Network


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

Hotspot


Dive into the research topics where Heather Oakervee is active.

Publication


Featured researches published by Heather Oakervee.


The New England Journal of Medicine | 2015

Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma

Abstr Act; Sagar Lonial; Meletios A. Dimopoulos; Antonio Palumbo; Darrell White; Sebastian Grosicki; Ivan Spicka; Adam Walter‑Croneck; Philippe Moreau; Maria Victoria Mateos; Hila Magen; Andrew R. Belch; Donna Reece; Meral Beksac; Andrew Spencer; Heather Oakervee; Robert Z. Orlowski; Masafumi Taniwaki; Christoph Röllig; Hermann Einsele; Ka Lung Wu; Anil Singhal; Jesús F. San Miguel; Morio Matsumoto; Jessica Katz; Eric Bleickardt; Valerie Poulart; Kenneth C. Anderson; Paul G. Richardson

BACKGROUND Elotuzumab, an immunostimulatory monoclonal antibody targeting signaling lymphocytic activation molecule F7 (SLAMF7), showed activity in combination with lenalidomide and dexamethasone in a phase 1b-2 study in patients with relapsed or refractory multiple myeloma. METHODS In this phase 3 study, we randomly assigned patients to receive either elotuzumab plus lenalidomide and dexamethasone (elotuzumab group) or lenalidomide and dexamethasone alone (control group). Coprimary end points were progression-free survival and the overall response rate. Final results for the coprimary end points are reported on the basis of a planned interim analysis of progression-free survival. RESULTS Overall, 321 patients were assigned to the elotuzumab group and 325 to the control group. After a median follow-up of 24.5 months, the rate of progression-free survival at 1 year in the elotuzumab group was 68%, as compared with 57% in the control group; at 2 years, the rates were 41% and 27%, respectively. Median progression-free survival in the elotuzumab group was 19.4 months, versus 14.9 months in the control group (hazard ratio for progression or death in the elotuzumab group, 0.70; 95% confidence interval, 0.57 to 0.85; P<0.001). The overall response rate in the elotuzumab group was 79%, versus 66% in the control group (P<0.001). Common grade 3 or 4 adverse events in the two groups were lymphocytopenia, neutropenia, fatigue, and pneumonia. Infusion reactions occurred in 33 patients (10%) in the elotuzumab group and were grade 1 or 2 in 29 patients. CONCLUSIONS Patients with relapsed or refractory multiple myeloma who received a combination of elotuzumab, lenalidomide, and dexamethasone had a significant relative reduction of 30% in the risk of disease progression or death. (Funded by Bristol-Myers Squibb and AbbVie Biotherapeutics; ELOQUENT-2 ClinicalTrials.gov number, NCT01239797.).


Blood | 2008

Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells

David Taussig; F Miraki-Moud; Fernando Anjos-Afonso; Daniel J. Pearce; Kirsty Allen; Colin Ridler; Debra M. Lillington; Heather Oakervee; Jamie Cavenagh; Samir G. Agrawal; T. A. Lister; John G. Gribben; Dominique Bonnet

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.


British Journal of Haematology | 2005

PAD combination therapy (PS‐341/bortezomib, doxorubicin and dexamethasone) for previously untreated patients with multiple myeloma

Heather Oakervee; Rakesh Popat; Nicola Curry; Pat Smith; Curly Morris; Mary Drake; Samir G. Agrawal; Jim Stec; David P. Schenkein; Dixie-Lee Esseltine; Jamie Cavenagh

Bortezomib (formerly PS‐341) has significant activity in patients with relapsed multiple myeloma (MM), its efficacy is increased with the addition of dexamethasone and it demonstrates synergy with doxorubicin, thus providing the rationale for combination therapy with bortezomib, doxorubicin and dexamethasone (PAD). Patients with untreated MM received four 21‐d cycles of PAD, comprising bortezomib 1·3 mg/m2 on days 1, 4, 8 and 11, along with dexamethasone 40 mg on days 1–4, 8–11 and 15–18 during cycle 1 and days 1–4 during cycles 2–4. During days 1–4, patients also received 0, 4·5 or 9 mg/m2 of doxorubicin at dose levels 1, 2, and 3 respectively. Following peripheral blood stem cell (PBSC) collection, patients received high‐dose melphalan (MEL200) with PBSC transplantation (PBSCT). After PAD induction alone, 20 of 21 patients (95%) achieved at least a partial response (PR), including complete response (CR) in five patients (24%). Twenty of 21 had PBSC mobilized, and 18 of 20 received MEL200/PBSCT. In an intention‐to‐treat analysis, response rates were: CR 43%, near CR 14%, very good PR 24%, PR 14% and stable disease 5%. PAD was effective, did not prejudice subsequent PBSC collection, and should be further evaluated in prospective randomized trials.


