmeyer, C.
Ngu, L.
Veale, D.
Bolam, S.
Orfali, N.
Parker, A.
Manson, C.
Parker, J.
Erblich, T.
Richardson, D.
Mokretar, K.
Potter, N.
Overgaard, U.M.
Roug, A.S.
Wei, A.H.
Esteve, J.
Jädersten, M.
Russell, N.
Dillon, R.
(2024). Venetoclax-based low intensity therapy in molecular failure of NPM1-mutated AML. Blood adv,
Vol.8
(2),
pp. 343-352.
show abstract
full text
Molecular failure in NPM1-mutated acute myeloid leukemia (AML) inevitably progresses to frank relapse if untreated. Recently published small case series show that venetoclax combined with low-dose cytarabine or azacitidine can reduce or eliminate measurable residual disease (MRD). Here, we report on an international multicenter cohort of 79 patients treated for molecular failure with venetoclax combinations and report an overall molecular response (≥1-log reduction in MRD) in 66 patients (84%) and MRD negativity in 56 (71%). Eighteen of 79 patients (23%) required hospitalization, and no deaths were reported during treatment. Forty-one patients were bridged to allogeneic transplant with no further therapy, and 25 of 41 were MRD negative assessed by reverse transcription quantitative polymerase chain reaction before transplant. Overall survival (OS) for the whole cohort at 2 years was 67%, event-free survival (EFS) was 45%, and in responding patients, there was no difference in survival in those who received a transplant using time-dependent analysis. Presence of FLT3-ITD mutation was associated with a lower response rate (64 vs 91%; P < .01), worse OS (hazard ratio [HR], 2.50; 95% confidence interval [CI], 1.06-5.86; P = .036), and EFS (HR, 1.87; 95% CI, 1.06-3.28; P = .03). Eighteen of 35 patients who did not undergo transplant became MRD negative and stopped treatment after a median of 10 months, with 2-year molecular relapse free survival of 62% from the end of treatment. Venetoclax-based low intensive chemotherapy is a potentially effective treatment for molecular relapse in NPM1-mutated AML, either as a bridge to transplant or as definitive therapy..
Smith, K.
Fathoala, D.
Anthias, C.
Easdale, S.
Ethell, M.
Mir, F.
Pickard, L.
Potter, M.
Taussig, D.
Nicholson, E.
(2023). Venetoclax-based salvage for management of relapsed acute leukaemia after allogeneic haematopoetic stem cell transplant: durable remissions only seen following consolidation with donor lymphocyte infusion or second transplant. Bone marrow transplant,
Vol.58
(10),
pp. 1152-1156.
Othman, J.
Potter, N.
Mokretar, K.
Taussig, D.
Khan, A.
Krishnamurthy, P.
Latif, A.-.
Cahalin, P.
Aries, J.
Amer, M.
Belsham, E.
Conneally, E.
Craddock, C.
Culligan, D.
Dennis, M.
Duncan, C.
Freeman, S.D.
Furness, C.
Gilkes, A.
Gkreka, P.
Hodgson, K.
Ingram, W.
Jain, M.
King, A.
Knapper, S.
Kottaridis, P.
McMullin, M.F.
Mohite, U.
Ngu, L.
O'Nions, J.
Patrick, K.
Rider, T.
Roberts, W.
Severinsen, M.T.
Storrar, N.
Taylor, T.
Russell, N.H.
Dillon, R.
(2023). FLT3 inhibitors as MRD-guided salvage treatment for molecular failure in FLT3 mutated AML. Leukemia,
Vol.37
(10),
pp. 2066-2072.
show abstract
Patients with FLT3-mutated AML have a high relapse rate and suboptimal outcomes. Many have co-mutations suitable for measurable residual disease (MRD) monitoring by RT-qPCR and those destined to relapse can be identified by high or rising levels of MRD, called molecular failure. This provides a window for pre-emptive intervention, but there is little evidence to guide treatment. The use of FLT3 inhibitors (FLT3i) appears attractive but their use has not yet been evaluated. We identified 56 patients treated with FLT3i at molecular failure. The FLT3 mutation was an ITD in 52, TKD in 7 and both in 3. Over half of patients had previously received midostaurin. Molecular failure occurred at a median 9.2 months from diagnosis and was treated with gilteritinib (n = 38), quizartinib (n = 7) or sorafenib (n = 11). 60% achieved a molecular response, with 45% reaching MRD negativity. Haematological toxicity was low, and 22 patients were bridged directly to allogeneic transplant with another 6 to donor lymphocyte infusion. 2-year overall survival was 80% (95%CI 69-93) and molecular event-free survival 56% (95%CI 44-72). High-sensitivity next-generation sequencing for FLT3-ITD at molecular failure identified patients more likely to benefit. FLT3i monotherapy for molecular failure is a promising strategy which merits evaluation in prospective studies..
Sekeres, M.A.
Montesinos, P.
Novak, J.
Wang, J.
Jeyakumar, D.
Tomlinson, B.
Mayer, J.
Jou, E.
Robak, T.
Taussig, D.C.
Dombret, H.
Merchant, A.
Shaik, N.
O'Brien, T.
Roh, W.
Liu, X.
Ma, W.
DiRienzo, C.G.
Chan, G.
Cortes, J.E.
(2023). Glasdegib plus intensive or non-intensive chemotherapy for untreated acute myeloid leukemia: results from the randomized, phase 3 BRIGHT AML 1019 trial. Leukemia,
Vol.37
(10),
pp. 2017-2026.
show abstract
full text
This is the primary report of the randomized, placebo-controlled phase 3 BRIGHT AML 1019 clinical trial of glasdegib in combination with intensive chemotherapy (cytarabine and daunorubicin) or non-intensive chemotherapy (azacitidine) in patients with untreated acute myeloid leukemia. Overall survival (primary endpoint) was similar between the glasdegib and placebo arms in the intensive (n = 404; hazard ratio [HR] 1.05; 95% confidence interval [CI]: 0.782-1.408; two-sided p = 0.749) and non-intensive (n = 325; HR 0.99; 95% CI: 0.768-1.289; two-sided p = 0.969) studies. The proportion of patients who experienced treatment-emergent adverse events was similar for glasdegib versus placebo (intensive: 99.0% vs. 98.5%; non-intensive: 99.4% vs. 98.8%). The most common treatment-emergent adverse events were nausea, febrile neutropenia, and anemia in the intensive study and anemia, constipation, and nausea in the non-intensive study. The addition of glasdegib to either cytarabine and daunorubicin or azacitidine did not significantly improve overall survival and the primary efficacy endpoint for the BRIGHT AML 1019 phase 3 trial was not met. Clinical trial registration: ClinicalTrials.gov: NCT03416179..
de Botton, S.
Fenaux, P.
Yee, K.
Récher, C.
Wei, A.H.
Montesinos, P.
Taussig, D.C.
Pigneux, A.
Braun, T.
Curti, A.
Grove, C.
Jonas, B.A.
Khwaja, A.
Legrand, O.
Peterlin, P.
Arnan, M.
Blum, W.
Cilloni, D.
Hiwase, D.K.
Jurcic, J.G.
Krauter, J.
Thomas, X.
Watts, J.M.
Yang, J.
Polyanskaya, O.
Brevard, J.
Sweeney, J.
Barrett, E.
Cortes, J.
(2023). Olutasidenib (FT-2102) induces durable complete remissions in patients with relapsed or refractory IDH1-mutated AML. Blood adv,
Vol.7
(13),
pp. 3117-3127.
show abstract
Olutasidenib (FT-2102) is a potent, selective, oral, small-molecule inhibitor of mutant isocitrate dehydrogenase 1 (mIDH1). Overall, 153 IDH1 inhibitor-naive patients with mIDH1R132 relapsed/refractory (R/R) acute myeloid leukemia (AML) received olutasidenib monotherapy 150 mg twice daily in the pivotal cohort of this study. The median age of participants was 71 years (range, 32-87 years) and the median number of prior regimens received by patients was 2 (1-7). The rate of complete remission (CR) plus CR with partial hematologic recovery (CRh) was 35%, and the overall response rate was 48%. Response rates were similar in patients who had, and who had not, received prior venetoclax. With 55% of patients censored at the time of data cut-off, the median duration of CR/CRh was 25.9 months. The median duration of overall response was 11.7 months, and the median overall survival was 11.6 months. Of 86 patients who were transfusion dependent at baseline, a 56-day transfusion independence was achieved in 29 (34%), which included patients in all response groups. Grade 3 or 4 treatment-emergent adverse events (≥10%) were febrile neutropenia and anemia (n = 31; 20% each), thrombocytopenia (n = 25; 16%), and neutropenia (n = 20; 13%). Differentiation syndrome adverse events of special interest occurred in 22 (14%) patients, with 14 (9%) grade ≥3 and 1 fatal case reported. Overall, olutasidenib induced durable remissions and transfusion independence with a well-characterized and manageable side effect profile. The observed efficacy represents a therapeutic advance in this molecularly defined, poor-prognostic population of patients with mIDH1 R/R AML. This trial was registered at www.clinicaltrials.gov as #NCT02719574..
Zhu, H.-.
Qin, Y.-.
Zhang, Z.-.
Liu, Y.-.
Wen, L.-.
You, M.J.
Zhang, C.
Such, E.
Luo, H.
Yuan, H.-.
Zhou, H.-.
Liu, H.-.
Xu, R.
Li, J.
Li, J.-.
Hao, J.-.
Jin, J.
Yu, L.
Zhang, J.-.
Liu, L.-.
Zhang, L.-.
Huang, R.-.
Shen, S.-.
Gao, S.-.
Wang, W.
Yan, X.-.
Zhang, X.-.
Du, X.
Chu, X.-.
Yu, Y.-.
Wang, Y.
Mi, Y.-.
Lu, Y.
Cai, Z.
Su, Z.
Taussig, D.C.
MacMahon, S.
Ball, E.D.
Wang, H.-.
Welch, J.S.
Yin, C.C.
Borthakur, G.
Sanz, M.A.
Kantarjian, H.M.
Huang, J.-.
Hu, J.
Chen, S.-.
