Network


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

Hotspot


Dive into the research topics where Catherine Williams is active.

Publication


Featured researches published by Catherine Williams.


Lancet Oncology | 2013

Vorinostat or placebo in combination with bortezomib in patients with multiple myeloma (VANTAGE 088): a multicentre, randomised, double-blind study

Meletios A. Dimopoulos; David Siegel; Sagar Lonial; Junyuan Qi; Roman Hájek; Thierry Facon; Laura Rosiñol; Catherine Williams; Hilary Blacklock; Hartmut Goldschmidt; Varnia Hungria; Andrew Spencer; Antonio Palumbo; Thorsten Graef; Joseph E. Eid; Jennifer Houp; Linda Sun; Scott Vuocolo; Kenneth C. Anderson

BACKGROUND We aimed to assess efficacy and tolerability of vorinostat in combination with bortezomib for treatment of patients with relapsed or refractory multiple myeloma. METHODS In our randomised, double-blind, placebo-controlled, phase 3 trial, we enrolled adults (≥18 years) at 174 university hospitals in 31 countries worldwide. Eligible patients had to have non-refractory multiple myeloma that previously responded to treatment (one to three regimens) but were currently progressing, ECOG performance statuses of 2 or less, and no continuing toxic effects from previous treatment. We excluded patients with known resistance to bortezomib. We randomly allocated patients (1:1) using an interactive voice response system to receive 21 day cycles of bortezomib (1·3 mg/m(2) intravenously on days 1, 4, 8, and 11) in combination with oral vorinostat (400 mg) or matching placebo once-daily on days 1-14. We stratified patients by baseline tumour stage (International Staging System stage 1 or stage ≥2), previous bone-marrow transplantation (yes or no), and number of previous regimens (1 or ≥2). The primary endpoint was progression-free survival (PFS) in the intention-to-treat population. We assessed adverse events in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number 00773747. FINDINGS Between Dec 24, 2008, and Sept 8, 2011, we randomly allocated 317 eligible patients to the vorinostat group (315 of whom received at least one dose) and 320 to the placebo group (all of whom received at least one dose). Median PFS was 7·63 months (95% CI 6·87-8·40) in the vorinostat group and 6·83 months (5·67-7·73) in the placebo group (hazard ratio [HR] 0·77, 95% CI 0·64-0·94; p=0·0100). 312 (99%) of 315 patients in the vorinostat group and 315 (98%) of 320 patients in the placebo group had adverse events (300 [95%] adverse events in the vorinostat group and 282 [88%] in the control group were regarded as related to treatment). The most common grade 3-4 adverse events were thrombocytopenia (143 [45%] patients in the vorinostat group vs 77 [24%] patients in the placebo group), neutropenia (89 [28%] vs 80 [25%]), and anaemia (53 [17%] vs 40 [13%]). INTERPRETATION Although the combination of vorinostat and bortezomib prolonged PFS relative to bortezomib and placebo, the clinical relevance of the difference in PFS between the two groups is not clear. Different treatment schedules of bortezomib and vorinostat might improve tolerability and enhance activity. FUNDING Merck.


Journal of Clinical Oncology | 2002

Role of nonmyeloablative allogeneic stem-cell transplantation after failure of autologous transplantation in patients with lymphoproliferative malignancies

Katharine Branson; Rajesh Chopra; Panagiotis D. Kottaridis; Grant McQuaker; Anne Parker; Stephen Schey; Ronjon Chakraverty; Charles Craddock; Donald Milligan; Ruth Pettengell; Judith Marsh; David C. Linch; Anthony H. Goldstone; Catherine Williams; Stephen Mackinnon

PURPOSE Conventional allogeneic stem-cell transplantation (SCT) after a prior failed autograft is associated with a transplant-related mortality rate of 50% to 80%. The aim of the current study was to evaluate the safety and efficacy of sibling, HLA-matched, nonmyeloablative allogeneic SCT with donor lymphocyte infusion (DLI) in patients with lymphoid malignancy after failure of autologous SCT. PATIENTS AND METHODS A total of 38 patients with refractory, progressive, or relapsed disease after autologous SCT were entered onto this study. The conditioning regimen consisted of the humanized monoclonal antibody CAMPATH-1H, fludarabine, and melphalan. Fifteen of 35 assessable patients received DLI after SCT. RESULTS Sustained neutrophil engraftment was achieved in 37 recipients, and platelet engraftment was achieved in 35 patients. The estimated transplant-related mortality was 7.9% at day 100 and 20% at 14 months, the median duration of follow-up. Eight patients experienced grade I/II acute graft-versus-host disease (GVHD) after transplantation, but no grade III/IV GVHD was observed in this setting. However, grade III/IV GVHD occurred in seven patients who received DLI. The actuarial overall survival at 14 months was 53%, with a progression-free survival of 50%. DLI produced a further response in three of 15 recipients. CONCLUSION Nonmyeloablative allogeneic SCT after CAMPATH-1H-containing conditioning is a relatively safe option compared with conventional allogeneic transplantation for patients who have failed previous autologous SCT. The low incidence of early GVHD enabled the subsequent administration of DLI to improve further clinical responses in this poor-risk group of lymphoma and myeloma patients.