Blood | 2010

Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34− fraction

David Taussig; Jacques Vargaftig; Farideh Miraki-Moud; Emmanuel Griessinger; Kirsty Sharrock; Tina Luke; Debra M. Lillington; Heather Oakervee; Jamie Cavenagh; Samir G. Agrawal; T. Andrew Lister; John G. Gribben; Dominique Bonnet

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.


British Journal of Haematology | 2008

Bortezomib, doxorubicin and dexamethasone (PAD) front‐line treatment of multiple myeloma: updated results after long‐term follow‐up

Rakesh Popat; Heather Oakervee; Simon Hallam; Nicola Curry; Liz Odeh; Nicola Foot; Dixie-Lee Esseltine; Mary Drake; Curly Morris; Jamie Cavenagh

Bortezomib, doxorubicin and dexamethasone (PAD) was evaluated as induction before stem cell transplantation in newly diagnosed multiple myeloma (MM) patients, using bortezomib 1·3 mg/m2 (PAD1, N = 21) or 1·0 mg/m2 (PAD2, N = 20). Complete/very good partial response rates with PAD1/PAD2 were 62%/42% postinduction and 81%/53% post‐transplant. Progression‐free survival (29 vs. 24 months), time to re‐treatment (36 vs. 29 months) and overall survival (1 year: 100% vs. 95%; 2 years: 95% vs. 73%) were statistically similar but favoured PAD1 versus PAD2. Toxicity was lower in PAD2; bortezomib dose reduction may help manage toxicities while retaining efficacy. PAD is highly active as front‐line induction in MM.


British Journal of Haematology | 2003

Thalidomide in multiple myeloma: current status and future prospects

Jamie Cavenagh; Heather Oakervee

Thalidomide was introduced in Europe during the 1950s as a sedative agent and was found to be particularly effective at alleviating the symptoms of morning sickness. As is widely known, it was subsequently withdrawn in 1961 after its teratogenic properties were recognized, in particular, its ability to cause phocomelia. Damage to the fetus occurs early in pregnancy (from d 28–42 post conception) and the limb bud abnormalities are thought to be due to inhibition of normal vessel formation. Since that time, thalidomide’s immunomodulatory effects have been recognized and it has been used, in a cautious and restricted manner, in the treatment of such disparate disorders as Behcet’s disease, erythema nodosum leprosum, human immunodeficiency virus-associated oral ulceration and chronic graft-versus-host disease. More recently, the drug has been tested in a variety of solid and haematological malignancies, and has shown remarkable efficacy in patients with advanced multiple myeloma. Indeed, the use of thalidomide is arguably the most significant advance in the treatment of myeloma since the introduction of highdose melphalan and autologous stem cell transplantation nearly 20 years ago.


Journal of Clinical Oncology | 2003

Durable Remissions of Myelodysplastic Syndrome and Acute Myeloid Leukemia After Reduced-Intensity Allografting

David Taussig; Andrew Davies; Jamie Cavenagh; Heather Oakervee; Denise Syndercombe-Court; S Kelsey; J. Amess; A. Z. S. Rohatiner; T. A. Lister; M. J. Barnett

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.


British Journal of Haematology | 2013

A phase 2 study of SRT501 (resveratrol) with bortezomib for patients with relapsed and or refractory multiple myeloma

Rakesh Popat; Torben Plesner; Faith E. Davies; Gordon Cook; Mark Cook; Peter J Elliott; Eric Jacobson; Todd Gumbleton; Heather Oakervee; Jamie Cavenagh