(2023). A global study for acute myeloid leukemia with RARG rearrangement. Blood adv,
Vol.7
(13),
pp. 2972-2982.
show abstract
Acute myeloid leukemia (AML) with retinoic acid receptor γ (RARG) rearrangement has clinical, morphologic, and immunophenotypic features similar to classic acute promyelocytic leukemia. However, AML with RARG rearrangement is insensitive to alltrans retinoic acid (ATRA) and arsenic trioxide (ATO) and carries a poor prognosis. We initiated a global cooperative study to define the clinicopathological features, genomic and transcriptomic landscape, and outcomes of AML with RARG rearrangements collected from 29 study groups/institutions worldwide. Thirty-four patients with AML with RARG rearrangements were identified. Bleeding or ecchymosis was present in 18 (54.5%) patients. Morphology diagnosed as M3 and M3v accounted for 73.5% and 26.5% of the cases, respectively. Immunophenotyping showed the following characteristics: positive for CD33, CD13, and MPO but negative for CD38, CD11b, CD34, and HLA-DR. Cytogenetics showed normal karyotype in 38% and t(11;12) in 26% of patients. The partner genes of RARG were diverse and included CPSF6, NUP98, HNRNPc, HNRNPm, PML, and NPM1. WT1- and NRAS/KRAS-mutations were common comutations. None of the 34 patients responded to ATRA and/or ATO. Death within 45 days from diagnosis occurred in 10 patients (∼29%). At the last follow-up, 23 patients had died, and the estimated 2-year cumulative incidence of relapse, event-free survival, and overall survival were 68.7%, 26.7%, and 33.5%, respectively. Unsupervised hierarchical clustering using RNA sequencing data from 201 patients with AML showed that 81.8% of the RARG fusion samples clustered together, suggesting a new molecular subtype. RARG rearrangement is a novel entity of AML that confers a poor prognosis. This study is registered with the Chinese Clinical Trial Registry (ChiCTR2200055810)..
Iland, H.J.
Russell, N.
Dillon, R.
Schuh, A.C.
Tedjaseputra, A.
Wei, A.
Khwaja, A.
Knapper, S.
Lane, S.W.
Reynolds, J.
McMullin, M.F.
Martin, A.
Tan, P.
Taussig, D.C.
Wong, A.
Taper, J.
Fraga, C.
Kelly, R.
Tawana, K.
Mehta, P.
Mina, A.
Altman, J.K.
Mølle, I.
Tauro, S.
Tholouli, E.
(2023). Characteristics and outcomes of patients with acute promyelocytic leukemia and extreme hyperleukocytosis at presentation. Blood adv,
Vol.7
(11),
pp. 2580-2585.
Casado, P.
Rio-Machin, A.
Miettinen, J.J.
Bewicke-Copley, F.
Rouault-Pierre, K.
Krizsan, S.
Parsons, A.
Rajeeve, V.
Miraki-Moud, F.
Taussig, D.C.
Bödör, C.
Gribben, J.
Heckman, C.
Fitzgibbon, J.
Cutillas, P.R.
(2023). Integrative phosphoproteomics defines two biologically distinct groups of KMT2A rearranged acute myeloid leukaemia with different drug response phenotypes. Signal transduct target ther,
Vol.8
(1),
p. 80.
show abstract
Acute myeloid leukaemia (AML) patients harbouring certain chromosome abnormalities have particularly adverse prognosis. For these patients, targeted therapies have not yet made a significant clinical impact. To understand the molecular landscape of poor prognosis AML we profiled 74 patients from two different centres (in UK and Finland) at the proteomic, phosphoproteomic and drug response phenotypic levels. These data were complemented with transcriptomics analysis for 39 cases. Data integration highlighted a phosphoproteomics signature that define two biologically distinct groups of KMT2A rearranged leukaemia, which we term MLLGA and MLLGB. MLLGA presented increased DOT1L phosphorylation, HOXA gene expression, CDK1 activity and phosphorylation of proteins involved in RNA metabolism, replication and DNA damage when compared to MLLGB and no KMT2A rearranged samples. MLLGA was particularly sensitive to 15 compounds including genotoxic drugs and inhibitors of mitotic kinases and inosine-5-monosphosphate dehydrogenase (IMPDH) relative to other cases. Intermediate-risk KMT2A-MLLT3 cases were mainly represented in a third group closer to MLLGA than to MLLGB. The expression of IMPDH2 and multiple nucleolar proteins was higher in MLLGA and correlated with the response to IMPDH inhibition in KMT2A rearranged leukaemia, suggesting a role of the nucleolar activity in sensitivity to treatment. In summary, our multilayer molecular profiling of AML with poor prognosis and KMT2A-MLLT3 karyotypes identified a phosphoproteomics signature that defines two biologically and phenotypically distinct groups of KMT2A rearranged leukaemia. These data provide a rationale for the potential development of specific therapies for AML patients characterised by the MLLGA phosphoproteomics signature identified in this study..
Marks, D.I.
Kirkwood, A.A.
Rowntree, C.J.
Aguiar, M.
Bailey, K.E.
Beaton, B.
Cahalin, P.
Castleton, A.Z.
Clifton-Hadley, L.
Copland, M.
Goldstone, A.H.
Kelly, R.
Lawrie, E.
Lee, S.
McMillan, A.K.
McMullin, M.F.
Menne, T.F.
Mitchell, R.J.
Moorman, A.V.
Patel, B.
Patrick, P.
Smith, P.
Taussig, D.
Yallop, D.
Alapi, K.Z.
Fielding, A.K.
(2022). Addition of four doses of rituximab to standard induction chemotherapy in adult patients with precursor B-cell acute lymphoblastic leukaemia (UKALL14): a phase 3, multicentre, randomised controlled trial. Lancet haematol,
Vol.9
(4),
pp. e262-e275.
show abstract
full text
BACKGROUND: Treatment for adults with acute lymphoblastic leukaemia requires improvement. UKALL14 was a UK National Cancer Research Institute Adult ALL group study that aimed to determine the benefit of adding the anti-CD20 monoclonal antibody, rituximab, to the therapy of adults with de novo B-precursor acute lymphoblastic leukaemia. METHODS: This was an investigator-initiated, phase 3, randomised controlled trial done in all UK National Health Service Centres treating patients with acute lymphoblastic leukaemia (65 centres). Patients were aged 25-65 years with de-novo BCR-ABL1-negative acute lymphoblastic leukaemia. Patients with de-novo BCR-ABL1-positive acute lymphoblastic leukaemia were eligible if they were aged 19-65 years. Participants were randomly assigned (1:1) to standard-of-care induction therapy or standard-of-care induction therapy plus four doses of intravenous rituximab (375 mg/m2 on days 3, 10, 17, and 24). Randomisation used minimisation and was stratified by sex, age, and white blood cell count. No masking was used for patients, clinicians, or staff (including the trial statistician), although the central laboratory analysing minimal residual disease and CD20 was masked to treatment allocation. The primary endpoint was event-free survival in the intention-to-treat population. Safety was assessed in all participants who started trial treatment. This study is registered with ClincialTrials.gov, NCT01085617. FINDINGS: Between April 19, 2012, and July 10, 2017, 586 patients were randomly assigned to standard of care (n=292) or standard of care plus rituximab (n=294). Nine patients were excluded from the final analysis due to misdiagnosis (standard of care n=4, standard of care plus rituximab n=5). In the standard-of-care group, median age was 45 years (IQR 22-65), 159 (55%) of 292 participants were male, 128 (44%) were female, one (<1%) was intersex, and 143 (59%) of 244 participants had high-risk cytogenetics. In the standard-of-care plus rituximab group, median age was 46 years (IQR 23-65), 159 (55%) of 294 participants were male, 130 (45%) were female, and 140 (60%) of 235 participants had high-risk cytogenetics. After a median follow-up of 53·7 months (IQR 40·3-70·4), 3-year event-free survival was 43·7% (95% CI 37·8-49·5) for standard of care versus 51·4% (45·4-57·1) for standard of care plus rituximab (hazard ratio [HR] 0·85 [95% CI 0·69-1·06]; p=0·14). The most common adverse events were infections and cytopenias, with no difference between the groups in the rates of adverse events. There were 11 (4%) fatal (grade 5) events in induction phases 1 and 2 in the standard-of-care group and 13 (5%) events in the standard-of-care plus rituximab group). 3-year non-relapse mortality was 23·7% (95% CI 19·0-29·4) in the standard-of-care group versus 20·6% (16·2-25·9) in the standard-of-care plus rituximab group (HR 0·88 [95% CI 0·62-1·26]; p=0·49). INTERPRETATION: Standard of care plus four doses of rituximab did not significantly improve event-free survival over standard of care. Rituximab is beneficial in acute lymphoblastic leukaemia but four doses during induction is likely to be insufficient. FUNDING: Cancer Research UK and Blood Cancer UK..
Zeidan, A.M.
Boss, I.
Beach, C.L.
Copeland, W.B.
Thompson, E.
Fox, B.A.
Hasle, V.E.
Hellmann, A.
Taussig, D.C.
Tormo, M.
Voso, M.T.
Cavenagh, J.
O'Connor, T.
Previtali, A.
Rose, S.
Silverman, L.R.
(2022). A randomized phase 2 trial of azacitidine with or without durvalumab as first-line therapy for older patients with AML. Blood adv,
Vol.6
(7),
pp. 2219-2229.
show abstract
full text
Evidence suggests that combining immunotherapy with hypomethylating agents may enhance antitumor activity. This phase 2 study investigated the activity and safety of durvalumab, a programmed death-ligand 1 (PD-L1) inhibitor, combined with azacitidine for patients aged ≥65 years with acute myeloid leukemia (AML), including analyses to identify biomarkers of treatment response. Patients were randomized to first-line therapy with azacitidine 75 mg/m2 on days 1 through 7 with (Arm A, n = 64) or without (Arm B, n = 65) durvalumab 1500 mg on day 1 every 4 weeks. Overall response rate (complete response [CR] + CR with incomplete blood recovery) was similar in both arms (Arm A, 31.3%; Arm B, 35.4%), as were overall survival (Arm A, 13.0 months; Arm B, 14.4 months) and duration of response (Arm A, 24.6 weeks; Arm B, 51.7 weeks; P = .0765). No new safety signals emerged with combination treatment. The most frequently reported treatment-emergent adverse events were constipation (Arm A, 57.8%; Arm B, 53.2%) and thrombocytopenia (Arm A, 42.2%; Arm B, 45.2%). DNA methylation, mutational status, and PD-L1 expression were not associated with response to treatment. In this study, first-line combination therapy with durvalumab and azacitidine in older patients with AML was feasible but did not improve clinical efficacy compared with azacitidine alone. ClinicalTrials.gov: NCT02775903..
Konopleva, M.Y.
Röllig, C.
Cavenagh, J.
Deeren, D.
Girshova, L.
Krauter, J.
Martinelli, G.
Montesinos, P.
Schäfer, J.A.
Ottmann, O.
Petrini, M.
Pigneux, A.
Rambaldi, A.
Recher, C.
Rodriguez-Veiga, R.
Taussig, D.
Vey, N.
Yoon, S.-.
Ott, M.
Muehlbauer, S.
Beckermann, B.M.
Catalani, O.
Genevray, M.
Mundt, K.
Jamois, C.
Fenaux, P.
Wei, A.H.
(2022). Idasanutlin plus cytarabine in relapsed or refractory acute myeloid leukemia: results of the MIRROS trial. Blood adv,
Vol.6
(14),
pp. 4147-4156.
show abstract
The phase 3 MIRROS (MDM2 antagonist Idasanutlin in Relapsed or Refractory acute myeloid leukemia [AML] for Overall Survival) trial (NCT02545283) evaluated the efficacy and safety of the small-molecule MDM2 antagonist idasanutlin plus cytarabine in patients with relapsed/refractory (R/R) AML. Adults (n = 447) with R/R AML whose disease relapsed or was refractory after ≤2 prior induction regimens as initial treatment or following salvage chemotherapy regimen, with Eastern Cooperative Oncology Group performance status ≤2 were enrolled regardless of TP53 mutation status and randomly assigned 2:1 to idasanutlin 300 mg or placebo orally twice daily plus cytarabine 1 g/m2 IV on days 1 to 5 of 28-day cycles. At primary analysis (cutoff, November 2019), 436 patients were enrolled, including 355 in the TP53 wild-type intention-to-treat (TP53WT-ITT) population. The primary endpoint, overall survival in the TP53WT-ITT population, was not met (median, 8.3 vs 9.1 months with idasanutlin-cytarabine vs placebo-cytarabine; stratified hazard ratio [HR], 1.08; 95% confidence interval [CI], 0.81-1.45; P = .58). The complete remission (CR) rate, a key secondary endpoint, was 20.3% vs 17.1% (odds ratio [OR], 1.23; 95% CI, 0.70-2.18). The overall response rate (ORR) was 38.8% vs 22.0% (OR, 2.25; 95% CI, 1.36-3.72). Common any-grade adverse events (≥10% incidence in any arm) were diarrhea (87.0% vs 32.9%), febrile neutropenia (52.8% vs 49.3%), and nausea (52.5% vs 31.5%). In summary, despite improved ORR, adding idasanutlin to cytarabine did not improve overall survival or CR rates in patients with R/R AML..
Tiong, I.S.
Dillon, R.
Ivey, A.
Teh, T.-.
Nguyen, P.
Cummings, N.
Taussig, D.C.
Latif, A.-.
Potter, N.E.
Runglall, M.
Russell, N.H.
Raj, K.
Schwarer, A.P.
Fong, C.Y.
Grigg, A.P.
Wei, A.H.
(2021). Venetoclax induces rapid elimination of NPM1 mutant measurable residual disease in combination with low-intensity chemotherapy in acute myeloid leukaemia. Br j haematol,
Vol.192
(6),
pp. 1026-1030.
show abstract
full text
Based on promising results in older adults with acute myeloid leukaemia (AML), we treated patients with NPM1mut measurable residual disease (MRD) using off-label venetoclax in combination with low-dose cytarabine or azacitidine. Twelve consecutive patients were retrospectively identified, including five with molecular persistence and seven with molecular relapse/progression. All patients with molecular persistence achieved durable molecular complete remission (CRMRD- ) without transplantation. Six of seven patients with molecular relapse/progression achieved CRMRD- after 1-2 cycles of venetoclax. This paper highlights the promising efficacy of venetoclax-based therapy to reduce the relapse risk in patients with persistent or rising NPM1mut MRD..
Heydt, Q.
Xintaropoulou, C.
Clear, A.
Austin, M.
Pislariu, I.
Miraki-Moud, F.
Cutillas, P.
Korfi, K.
Calaminici, M.
Cawthorn, W.
Suchacki, K.
Nagano, A.
Gribben, J.G.
Smith, M.
Cavenagh, J.D.
Oakervee, H.
Castleton, A.
Taussig, D.
Peck, B.
Wilczynska, A.
McNaughton, L.
Bonnet, D.
Mardakheh, F.
Patel, B.
(2021). Adipocytes disrupt the translational programme of acute lymphoblastic leukaemia to favour tumour survival and persistence. Nat commun,
Vol.12
(1),
p. 5507.
show abstract
full text
The specific niche adaptations that facilitate primary disease and Acute Lymphoblastic Leukaemia (ALL) survival after induction chemotherapy remain unclear. Here, we show that Bone Marrow (BM) adipocytes dynamically evolve during ALL pathogenesis and therapy, transitioning from cellular depletion in the primary leukaemia niche to a fully reconstituted state upon remission induction. Functionally, adipocyte niches elicit a fate switch in ALL cells towards slow-proliferation and cellular quiescence, highlighting the critical contribution of the adipocyte dynamic to disease establishment and chemotherapy resistance. Mechanistically, adipocyte niche interaction targets posttranscriptional networks and suppresses protein biosynthesis in ALL cells. Treatment with general control nonderepressible 2 inhibitor (GCN2ib) alleviates adipocyte-mediated translational repression and rescues ALL cell quiescence thereby significantly reducing the cytoprotective effect of adipocytes against chemotherapy and other extrinsic stressors. These data establish how adipocyte driven restrictions of the ALL proteome benefit ALL tumours, preventing their elimination, and suggest ways to manipulate adipocyte-mediated ALL resistance..
Waclawiczek, A.
Hamilton, A.
Rouault-Pierre, K.
Abarrategi, A.
Albornoz, M.G.
Miraki-Moud, F.
Bah, N.
Gribben, J.
Fitzgibbon, J.
Taussig, D.
Bonnet, D.
(2020). Mesenchymal niche remodeling impairs hematopoiesis via stanniocalcin 1 in acute myeloid leukemia. J clin invest,
Vol.130
(6),
pp. 3038-3050.
show abstract
full text
Acute myeloid leukemia (AML) disrupts the generation of normal blood cells, predisposing patients to hemorrhage, anemia, and infections. Differentiation and proliferation of residual normal hematopoietic stem and progenitor cells (HSPCs) are impeded in AML-infiltrated bone marrow (BM). The underlying mechanisms and interactions of residual hematopoietic stem cells (HSCs) within the leukemic niche are poorly understood, especially in the human context. To mimic AML infiltration and dissect the cellular crosstalk in human BM, we established humanized ex vivo and in vivo niche models comprising AML cells, normal HSPCs, and mesenchymal stromal cells (MSCs). Both models replicated the suppression of phenotypically defined HSPC differentiation without affecting their viability. As occurs in AML patients, the majority of HSPCs were quiescent and showed enrichment of functional HSCs. HSPC suppression was largely dependent on secreted factors produced by transcriptionally remodeled MSCs. Secretome analysis and functional validation revealed MSC-derived stanniocalcin 1 (STC1) and its transcriptional regulator HIF-1α as limiting factors for HSPC proliferation. Abrogation of either STC1 or HIF-1α alleviated HSPC suppression by AML. This study provides a humanized model to study the crosstalk among HSPCs, leukemia, and their MSC niche, and a molecular mechanism whereby AML impairs normal hematopoiesis by remodeling the mesenchymal niche..
Moore, A.S.
Faisal, A.
Mak, G.W.
Miraki-Moud, F.
Bavetsias, V.
Valenti, M.
Box, G.
Hallsworth, A.
de Haven Brandon, A.
Xavier, C.P.
Stronge, R.
Pearson, A.D.
Blagg, J.
Raynaud, F.I.
Chopra, R.
Eccles, S.A.
Taussig, D.C.
Linardopoulos, S.
(2020). Quizartinib-resistant FLT3-ITD acute myeloid leukemia cells are sensitive to the FLT3-Aurora kinase inhibitor CCT241736. Blood adv,
Vol.4
(7),
pp. 1478-1491.
show abstract
full text
Internal tandem duplication of FLT3 (FLT3-ITD) is one of the most common somatic mutations in acute myeloid leukemia (AML); it causes constitutive activation of FLT3 kinase and is associated with high relapse rates and poor survival. Small-molecule inhibition of FLT3 represents an attractive therapeutic strategy for this subtype of AML, although resistance from secondary FLT3 tyrosine kinase domain (FLT3-TKD) mutations is an emerging clinical problem. CCT241736 is an orally bioavailable, selective, and potent dual inhibitor of FLT3 and Aurora kinases. FLT3-ITD+ cells with secondary FLT3-TKD mutations have high in vitro relative resistance to the FLT3 inhibitors quizartinib and sorafenib, but not to CCT241736. The mechanism of action of CCT241736 results in significant in vivo efficacy, with inhibition of tumor growth observed in efficacy studies in FLT3-ITD and FLT3-ITD-TKD human tumor xenograft models. The efficacy of CCT241736 was also confirmed in primary samples from AML patients, including those with quizartinib-resistant disease, which induces apoptosis through inhibition of both FLT3 and Aurora kinases. The unique combination of CCT241736 properties based on robust potency, dual selectivity, and significant in vivo activity indicate that CCT241736 is a bona fide clinical drug candidate for FLT3-ITD and TKD AML patients with resistance to current drugs..
Ghazaly, E.A.
Miraki-Moud, F.
Smith, P.
Gnanaranjan, C.
Koniali, L.
Oke, A.
Saied, M.H.
Petty, R.
Matthews, J.
Stronge, R.
Joel, S.P.
Young, B.D.
Gribben, J.
Taussig, D.C.
(2020). Repression of sphingosine kinase (SK)-interacting protein (SKIP) in acute myeloid leukemia diminishes SK activity and its re-expression restores SK function. J biol chem,
Vol.295
(16),
pp. 5496-5508.
show abstract
full text
Previous studies have shown that sphingosine kinase interacting protein (SKIP) inhibits sphingosine kinase (SK) function in fibroblasts. SK phosphorylates sphingosine producing the potent signaling molecule sphingosine-1-phosphate (S1P). SKIP gene (SPHKAP) expression is silenced by hypermethylation of its promoter in acute myeloid leukemia (AML). However, why SKIP activity is silenced in primary AML cells is unclear. Here, we investigated the consequences of SKIP down-regulation in AML primary cells and the effects of SKIP re-expression in leukemic cell lines. Using targeted ultra-HPLC-tandem MS (UPLC-MS/MS), we measured sphingolipids (including S1P and ceramides) in AML and control cells. Primary AML cells had significantly lower SK activity and intracellular S1P concentrations than control cells, and SKIP-transfected leukemia cell lines exhibited increased SK activity. These findings show that SKIP re-expression enhances SK activity in leukemia cells. Furthermore, other bioactive sphingolipids such as ceramide were also down-regulated in primary AML cells. Of note, SKIP re-expression in leukemia cells increased ceramide levels 2-fold, inactivated the key signaling protein extracellular signal-regulated kinase, and increased apoptosis following serum deprivation or chemotherapy. These results indicate that SKIP down-regulation in AML reduces SK activity and ceramide levels, an effect that ultimately inhibits apoptosis in leukemia cells. The findings of our study contrast with previous results indicating that SKIP inhibits SK function in fibroblasts and therefore challenge the notion that SKIP always inhibits SK activity..
Rio-Machin, A.
Vulliamy, T.
Hug, N.
Walne, A.
Tawana, K.
Cardoso, S.
Ellison, A.
Pontikos, N.
Wang, J.
Tummala, H.
Al Seraihi, A.F.
Alnajar, J.
Bewicke-Copley, F.
Armes, H.
Barnett, M.
Bloor, A.
Bödör, C.
Bowen, D.
Fenaux, P.
Green, A.
Hallahan, A.
Hjorth-Hansen, H.
Hossain, U.
Killick, S.
Lawson, S.
Layton, M.
Male, A.M.
Marsh, J.
Mehta, P.
Mous, R.
Nomdedéu, J.F.
Owen, C.
Pavlu, J.
Payne, E.M.
Protheroe, R.E.
Preudhomme, C.
Pujol-Moix, N.
Renneville, A.
Russell, N.
Saggar, A.
Sciuccati, G.
Taussig, D.
Toze, C.L.
Uyttebroeck, A.
Vandenberghe, P.
Schlegelberger, B.
Ripperger, T.
Steinemann, D.
Wu, J.
Mason, J.
Page, P.
Akiki, S.
Reay, K.
Cavenagh, J.D.
Plagnol, V.
Caceres, J.F.
Fitzgibbon, J.
Dokal, I.
(2020). The complex genetic landscape of familial MDS and AML reveals pathogenic germline variants. Nat commun,
Vol.11
(1),
p. 1044.
show abstract
The inclusion of familial myeloid malignancies as a separate disease entity in the revised WHO classification has renewed efforts to improve the recognition and management of this group of at risk individuals. Here we report a cohort of 86 acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) families with 49 harboring germline variants in 16 previously defined loci (57%). Whole exome sequencing in a further 37 uncharacterized families (43%) allowed us to rationalize 65 new candidate loci, including genes mutated in rare hematological syndromes (ADA, GP6, IL17RA, PRF1 and SEC23B), reported in prior MDS/AML or inherited bone marrow failure series (DNAH9, NAPRT1 and SH2B3) or variants at novel loci (DHX34) that appear specific to inherited forms of myeloid malignancies. Altogether, our series of MDS/AML families offer novel insights into the etiology of myeloid malignancies and provide a framework to prioritize variants for inclusion into routine diagnostics and patient management..
Dillon, R.
Hills, R.
Freeman, S.
Potter, N.
Jovanovic, J.
Ivey, A.
Kanda, A.S.
Runglall, M.
Foot, N.
Valganon, M.
Khwaja, A.
Cavenagh, J.
Smith, M.
Ommen, H.B.
Overgaard, U.M.
Dennis, M.
Knapper, S.
Kaur, H.
Taussig, D.
Mehta, P.
Raj, K.
Novitzky-Basso, I.
Nikolousis, E.
Danby, R.
Krishnamurthy, P.
Hill, K.
Finnegan, D.
Alimam, S.
Hurst, E.
Johnson, P.
Khan, A.
Salim, R.
Craddock, C.
Spearing, R.
Gilkes, A.
Gale, R.
Burnett, A.
Russell, N.H.
Grimwade, D.
(2020). Molecular MRD status and outcome after transplantation in NPM1-mutated AML. Blood,
Vol.135
(9),
pp. 680-688.
show abstract
Relapse remains the most common cause of treatment failure for patients with acute myeloid leukemia (AML) who undergo allogeneic stem cell transplantation (alloSCT), and carries a grave prognosis. Multiple studies have identified the presence of measurable residual disease (MRD) assessed by flow cytometry before alloSCT as a strong predictor of relapse, but it is not clear how these findings apply to patients who test positive in molecular MRD assays, which have far greater sensitivity. We analyzed pretransplant blood and bone marrow samples by reverse-transcription polymerase chain reaction in 107 patients with NPM1-mutant AML enrolled in the UK National Cancer Research Institute AML17 study. After a median follow-up of 4.9 years, patients with negative, low (<200 copies per 105ABL in the peripheral blood and <1000 copies in the bone marrow aspirate), and high levels of MRD had an estimated 2-year overall survival (2y-OS) of 83%, 63%, and 13%, respectively (P < .0001). Focusing on patients with low-level MRD before alloSCT, those with FLT3 internal tandem duplications(ITDs) had significantly poorer outcome (hazard ratio [HR], 6.14; P = .01). Combining these variables was highly prognostic, dividing patients into 2 groups with 2y-OS of 17% and 82% (HR, 13.2; P < .0001). T-depletion was associated with significantly reduced survival both in the entire cohort (2y-OS, 56% vs 96%; HR, 3.24; P = .0005) and in MRD-positive patients (2y-OS, 34% vs 100%; HR, 3.78; P = .003), but there was no significant effect of either conditioning regimen or donor source on outcome. Registered at ISRCTN (http://www.isrctn.com/ISRCTN55675535)..
Potter, N.
Miraki-Moud, F.
Ermini, L.
Titley, I.
Vijayaraghavan, G.
Papaemmanuil, E.
Campbell, P.
Gribben, J.
Taussig, D.
Greaves, M.
(2019). Single cell analysis of clonal architecture in acute myeloid leukaemia. Leukemia,
Vol.33
(5),
pp. 1113-1123.
show abstract
full text
We used single cell Q-PCR on a micro-fluidic platform (Fluidigm) to analyse clonal, genetic architecture and phylogeny in acute myeloid leukaemia (AML) using selected mutations. Ten cases of NPM1c mutant AML were screened for 111 mutations that are recurrent in AML and cancer. Clonal architectures were relatively simple with one to six sub-clones and were branching in some, but not all, patients. NPM1 mutations were secondary or sub-clonal to other driver mutations (DNM3TA, TET2, WT1 and IDH2) in all cases. In three of the ten cases, single cell analysis of enriched CD34+/CD33- cells revealed a putative pre-leukaemic sub-clone, undetectable in the bulk CD33+ population that had one or more driver mutations but lacked NPM1c. Cells from all cases were transplanted into NSG mice and in most (8/10), more than one sub-clone (#2-5 sub-clones) transplanted. However, the dominant regenerating sub-clone in 9/10 cases was NPM1+ and this sub-clone was either dominant or minor in the diagnostic sample from which it was derived. This study provides further evidence, at the single cell level, for genetic variegation in sub-clones and stem cells in acute leukaemia and demonstrates both a preferential order of mutation accrual and parallel evolution of sub-clones..
Morilla, R.
Moss, K.
Nikolova, V.
Marquardt, K.
Duke, S.
Adamowska, K.
Fuller, L.
Taifoor, A.
Johnson, N.
Zeisig, A.
Morilla, A.
Atra, A.
Taussig, D.C.
(2018). The use of unfixed bone marrow trephines for multicolour flow cytometry. Methods,
Vol.134-135,
pp. 80-86.
show abstract
full text
An adequate bone marrow aspirate is essential for a rapid diagnosis of acute leukaemia by multicolour flow cytometry enabling the simultaneous assessment of multiple antigens on the cell surface as well as intracellular or nuclear ones. In the context of acute leukaemia, it is important to have a diagnosis of the blasts lineage as soon as possible to decide the appropriate treatment. This is sometimes delayed due to difficulties in obtaining a bone marrow aspirate due to a "dry tap". In this study we evaluated retrospectively cell markers results by flow cytometry of unfixed bone marrow trephines of 65 patients with leukaemia at diagnosis and including a few after treatment. Our aims were: 1) To compare cell markers results between bone marrow trephine (BMT) and bone marrow aspirate (BMA) 24 cases and BMT with peripheral blood (PB) 14 cases in paired samples to establish if they were reproducible with results of the unfixed bone marrow trephine biopsies. 2) To ascertain a precise diagnosis in 27 (42%) of the cases in which only a bone marrow trephine was available. We demonstrated that unfixed bone marrow trephine provides an adequate and representative cell suspension for flow cytometry and it is a powerful tool when no other material (bone marrow aspirate or peripheral blood) is available to make a rapid diagnosis. Furthermore when marrow aspirate or peripheral blood paired samples were available, flow cytometry results obtained were identical across all the sample types. Applicability to the clinical laboratory: We described a method to obtain a cell suspension from core biopsies that can easily be implemented routinely in a laboratory that performs diagnostic flow cytometry immunophenotyping. This method is simple, inexpensive and it doesn't require extra equipment..
Davies, J.K.
Hassan, S.
Sarker, S.-.
Besley, C.
Oakervee, H.
Smith, M.
Taussig, D.
Gribben, J.G.
Cavenagh, J.D.
(2018). Durable graft-versus-leukaemia effects without donor lymphocyte infusions - results of a phase II study of sequential T-replete allogeneic transplantation for high-risk acute myeloid leukaemia and myelodysplasia. Br j haematol,
Vol.180
(3),
pp. 346-355.
show abstract
Allogeneic haematopoietic stem-cell transplantation remains the only curative treatment for relapsed/refractory acute myeloid leukaemia (AML) and high-risk myelodysplasia but has previously been limited to patients who achieve remission before transplant. New sequential approaches employing T-cell depleted transplantation directly after chemotherapy show promise but are burdened by viral infection and require donor lymphocyte infusions (DLI) to augment donor chimerism and graft-versus-leukaemia effects. T-replete transplantation in sequential approaches could reduce both viral infection and DLI usage. We therefore performed a single-arm prospective Phase II clinical trial of sequential chemotherapy and T-replete transplantation using reduced-intensity conditioning without planned DLI. The primary endpoint was overall survival. Forty-seven patients with relapsed/refractory AML or high-risk myelodysplasia were enrolled; 43 proceeded to transplantation. High levels of donor chimerism were achieved spontaneously with no DLI. Overall survival of transplanted patients was 45% and 33% at 1 and 3 years. Only one patient developed cytomegalovirus disease. Cumulative incidences of treatment-related mortality and relapse were 35% and 20% at 1 year. Patients with relapsed AML and myelodysplasia had the most favourable outcomes. Late-onset graft-versus-host disease protected against relapse. In conclusion, a T-replete sequential transplantation using reduced-intensity conditioning is feasible for relapsed/refractory AML and myelodysplasia and can deliver graft-versus-leukaemia effects without DLI..
Griessinger, E.
Vargaftig, J.
Horswell, S.
Taussig, D.C.
Gribben, J.
Bonnet, D.
(2018). Acute myeloid leukemia xenograft success prediction: Saving time. Exp hematol,
Vol.59,
pp. 66-71.e4.
show abstract
full text
Xenograft assay allows functional analysis of leukemia-initiating cells of acute myeloid leukemia primary samples. However, 40% of samples derived from patients with better outcomes fail to engraft in immunodeficient mouse recipients when conventional protocols are followed. At diagnosis, the engraftment of intermediate-risk group samples cannot be anticipated. In this study, we decided to further explore the reasons for xenograft success and failure. No differences in extracellular phenotype, apoptosis, or cell cycle profile could distinguish samples that engraft (engrafter [E]) from samples that do not engraft (nonengrafter [NE]) in NSG mice. In addition, ex vivo long-term culture assay revealed, after 5 weeks, a lower content of leukemic-LTC-initiating cells in the NE samples associated with a lower expansion rate capacity. One-week co-cultures with mesenchymal or osteoblastic or endothelial cells did not influence the proliferation rate, suggesting that E and NE samples are genuinely rapidly or slowly expanding independent of external cue. Engraftment success for some NE samples was consistently observed in recipient mice analyzed 6 months later than the conventional 3-month period. Eventually we implemented a flow cytometry-based assay, which allowed us to predict, in 1 week, the fast or delayed engraftment potential of a noncharacterized acute myeloid leukemia sample. This approach will be especially useful in selecting intermediate-risk-group patient samples and restricting the experimental duration to a 3-month period and, eventually, in reducing the number of animals and the cost and effort of unnecessary xenograft failures..
Iyengar, S.
Ariza-McNaughton, L.
Clear, A.
Taussig, D.
Auer, R.
Roe, A.
Lillington, D.
Iqbal, S.
Joel, S.
Gribben, J.
Bonnet, D.
(2016). Characteristics of human primary mantle cell lymphoma engraftment in NSG mice. Br j haematol,
Vol.173
(1),
pp. 165-169.
Berthon, C.
Raffoux, E.
Thomas, X.
Vey, N.
Gomez-Roca, C.
Yee, K.
Taussig, D.C.
Rezai, K.
Roumier, C.
Herait, P.
Kahatt, C.
Quesnel, B.
Michallet, M.
Recher, C.
Lokiec, F.
Preudhomme, C.
Dombret, H.
(2016). Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet haematol,
Vol.3
(4),
pp. e186-e195.
show abstract
full text
BACKGROUND: Bromodomain and extraterminal (BET) proteins are chromatin readers that preferentially affect the transcription of genes with super-enhancers, including oncogenes. BET proteins bind acetylated histone tails via their bromodomain, bringing the elongation complex to the promoter region. OTX015 (MK-8628) specifically binds to BRD2, BRD3, and BRD4, preventing BET proteins from binding to the chromatin, thus inhibiting gene transcription. OTX015 inhibits proliferation in many haematological malignancy cell lines and patient cells, in vitro and in vivo. We aimed to establish the recommended dose of OTX015 in patients with haematological malignancies. We report the results of patients with acute leukaemia (leukaemia cohort). METHODS: In this dose-escalation, phase 1 study we recruited patients from seven university hospital centres (in France [five], UK [one], and Canada [one]). Adults with acute leukaemia who had failed or had a contraindication to standard therapies were eligible to participate. OTX015 was given orally at increasing doses from 10 mg/day to 160 mg/day (14 of 21 days), using a conventional 3 + 3 design. In this open-label trial, OTX015 was initially administered once a day, with allowance for exploration of other schedules. The primary endpoint was dose-limiting toxicity (DLT), assessed during the first treatment cycle (21 days). The study is ongoing and is registered with ClinicalTrials.gov, NCT01713582. FINDINGS: Between Jan 18, 2013, and Sept 9, 2014, 41 patients, 36 with acute myeloid leukaemia, a median age of 70 years (IQR 60-75) and two lines of previous therapy, were recruited and treated across six dose levels of OTX015. No DLT was recorded until 160 mg/day, when one patient had grade 3 diarrhoea and another had grade 3 fatigue. However, concomitant grade 1-2 non-DLT toxic effects (ie, gastrointestinal, fatigue, or cutaneous) from 120 mg doses hampered patient compliance and 80 mg once a day was judged the recommended dose with a 14 days on, 7 days off schedule. Common toxic effects for all OTX015 doses were fatigue (including grade 3 in three patients) and bilirubin concentration increases (including grade 3-4 in two patients). OTX015 plasma exposure increased proportionally up to 120 mg/day with trough concentrations in the in-vitro active range from 80 mg/day (274 nmol/L). Three patients (receiving 40 mg/day, 80 mg/day, and 160 mg/day) achieved complete remission or complete remission with incomplete recovery of platelets lasting 2-5 months, and two additional patients had partial blast clearance. No predictive biomarkers for response have been identified so far. INTERPRETATION: The once-daily recommended dose for oral, single agent oral OTX015 use in patients with acute leukaemia for further phase 2 studies is 80 mg on a 14 days on, 7 days off schedule. FUNDING: Oncoethix GmbH, a wholly owned subsidiary of Merck Sharp & Dohme Corp..
Griessinger, E.
Anjos-Afonso, F.
Vargaftig, J.
Taussig, D.C.
Lassailly, F.
Prebet, T.
Imbert, V.
Nebout, M.
Vey, N.
Chabannon, C.
Filby, A.
Bollet-Quivogne, F.
Gribben, J.G.
Peyron, J.-.
Bonnet, D.
(2016). Frequency and Dynamics of Leukemia-Initiating Cells during Short-term Ex Vivo Culture Informs Outcomes in Acute Myeloid Leukemia Patients. Cancer res,
Vol.76
(8),
pp. 2082-2086.
show abstract
Acute myeloid leukemia (AML) is sustained by a subpopulation of rare leukemia-initiating cells (LIC) detected in the xenograft assay by their capacity to self-renew and to generate non-LICs in vivo The xenotransplantation model captures functional properties of LICs that have clinical prognostic value. However, the long duration of this in vivo assay has hampered its use as a prognostic tool. Here, we show, using an ex vivo coculture system, that intermediate and poor risk AML patient samples at diagnosis have a 5 to 7 times higher frequency of leukemic long-term culture-initiating cells (L-LTC-IC) compared with the good risk group. We defined a fluorescence dilution factor (FDF) parameter that monitors sample proliferation over 1 week and established a strong correlation of this parameter with the L-LTC-IC frequency. A higher FDF was found for poor prognostic AMLs or for samples capable of engrafting NSG mice compared with good risk AMLs or nonengrafters. Importantly, FDF could classify normal karyotype intermediate risk patients into two groups with a significant difference in their overall survival, thus making this nongenetic and non-in vivo approach a new clinically relevant tool for better diagnosis of AML patients. Cancer Res; 76(8); 2082-6. ©2016 AACR..
Sud, A.
Morilla, R.
Ethell, M.
Taussig, D.
Liapis, K.
(2015). Green-grey crystals in acute myeloid leukaemia. Br j haematol,
Vol.168
(5),
p. 618.
Miraki-Moud, F.
Ghazaly, E.
Ariza-McNaughton, L.
Hodby, K.A.
Clear, A.
Anjos-Afonso, F.
Liapis, K.
Grantham, M.
Sohrabi, F.
Cavenagh, J.
Bomalaski, J.S.
Gribben, J.G.
Szlosarek, P.W.
Bonnet, D.
Taussig, D.C.
(2015). Arginine deprivation using pegylated arginine deiminase has activity against primary acute myeloid leukemia cells in vivo. Blood,
Vol.125
(26),
pp. 4060-4068.
show abstract
The strategy of enzymatic degradation of amino acids to deprive malignant cells of important nutrients is an established component of induction therapy of acute lymphoblastic leukemia. Here we show that acute myeloid leukemia (AML) cells from most patients with AML are deficient in a critical enzyme required for arginine synthesis, argininosuccinate synthetase-1 (ASS1). Thus, these ASS1-deficient AML cells are dependent on importing extracellular arginine. We therefore investigated the effect of plasma arginine deprivation using pegylated arginine deiminase (ADI-PEG 20) against primary AMLs in a xenograft model and in vitro. ADI-PEG 20 alone induced responses in 19 of 38 AMLs in vitro and 3 of 6 AMLs in vivo, leading to caspase activation in sensitive AMLs. ADI-PEG 20-resistant AMLs showed higher relative expression of ASS1 than sensitive AMLs. This suggests that the resistant AMLs survive by producing arginine through this metabolic pathway and ASS1 expression could be used as a biomarker for response. Sensitive AMLs showed more avid uptake of arginine from the extracellular environment consistent with their auxotrophy for arginine. The combination of ADI-PEG 20 and cytarabine chemotherapy was more effective than either treatment alone resulting in responses in 6 of 6 AMLs tested in vivo. Our data show that arginine deprivation is a reasonable strategy in AML that paves the way for clinical trials..
Liapis, K.
Taussig, D.
Cotter, F.E.
Gribben, J.G.
(2014). Cutaneous cryptococcosis in Hodgkin lymphoma. Br j haematol,
Vol.164
(4),
p. 467.
Griessinger, E.
Anjos-Afonso, F.
Pizzitola, I.
Rouault-Pierre, K.
Vargaftig, J.
Taussig, D.
Gribben, J.
Lassailly, F.
Bonnet, D.
(2014). A niche-like culture system allowing the maintenance of primary human acute myeloid leukemia-initiating cells: a new tool to decipher their chemoresistance and self-renewal mechanisms. Stem cells transl med,
Vol.3
(4),
pp. 520-529.
show abstract
Acute myeloid leukemia-initiating cells (LICs) are responsible for the emergence of leukemia and relapse after chemotherapy. Despite their identification more than 15 years ago, our understanding of the mechanisms responsible for their self-renewal activity and their chemoresistance remains poor. The slow progress in this area is partly due to the difficulty of studying these cells ex vivo. Indeed, current studies are reliant on xenotransplantation assays in immunodeficient mice. In this paper, we report that by modeling key elements of the bone marrow niche using different stromal feeder layers and hypoxic culture conditions, we can maintain LICs over at least 3 weeks and support their self-renewal properties demonstrated through primary and secondary successful xenograft. We provide a proof of principle that this niche-like culture system can be used to study LIC chemoresistance following in vitro cytarabine treatment similarly to the xenograft chemotherapy model. We found that although LICs are believed to be more chemoresistant than non-LICs, functionally defined LICs are not enriched after cytarabine treatment, and heterogeneity in their resistance to treatment can be seen between patients and even within the same patient. We present a culture system that can be used as an in vitro surrogate for xenotransplantation and that has the potential to dramatically increase the throughput of the investigation of LICs. This would further provide the means by which to identify and target the functionality of the different signaling pathways involved in the maintenance and resistance of LICs to improve acute myeloid leukemia treatments..
Liapis, K.
Chung, Y.
Akhtar, W.
Taussig, D.
(2014). A patient with rheumatoid arthritis, cryoglobulinaemia, and an "accidental" finding. Bmj,
Vol.348,
p. g2701.
Manodoro, F.
Marzec, J.
Chaplin, T.
Miraki-Moud, F.
Moravcsik, E.
Jovanovic, J.V.
Wang, J.
Iqbal, S.
Taussig, D.
Grimwade, D.
Gribben, J.G.
Young, B.D.
Debernardi, S.
(2014). Loss of imprinting at the 14q32 domain is associated with microRNA overexpression in acute promyelocytic leukemia. Blood,
Vol.123
(13),
pp. 2066-2074.
show abstract
Distinct patterns of DNA methylation characterize the epigenetic landscape of promyelocytic leukemia/retinoic acid receptor-α (PML-RARα)-associated acute promyelocytic leukemia (APL). We previously reported that the microRNAs (miRNAs) clustered on chromosome 14q32 are overexpressed only in APL. Here, using high-throughput bisulfite sequencing, we identified an APL-associated hypermethylation at the upstream differentially methylated region (DMR), which also included the site motifs for the enhancer blocking protein CCCTC-binding factor (CTCF). Comparing the profiles of diagnostic/remission paired patient samples, we show that hypermethylation was acquired in APL in a monoallelic manner. The cytosine guanine dinucleotide status of the DMR correlated with expression of the miRNAs following a characteristic position-dependent pattern. Moreover, a signature of hypermethylation was also detected in leukemic cells from an established transgenic PML-RARA APL mouse model at the orthologous region on chromosome 12, including the CTCF binding site located upstream from the mouse miRNA cluster. These results, together with the demonstration that the region does not show DNA methylation changes during myeloid differentiation, provide evidence that 14q32 hypermethylation is implicated in the pathogenesis of APL. We propose a model in which loss of imprinting at the 14q32 domain leads to overexpression of the miRNAs in APL..
Anjos-Afonso, F.
Currie, E.
Palmer, H.G.
Foster, K.E.
Taussig, D.C.
Bonnet, D.
(2013). CD34(-) cells at the apex of the human hematopoietic stem cell hierarchy have distinctive cellular and molecular signatures. Cell stem cell,
Vol.13
(2),
pp. 161-174.
show abstract
In addition to well-characterized CD34(+) hematopoietic stem and progenitor cells (HSPCs), the human hematopoietic stem cell (HSC) hierarchy contains a rare CD34(-) population with severe combined immunodeficiency-repopulating capacity. However, little is known about the molecular characteristics of these CD34(-) cells or their relationship to the CD34(+) populations. Here, we show that the self-renewing Lin(-)CD34(-)CD38(-)CD93(hi) population contains cells that not only function as HSCs, but can also be placed above the CD34(+) populations in the hematopoietic hierarchy. These cells have an active Notch pathway, in which signaling through Delta4 is crucial for maintenance of the primitive state, and combined signals from Jagged1 and TGF-β are important in controlling its quiescence. They are also refractory to proliferative signals and show a repressed canonical Wnt pathway, in part regulated by Notch. Overall, therefore, CD34(-) cells represent an immature and quiescent human HSC population maintained through a distinctive network of cellular signaling interactions..
Rouault-Pierre, K.
Lopez-Onieva, L.
Foster, K.
Anjos-Afonso, F.
Lamrissi-Garcia, I.
Serrano-Sanchez, M.
Mitter, R.
Ivanovic, Z.
de Verneuil, H.
Gribben, J.
Taussig, D.
Rezvani, H.R.
Mazurier, F.
Bonnet, D.
(2013). HIF-2α protects human hematopoietic stem/progenitors and acute myeloid leukemic cells from apoptosis induced by endoplasmic reticulum stress. Cell stem cell,
Vol.13
(5),
pp. 549-563.
show abstract
Hematopoietic stem and progenitor cells (HSPCs) are exposed to low levels of oxygen in the bone marrow niche, and hypoxia-inducible factors (HIFs) are the main regulators of cellular responses to oxygen variation. Recent studies using conditional knockout mouse models have unveiled a major role for HIF-1α in the maintenance of murine HSCs; however, the role of HIF-2α is still unclear. Here, we show that knockdown of HIF-2α, and to a much lesser extent HIF-1α, impedes the long-term repopulating ability of human CD34(+) umbilical cord blood cells. HIF-2α-deficient HSPCs display increased production of reactive oxygen species (ROS), which subsequently stimulates endoplasmic reticulum (ER) stress and triggers apoptosis by activation of the unfolded-protein-response (UPR) pathway. HIF-2α deregulation also significantly decreased engraftment ability of human acute myeloid leukemia (AML) cells. Overall, our data demonstrate a key role for HIF-2α in the maintenance of human HSPCs and in the survival of primary AML cells..
Davies, J.K.
Taussig, D.
Oakervee, H.
Smith, M.
Agrawal, S.
Cavenagh, J.D.
Gribben, J.G.
(2013). Long-term survival with low toxicity after allogeneic transplantation for acute myeloid leukaemia and myelodysplasia using non-myeloablative conditioning without T cell depletion. Br j haematol,
Vol.162
(4),
pp. 525-529.
show abstract
The toxicity burden and long-term anti-leukaemic effect of non-myeloablative (NMA) allogeneic haematopoietic stem-cell transplantation (AHSCT) for acute myeloid leukaemia (AML) and myelodysplasia (MDS) remains undefined. We report the outcome of 56 patients with AML/MDS transplanted from human leucocyte antigen-matched donors using NMA conditioning without T-cell depletion. With a median follow-up of 5 years, treatment-related mortality was 9% and current disease-free survival (CDFS) was 45% (overall) and 55% (patients transplanted in remission). Development of graft-versus-host disease upon withdrawal of post-transplant immunosuppression was associated with less relapse and better CDFS. These data confirm that NMA AHSCT without T-cell depletion is safe and can result in sustained remissions of AML/MDS..
Miraki-Moud, F.
Anjos-Afonso, F.
Hodby, K.A.
Griessinger, E.
Rosignoli, G.
Lillington, D.
Jia, L.
Davies, J.K.
Cavenagh, J.
Smith, M.
Oakervee, H.
Agrawal, S.
Gribben, J.G.
Bonnet, D.
Taussig, D.C.
(2013). Acute myeloid leukemia does not deplete normal hematopoietic stem cells but induces cytopenias by impeding their differentiation. Proc natl acad sci u s a,
Vol.110
(33),
pp. 13576-13581.
show abstract
Acute myeloid leukemia (AML) induces bone marrow (BM) failure in patients, predisposing them to life-threatening infections and bleeding. The mechanism by which AML mediates this complication is unknown but one widely accepted explanation is that AML depletes the BM of hematopoietic stem cells (HSCs) through displacement. We sought to investigate how AML affects hematopoiesis by quantifying residual normal hematopoietic subpopulations in the BM of immunodeficient mice transplanted with human AML cells with a range of genetic lesions. The numbers of normal mouse HSCs were preserved whereas normal progenitors and other downstream hematopoietic cells were reduced following transplantation of primary AMLs, findings consistent with a differentiation block at the HSC-progenitor transition, rather than displacement. Once removed from the leukemic environment, residual normal hematopoietic cells differentiated normally and outcompeted steady-state hematopoietic cells, indicating that this effect is reversible. We confirmed the clinical significance of this by ex vivo analysis of normal hematopoietic subpopulations from BM of 16 patients with AML. This analysis demonstrated that the numbers of normal CD34(+)CD38(-) stem-progenitor cells were similar in the BM of AML patients and controls, whereas normal CD34(+)CD38(+) progenitors were reduced. Residual normal CD34(+) cells from patients with AML were enriched in long-term culture, initiating cells and repopulating cells compared with controls. In conclusion the data do not support the idea that BM failure in AML is due to HSC depletion. Rather, AML inhibits production of downstream hematopoietic cells by impeding differentiation at the HSC-progenitor transition..
Vargaftig, J.
Taussig, D.C.
Griessinger, E.
Anjos-Afonso, F.
Lister, T.A.
Cavenagh, J.
Oakervee, H.
Gribben, J.
Bonnet, D.
(2012). Frequency of leukemic initiating cells does not depend on the xenotransplantation model used. Leukemia,
Vol.26
(4),
pp. 858-860.
full text
Joshi, N.M.
Hassan, S.
Jasani, P.
Dixon, S.
Cavenagh, J.D.
Oakervee, H.E.
Smith, M.
Agrawal, S.A.
Auer, R.
de Vos, J.
Langmead, L.
Rampton, D.S.
Gribben, J.G.
Taussig, D.C.
(2012). Bile acid malabsorption in patients with graft-versus-host disease of the gastrointestinal tract. Br j haematol,
Vol.157
(3),
pp. 403-407.
Auer, R.L.
MacDougall, F.
Oakervee, H.E.
Taussig, D.
Davies, J.K.
Syndercombe-Court, D.
Agrawal, S.
Cavenagh, J.D.
Lister, T.A.
Gribben, J.G.
(2012). T-cell replete fludarabine/cyclophosphamide reduced intensity allogeneic stem cell transplantation for lymphoid malignancies. Br j haematol,
Vol.157
(5),
pp. 580-585.
show abstract
The relative merits of reduced-intensity allogeneic stem cell transplantation (RISCT) for high-risk indolent lymphoid malignancies are emerging, although the preferred conditioning regimen to manage the risks of graft-versus-host disease (GVHD) is not clearly defined. Here we report the outcome of 73 patients with lymphoid malignancies who received RISCT with a fludarabine/cyclophosphosphamide conditioning regimen and a median follow-up of 3 years. Median age was 54 years. Forty-eight per cent of patients had previously undergone autologous stem cell transplantation with a median of three prior therapies. Non-relapse mortality at 3 years was 19% but only 5% for patients with multiple myeloma (MM). Three-year overall survival and current progression-free survival was 67% and 63% respectively. Grade 2-4 acute GVHD occurred in 14% of patients while 49% had chronic GVHD requiring systemic immunosuppression. The preparatory regimen in this study has the advantage of reduced acute GVHD and low mortality, notably in patients with MM. In addition, this strategy provides long-term disease control in a significant proportion of patients with particular benefit in those with high-risk follicular lymphoma..
Szlosarek, P.W.
Luong, F.
Clear, A.
Taussig, D.
Joel, S.
Calaminici, M.
Debernardi, S.
Fitzgibbon, J.
Bomalaski, J.S.
Frankel, A.E.
Bonnet, D.
(2011). Pegylated arginine deiminase (ADI-PEG20) as a potential novel therapy for argininosuccinate synthetase-deficient acute myeloid leukemia. Cancer research,
Vol.71.
Farren, T.W.
Giustiniani, J.
Liu, F.-.
Tsitsikas, D.A.
Macey, M.G.
Cavenagh, J.D.
Oakervee, H.E.
Taussig, D.
Newland, A.C.
Calaminici, M.
Bensussan, A.
Jenner, M.
Gribben, J.G.
Agrawal, S.G.
(2011). Differential and tumor-specific expression of CD160 in B-cell malignancies. Blood,
Vol.118
(8),
pp. 2174-2183.
show abstract
CD160 is a human natural killer (NK)-cell-activating receptor that is also expressed on T-cell subsets. In the present study, we examined 811 consecutive cases of B-cell lymphoproliferative disorders (B-LPDs), and demonstrated CD160 expression in 98% (590 of 600) of chronic lymphocytic leukemia (CLL) cases, 100% (32 of 32) of hairy cell leukemia (HCL) cases, 15% (5 of 34) of mantle cell lymphoma (MCL) in the leukemic phase, and 16% (23 of 145) of other B-LPD cases. CD160 transcript and protein were absent in the normal B-cell hierarchy, from stem cells, B-cell precursors, maturing B cells in the germinal center, and circulating B cells, including CD5(+)CD19(+) B1 cells in umbilical cord. CD160 positivity was significantly higher in CLL and HCL in terms of percentage (65.9% and 67.8%, respectively, P < .0001) and median fluorescence intensity (552 and 857, respectively, P < .0001) compared with all other B-LPD cases. Lymph node CLL samples were also CD160(+). Using the disease-specific expression of CD5, CD23, and CD160, a score of 3 characterized CLL (diagnostic odds ratio, 1430); a score of 0 excluded CLL, MCL, and HCL; and the CD23/CD5 ratio differentiated CLL from leukemic CD23(+) MCL. In the B-cell lineage, CD160 is a tumor-specific antigen known to mediate cellular activation signals in CLL, and is a novel target for therapeutic manipulation and monitoring of minimal residual disease..
Taussig, D.C.
Vargaftig, J.
Miraki-Moud, F.
Griessinger, E.
Sharrock, K.
Luke, T.
Lillington, D.
Oakervee, H.
Cavenagh, J.
Agrawal, S.G.
Lister, T.A.
Gribben, J.G.
Bonnet, D.
(2010). Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood,
Vol.115
(10),
pp. 1976-1984.
show abstract
Leukemia-initiating cells (LICs) in acute myeloid leukemia (AML) are believed to be restricted to the CD34(+) fraction. However, one of the most frequently mutated genes in AML is nucleophosmin (NPM), and this is associated with low CD34 expression. We, therefore, investigated whether NPM-mutated AMLs have LICs restricted to the CD34(+) fraction. We transplanted sorted fractions of primary NPM-mutated AML into immunodeficient mice to establish which fractions initiate leukemia. Approximately one-half of cases had LICs exclusively within the CD34(-) fraction, whereas the CD34(+) fraction contained normal multilineage hematopoietic repopulating cells. Most of the remaining cases had LICs in both CD34(+) and CD34(-) fractions. When samples were sorted based on CD34 and CD38 expression, multiple fractions initiated leukemia in primary and secondary recipients. The data indicate that the phenotype of LICs is more heterogeneous than previously realized and can vary even within a single sample. This feature of LICs may make them particularly difficult to eradicate using therapies targeted against surface antigens..
Anjos-Afonso, F.
Taussig, D.
Bonnet, D.
(2009). PERTURBED BONE MARROW MESENCHYMAL STROMAL (MSC) COMPARTMENT IN AML PATIENTS: IMPLICATION FOR UNDERSTANDING LEUKEMIC MICROENVIRONMENT. Experimental hematology,
Vol.37
(9),
pp. S52-S53.
Le Dieu, R.
Taussig, D.C.
Ramsay, A.G.
Mitter, R.
Miraki-Moud, F.
Fatah, R.
Lee, A.M.
Lister, T.A.
Gribben, J.G.
(2009). Peripheral blood T cells in acute myeloid leukemia (AML) patients at diagnosis have abnormal phenotype and genotype and form defective immune synapses with AML blasts. Blood,
Vol.114
(18),
pp. 3909-3916.
show abstract
Understanding how the immune system in patients with cancer interacts with malignant cells is critical for the development of successful immunotherapeutic strategies. We studied peripheral blood from newly diagnosed patients with acute myeloid leukemia (AML) to assess the impact of this disease on the patients' T cells. The absolute number of peripheral blood T cells is increased in AML compared with healthy controls. An increase in the absolute number of CD3+56+ cells was also noted. Gene expression profiling on T cells from AML patients compared with healthy donors demonstrated global differences in transcription suggesting aberrant T-cell activation patterns. These gene expression changes differ from those observed in chronic lymphocytic leukemia (CLL), indicating the heterogeneous means by which different tumors evade the host immune response. However, in common with CLL, differentially regulated genes involved in actin cytoskeletal formation were identified, and therefore the ability of T cells from AML patients to form immunologic synapses was assessed. Although AML T cells could form conjugates with autologous blasts, their ability to form immune synapses and recruit phosphotyrosine signaling molecules to the synapse was significantly impaired. These findings identify T-cell dysfunction in AML that may contribute to the failure of a host immune response against leukemic blasts..
Le Dieu, R.
Taussig, D.
Lister, T.A.
Gribben, J.G.
(2009). Negative immunomagnetic selection of T cells from peripheral blood of presentation AML specimens. J immunol methods,
Vol.348
(1-2),
pp. 95-100.
show abstract
To date, studies on T cells in acute myeloid leukemia (AML) have been limited to flow cytometric analysis of whole peripheral blood mononuclear cell (PBMC) specimens or functional work looking at the impact of AML myeloblasts on normal or remission T cells. This lack of information on T cells at the time of presentation with disease is due in part to the difficulty in isolating sufficiently pure T cells from these specimens for further study. Negative immunomagnetic selection has been the method of choice for isolating immune cells for functional studies due to concerns that binding antibodies to the cell surface may induce cellular activation, block ligand-receptor interactions or result in immune clearance. In order specifically to study T cells in presentation AML specimens, we set out to develop a method of isolating highly pure CD4 and CD8 T cells by negative selection from the peripheral blood (PB) of newly diagnosed AML patients. This technique, unlike T cell selection from PB from normal individuals or from patients with chronic lymphocytic leukaemia, was extremely problematic due to properties of the leukaemic myeloblasts. A successful method was eventually optimized requiring the use of a custom antibody cocktail consisting of CD33, CD34, CD123, CD11c and CD36, to deplete myeloblasts..
Taussig, D.C.
Miraki-Moud, F.
Anjos-Afonso, F.
Pearce, D.J.
Allen, K.
Ridler, C.
Lillington, D.
Oakervee, H.
Cavenagh, J.
Agrawal, S.G.
Lister, T.A.
Gribben, J.G.
Bonnet, D.
(2008). Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells. Blood,
Vol.112
(3),
pp. 568-575.
show abstract
Immunodeficient mice are increasingly used to assay human hematopoietic repopulating cells as well as leukemia-initiating cells. One method commonly used to isolate these rare cells is to sort cells stained with fluorochrome-conjugated antibodies into fractions, then transplant the different fractions into immunodeficient mice to test their repopulating ability. The antibodies are generally treated as being neutral in terms of their effects on the experiment. Human repopulating cells are thought to express CD34 and lack CD38. Here we present evidence that anti-CD38 antibodies have a profound inhibitory effect on engraftment of cord blood and leukemia cells. We show that this effect is Fc-mediated and can be overcome by treating mice with immunosuppressive antibodies. When this inhibitory effect is prevented, we demonstrate that the CD34(+)CD38(+) fraction of certain acute myeloid leukemia samples contains all, or at least most, leukemia-initiating cell capacity. This study highlights the potential pitfall of antibody-mediated clearance of repopulating cells and is important for any groups working with this model. More importantly, the work suggests that there is greater variation in the phenotypes of leukemia-initiating cells than previously suggested..
Le Dieu, R.
Ramsay, A.G.
Taussig, D.
Mitter, R.
Gribben, J.G.
(2008). Differential Gene Expression Profile Identifies the Nature of T Cell Defects in AML Patients at Diagnosis. Blood,
Vol.112
(11),
pp. 433-433.
Pearce, D.J.
Taussig, D.
Zibara, K.
Smith, L.-.
Ridler, C.M.
Preudhomme, C.
Young, B.D.
Rohatiner, A.Z.
Lister, T.A.
Bonnet, D.
(2006). AML engraftment in the NOD/SCID assay reflects the outcome of AML: implications for our understanding of the heterogeneity of AML. Blood,
Vol.107
(3),
pp. 1166-1173.
show abstract
The nonobese diabetic/severe combined immunodeficient (NOD/SCID) assay is the current model for assessment of human normal and leukemic stem cells. We explored why 51% of 59 acute myeloid leukemia (AML) patients were unable to initiate leukemia in NOD/SCID mice. Increasing the cell dose, using more permissive recipients, and alternative tissue sources did not cause AML engraftment in most previously nonengrafting AML samples. Homing of AML cells to the marrow was the same between engrafters and nonengrafters. FLT3 internal tandem duplication (ITD) and nucleophosmin mutations occurred at a similar frequency in engrafters and nonengrafters. The only variable that was related to engraftment ability was the karyotypically defined risk stratification of individual AML cases. Of interest, follow-up of younger patients with intermediate-risk AML revealed a significant difference in overall survival between NOD/SCID engrafting and nonengrafting AMLs. Hence, the ability of AML to engraft in the NOD/SCID assay seems to be an inherent property of AML cells, independent of homing, conditioning, or cell frequency/source, which is directly related to prognosis. Our results suggest an important difference between leukemic initiating cells between engrafting and nonengrafting AML cases that correlates with treatment response..
Pearce, D.J.
Taussig, D.C.
Bonnet, D.
(2006). Implications of the expression of myeloid markers on normal and leukemic stem cells. Cell cycle,
Vol.5
(3),
pp. 271-273.
show abstract
We have recently described the expression of CD33 and other antigens previously thought to be myeloid specific on both acute myeloid leukemia-initiating cells (AML-IC) and normal hematopoietic stem cells (HSC) that are capable of repopulating immuno-deficient mice. Here, we discuss that the presence of myeloid markers on AML-ICs and HSCs makes identification of an antigen for targeted therapy extremely difficult. Xenotransplantation assays should be used wherever possible to test the suitability of candidate antigens for targeted therapy, including the assessment of the effect on normal cells..
Davies, J.K.
Taussig, D.C.
Oakervee, H.
Davies, A.J.
Agrawal, S.G.
Gribben, J.G.
Lister, T.A.
Cavenagh, J.D.
(2006). Long-term follow-up after reduced-intensity conditioning allogeneic transplantation for acute myeloid leukemia/myelodysplastic syndrome: late CNS relapses despite graft-versus-host disease. J clin oncol,
Vol.24
(14),
pp. e23-e25.
Taussig, D.C.
Pearce, D.J.
Simpson, C.
Rohatiner, A.Z.
Lister, T.A.
Kelly, G.
Luongo, J.L.
Danet-Desnoyers, G.-.
Bonnet, D.
(2005). Hematopoietic stem cells express multiple myeloid markers: implications for the origin and targeted therapy of acute myeloid leukemia. Blood,
Vol.106
(13),
pp. 4086-4092.
show abstract
Human hematopoietic stem cells (HSCs) are generally regarded as being devoid of the markers expressed by differentiated blood cells, the lineage-specific antigens. However, recent work suggests that genes associated with the myeloid lineage are transcribed in mouse HSCs. Here, we explore whether myeloid genes are actually translated in human HSCs. We show that CD33, CD13, and CD123, well-established myeloid markers, are expressed on human long-term repopulating cells from cord blood and bone marrow. In addition, we demonstrate that nonobese diabetic/severe combined immunodeficiency (NOD/SCID) leukemia-initiating cells (SL-ICs) are restricted to the CD33+ fraction in 11 of 12 acute myeloid leukemia (AML) samples studied, indicating that leukemic stem cells (LSCs) express this antigen. This study changes our view of HSCs and the process of differentiation. Furthermore, based on the phenotypic similarity of HSCs and LSCs, our data provide support for the hypothesis that AML derives from an HSC. Our findings also provide a challenge to contemporary attempts to improve the outcome of AML using myeloid antigen-targeted therapies, given the potential for HSC killing..
Pearce, D.J.
Taussig, D.
Simpson, C.
Allen, K.
Rohatiner, A.Z.
Lister, T.A.
Bonnet, D.
(2005). Characterization of cells with a high aldehyde dehydrogenase activity from cord blood and acute myeloid leukemia samples. Stem cells,
Vol.23
(6),
pp. 752-760.
show abstract
Aldehyde dehydrogenase (ALDH) is a cytosolic enzyme that is responsible for the oxidation of intracellular aldehydes. Elevated levels of ALDH have been demonstrated in murine and human progenitor cells compared with other hematopoietic cells, and this is thought to be important in chemoresistance. A method for the assessment of ALDH activity in viable cells recently has been developed and made commercially available in a kit format. In this study, we confirmed the use of the ALDH substrate kit to identify cord blood stem/progenitor cells. Via multicolor flow cytometry of cord blood ALDH+ cells, we have expanded on their phenotypic analysis. We then assessed the incidence, morphology, phenotype, and nonobese diabetic/ severe combined immunodeficiency engraftment ability of ALDH+ cells from acute myeloid leukemia (AML) samples. AML samples had no ALDH+ cells at all, an extremely rare nonmalignant stem/progenitor cell population, or a less rare, leukemic stem cell population. Hence, in addition to identifying nonmalignant stem cells within some AML samples, a high ALDH activity also identifies some patients' CD34+/ CD38- leukemic stem cells. The incidence of normal or leukemic stem cells with an extremely high ALDH activity may have important implications for resistance to chemotherapy. Identification and isolation of leukemic cells on the basis of ALDH activity provides a tool for their isolation and further analysis..
Taussig, D.C.
Davies, A.J.
Cavenagh, J.D.
Oakervee, H.
Syndercombe-Court, D.
Kelsey, S.
Amess, J.A.
Rohatiner, A.Z.
Lister, T.A.
Barnett, M.J.
(2003). Durable remissions of myelodysplastic syndrome and acute myeloid leukemia after reduced-intensity allografting. J clin oncol,
Vol.21
(16),
pp. 3060-3065.
show abstract
PURPOSE: To evaluate the use of reduced-intensity (RI) conditioning with allogeneic hematopoietic stem cell transplantation (HSCT) from HLA-identical family donors in patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). PATIENTS AND METHODS: Sixteen patients (median age, 54 years; range, 37 to 66 years) underwent RI-HSCT using a conditioning regimen of fludarabine 25 mg/m2 daily for 5 days and either cyclophosphamide 1 g/m2 daily for 2 days (14 patients) or melphalan 140 mg/m2 for 1 day (two patients). The median number of CD34+ cells and CD3+ cells infused per kilogram of recipient weight was 4.5 x 106 (range, 1.8 to 7.3 x 106 cells) and 2.9 x 108 (range, 0.1 to 9.6 x 108 cells), respectively. RESULTS: There was no transplant-related mortality (TRM) within 100 days of HSCT. Grade 1 to 2 acute graft-versus-host disease (GVHD) occurred in three patients, but neither grade 3 nor grade 4 disease was observed. Chronic GVHD occurred in 10 patients. One patient had cytomegalovirus (CMV) reactivation but did not develop CMV disease. With a median follow-up of 26 months (range, 15 to 45 months), 11 patients are alive (nine in continuous complete remission and one in complete remission after a second transplantation), and five have died (four from disease progression and one from bone-marrow aplasia induced by cyclosporine withdrawal). The 2-year actuarial overall and event-free survival rates were 69% (95% confidence interval [CI], 40% to 86%) and 56% (95% CI, 30% to 68%), respectively. CONCLUSION: This strategy of RI-HSCT resulted in reliable engraftment with low incidence of acute GVHD and TRM. Durable remissions were observed in patients with MDS and AML consistent with a graft-versus-leukemia effect..
Longhurst, H.J.
Taussig, D.
Haque, T.
Syndercombe-Court, D.
Cavenagh, J.
Edgar, J.D.
Helbert, M.R.
(2002). Non-myeloablative bone marrow transplantation in an adult with Wiskott-Aldrich syndrome. Br j haematol,
Vol.116
(2),
pp. 497-499.
show abstract
Early bone marrow transplant is now standard treatment for infants with severe immunodeficiencies such as Wiskott-Aldrich Syndrome (WAS), but results in older children and adults are poor. Non-myeloablative transplant has shown promise in the treatment of older children, who are likely to have active infections and organ damage. We describe a non-myeloablative transplant of a 26-year-old man with WAS, undertaken because of severe infections and vasculitis. Partial engraftment and immunorestoration were achieved. The patient is well 1 year post transplantation..
Allen, D.
Ng, S.
Beaton, K.
Taussig, D.
(2002). Sternal osteomyelitis caused by Aspergillus fumigatus in a patient with previously treated Hodgkin's disease. J clin pathol,
Vol.55
(8),
pp. 616-618.
show abstract
This report details the case of a 67 year old woman with sternal osteomyelitis caused by Aspergillus fumigatus. She was diagnosed with Hodgkin's disease in 1975 and was successfully treated with chemotherapy. A lobectomy for recurrence localised to the left lung was complicated nine years later by severe bronchiectasis, for which she required a total left sided pneumonectomy. At surgery, a non-invasive aspergillus was found. She presented eight years later with symptoms that were initially attributed to recurrence of Hodgkins's disease, but on investigation were found to be caused by fungal sternal osteomyelitis. Treatment with itraconazole suspension at a dose of 400 mg daily was successful..
Taussig, D.C.
Pearce, D.J.
Bonnet, D.A.
(2002). Male cells in female recipients of hematopoietic-cell transplants. N engl j med,
Vol.347
(3),
pp. 218-220.
Taussig, D.C.
Pearce, D.J.
Bonnet, D.A.
(2002). Male cells in female recipients of hematopoietic-cell transplants. New england journal of medicine,
Vol.347
(3),
pp. 219-219.
Foran, J.M.
Norton, A.J.
Micallef, I.N.
Taussig, D.C.
Amess, J.A.
Rohatiner, A.Z.
Lister, T.A.
(2001). Loss of CD20 expression following treatment with rituximab (chimaeric monoclonal anti-CD20): a retrospective cohort analysis. Br j haematol,
Vol.114
(4),
pp. 881-883.
show abstract
A retrospective analysis of CD20 expression following rituximab for B-cell non-Hodgkin's lymphoma demonstrated a significant change in immunophenotype in 6/25 (24%) patients with persistent bone marrow (BM) infiltration. In three out of six patients, the B cells were uniformly CD20-/CD79alpha+, consistent with frank loss of CD20 expression. In the remaining three cases, the BM infiltrate was predominantly (> 80%) CD20-/CD79alpha+. Two of the former but none of the latter three cases achieved a clinical response. In three further cases, the post-treatment BM infiltrate was composed entirely of benign or reactive CD3+ T cells. Frank loss of CD20 was not seen in 25 post-treatment lymph node biopsies. Immunophenotyping is therefore an important adjunct in the diagnosis of BM infiltration following rituximab..
Taussig, D.
Williams, M.A.
Wiggins, C.
Feakins, R.M.
McBride, N.
Newland, A.C.
Kelsey, S.M.
(1998). Clinical Regression and Remission of Primary Refractory Angiocentric Lymphoma Following Autologous Tumour Peptide Antigen-charged Dendritic Cells After High-dose Chemotherapy and Autologous Stem Cell Rescue. Hematology,
Vol.3
(4),
pp. 277-289.
show abstract
We report on the case of a 34-year old man with a previously refractory high-grade non-Hodgkin's Lymphoma which regressed following dendritic cell based immunotherapy after high-dose chemotherapy. Antigen presenting cells known as dendritic cells were cultured from harvested autologous peripheral blood progenitor cells. The dendritic cells were then exposed to unfractionated tumour peptides derived from a skin biopsy section inflitrated with lymphoma. These charged dendritic cells infused into the patient together with autologous T-lymphocytes after high-dose chemotherapy with peripheral blood stem cell rescue. Although the lymphoma relapsed after the high-dose chemotherapy it regressed again following the dendritic cell infusion. The patient's T-lymphocytes demonstrated in vitro reactivity to tumour peptides whereas the lymphocytes of controls did not. We propose that the lymphoma regression occurred because of a T cell mediated response against the tumour induced by the charged dendritic cells..
Ho, T.
Taussig, D.
McAnulty, G.
(1997). Effect of atenolol on mortality and cardiovascular morbidity after noncardiac surgery. N engl j med,
Vol.336
(20),
p. 1452.