Journal of Clinical Oncology | 1998

Back-up bone marrow is frequently ineffective in patients with poor peripheral-blood stem-cell mobilization.

Michael J. Watts; Angela Michelle Sullivan; D Leverett; Andrew Peniket; Ar Perry; Catherine Williams; S Devereux; Anthony H. Goldstone; David C. Linch

PURPOSE To assess hematologic recovery and procedure-related mortality in patients who received high-dose therapy with stem-cell support, in whom the peripheral-blood stem-cell (PBSC) collection fails (CD34+ cells < 1 x 10(6)/kg). The predictive value of granulocyte-monocyte colony-forming cell (GM-CFC) measurements and the value of bone marrow obtained after PBSC collection failure was assessed. PATIENTS AND METHODS The study group comprised 324 consecutive patients mobilized with granulocyte colony-stimulating factor (G-CSF) and cyclophosphamide (273 patients), G-CSF with other chemotherapy (37 patients), and G-CSF alone (14 patients). Between one and four aphereses were performed. RESULTS In 51 of 324 patients, there was failure to obtain 1 x 10(6)/kg CD34+ cells. Twenty-three patients had greater than 1 x 10(5)/kg GM-CFC; 22 patients proceeded to high-dose therapy. Neutrophil recovery occurred within 21 days, but platelet independence was delayed (> 28 days) in eight patients. Of 28 patients with less than 1 x 10(5)/kg GM-CFC, six received high-dose therapy with PBSC alone and five had delayed engraftment. Twelve patients with less than 1 x 10(5)/kg GM-CFC received high-dose therapy supported by bone marrow collected after PBSC collection failure. Eleven patients were assessable for engraftment; four patients had slow (> 21 days) or delayed (> 28 days) neutrophil recovery and eight patients had delayed platelet recovery. In the group of patients who received less than 1 x 10(5)/kg GM-CFC, there were five procedure-related deaths. CONCLUSION This study shows that delayed hematologic recovery is frequent if less than 1 x 10(6)/kg CD34+ cells are infused after high-dose therapy, particularly with GM-CFC less than 1 x 10(5)/kg. The procedure-related mortality in this latter group is high. In most patients whose PBSC collection contains less than 1 x 10(5)/kg GM-CFC, the use of bone marrow cells does not improve engraftment, which suggests that poor PBSC mobilization usually indicates poor marrow function.


Journal of Biological Chemistry | 1998

p130, p107, and pRb are differentially regulated in proliferating cells and during cell cycle arrest by alpha-interferon.

Nicholas S. B. Thomas; Arnold Pizzey; Sanjay Tiwari; Catherine Williams; Jiewu Yang

We have determined how the phosphorylation of the retinoblastoma family (pRb, p107, and p130) is governed in individual cell cycle phases of Daudi B-cells during cell cycle exit triggered by α-interferon (α-IFN). α-IFN causes dephosphorylation of pRb and loss of p130 phosphorylated Form 3. However, the change in p130 phosphorylation in response to α-IFN occurs before dephosphorylation of pRb is complete because loss of p130 Form 3 occurs throughout the cell cycle prior to complete arrest in G1, whereas pRb is dephosphorylated only in G1. In contrast, p107 is dephosphorylated and is then depleted from cells as they exit the cell cycle. p130, predominantly in Form 1, and hypophosphorylated pRb bind an E2F DNA binding site; p130 complexes E2F-4, whereas pRb binds both E2F-4 and E2F-1. The phosphorylated forms of E2F-4 that bind to the E2F DNA site are different from hyperphosphorylated E2F-4, which predominates in primary hemopoietic cells in G0. We conclude that although cell cycle arrest induced by α-IFN may be mediated in part by formation of a complex containing p130 and E2F-4, α-IFN does not induce hyperphosphorylation of E2F-4, which characterizes primary hemopoietic cells in G0.


Cytotherapy | 2001

In vivo CAMPATH-1H prevents GvHD following nonmyeloablative stem-cell transplantation

Panagiotis D. Kottaridis; Donald Milligan; Rajesh Chopra; R K Chakraverty; S Chakrabarti; Sa Robinson; Karl S. Peggs; S Verfuerth; Ruth Pettengell; J. C. W. Marsh; Steve Schey; Premini Mahendra; Gareth J. Morgan; Geoff Hale; H Waldmann; M C Ruiz de Elvira; Catherine Williams; Stephen Devereux; David C. Linch; Anthony H. Goldstone; Stephen Mackinnon

BACKGROUND We have investigated a novel nonmyeloablative conditioning regimen in 44 patients with hematological malignancies. The median patient age was 41 years. Many of the patients had high-risk features, including 19 patients with a previous failed transplant. METHODS Recipient conditioning consisted of CAMPATH-1H 20 mg/day on Days -8 to -4, fludarabine 30 mg/m(2) on Days -7 to -3 and melphalan 140 mg/m(2) on Day -2. Thirty-six recipients received unmanipulated G-CSF mobilized PBSC from HLA identical siblings and eight received unmanipulated BM from MUD. GvHD prophylaxis was with CYA alone for 38 patients and CYA plus MTX for six sibling recipients. RESULTS Forty-two of the 43 evaluable patients had sustained engraftment. Results of chimerism analysis using microsatellite PCR indicate that 18 of 31 patients studied were full donor chimeras, while the other