Bagnara, D., Kaufman, M.S., Calissano, C., Marsilio, S., Patten, P.E., Simone, R., Chum, P., Yan, X.J., Allen, S.L., Kolitz, J.E., Baskar, S., Rader, C., Mellstedt, H., Rabbani, H., Lee, A., Gregersen, P.K., Rai, K.R. & Chiorazzi, N. (2011) A novel adoptive transfer model of chronic lymphocytic leukemia suggests a key role for T lymphocytes in the disease. Blood, 117, 5463–5472. Brachtl, G., Sahakyan, K., Denk, U., Girbl, T., Alinger, B., Hofbauer, S.W., Neureiter, D., Hofbauer, J.P., Egle, A., Greil, R. & Hartmann, T.N. (2011) Differential bone marrow homing capacity of VLA-4 and CD38 high expressing chronic lymphocytic leukemia cells. PLoS ONE, 6, e23758. Burger, J.A., Ghia, P., Rosenwald, A. & CaligarisCappio, F. (2009) The microenvironment in mature B-cell malignancies: a target for new treatment strategies. Blood, 114, 3367–3375. Hallaert, D.Y., Jaspers, A., van Noesel, C.J., van Oers, M.H., Kater, A.P. & Eldering, E. (2008) c-Abl kinase inhibitors overcome CD40-mediated drug resistance in CLL: implications for therapeutic targeting of chemoresistant niches. Blood, 112, 5141–5149. Hamilton, E., Pearce, L., Morgan, L., Robinson, S., Ware, V., Brennan, P., Thomas, N.S., Yallop, D., Devereux, S., Fegan, C., Buggins, A.G. & Pepper, C. (2012) Mimicking the tumour microenvironment: three different co-culture systems induce a similar phenotype but distinct proliferative signals in primary chronic lymphocytic leukaemia cells. British Journal of Haematology, 158, 589–599. Herishanu, Y., Perez-Galan, P., Liu, D., Biancotto, A., Pittaluga, S., Vire, B., Gibellini, F., Njuguna, N., Lee, E., Stennett, L., Raghavachari, N., Liu, P., McCoy, J.P., Raffeld, M., Stetler-Stevenson, M., Yuan, C., Sherry, R., Arthur, D.C., Maric, I., White, T., Marti, G.E., Munson, P., Wilson, W. H. & Wiestner, A. (2011) The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in chronic lymphocytic leukemia. Blood, 117, 563–574. Hofbauer, J.P., Heyder, C., Denk, U., Kocher, T., Holler, C., Trapin, D., Asslaber, D., Tinhofer, I., Greil, R. & Egle, A. (2011) Development of CLL in the TCL1 transgenic mouse model is associated with severe skewing of the T-cell compartment homologous to human CLL. Leukemia, 25, 1452–1458. Messmer, B.T., Messmer, D., Allen, S.L., Kolitz, J. E., Kudalkar, P., Cesar, D., Murphy, E.J., Koduru, P., Ferrarini, M., Zupo, S., Cutrona, G., Damle, R.N., Wasil, T., Rai, K.R., Hellerstein, M.K. & Chiorazzi, N. (2005) In vivo measurements document the dynamic cellular kinetics of chronic lymphocytic leukemia B cells. J Clin Invest, 115, 755–764. Patten, P.E., Buggins, A.G., Richards, J., Wotherspoon, A., Salisbury, J., Mufti, G.J., Hamblin, T. J. & Devereux, S. (2008) CD38 expression in chronic lymphocytic leukemia is regulated by the tumor microenvironment. Blood, 111, 5173–5181. Plander, M., Seegers, S., Ugocsai, P., DiermeierDaucher, S., Ivanyi, J., Schmitz, G., Hofstadter, F., Schwarz, S., Orso, E., Knuchel, R. & Brockhoff, G. (2009) Different proliferative and survival capacity of CLL-cells in a newly established in vitro model for pseudofollicles. Leukemia, 23, 2118–2128.


Leukemia Research | 2003

An UK myeloma forum phase II study of thalidomide; long term follow-up and recommendations for treatment

Stephen Schey; Jamie Cavenagh; R Johnson; J A Child; Heather Oakervee; R W Jones

Myeloma remains incurable with conventional treatment in the vast majority of patients. The introduction of thalidomide in 1999 for the treatment of relapsed disease offers the opportunity to treat patients who have developed myelotoxicity or who are refractory to conventional chemotherapy. The optimal schedule remains unresolved and only two studies have reported long term follow-up data. We report a phase II low dose escalation study of thalidomide with long term follow-up showing overall survival (OS) of 19 months and progression free survival (PFS) of 14 months. In addition we report on the side effects and toxicity and give recommendations for the use of thalidomide in the relapsed setting based upon these findings.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Acute myeloid leukemia does not deplete normal hematopoietic stem cells but induces cytopenias by impeding their differentiation

Farideh Miraki-Moud; Fernando Anjos-Afonso; Katharine A. Hodby; Emmanuel Griessinger; Guglielmo Rosignoli; Debra M. Lillington; Li Jia; Jeff K. Davies; Jamie Cavenagh; Matthew Smith; Heather Oakervee; Samir G. Agrawal; John G. Gribben; Dominique Bonnet; David Taussig

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.

Collaboration


Dive into the Heather Oakervee's collaboration.

Top Co-Authors

Avatar

Jamie Cavenagh

St Bartholomew's Hospital

View shared research outputs
Top Co-Authors

Avatar

Rakesh Popat

University College London

View shared research outputs
Top Co-Authors

Avatar

J Cavenagh

St Bartholomew's Hospital

View shared research outputs
Top Co-Authors

Avatar

Samir G. Agrawal

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

John G. Gribben

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Taussig

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Nicola Foot

St Bartholomew's Hospital

View shared research outputs
Top Co-Authors

Avatar

Matthew Streetly

Guy's and St Thomas' NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge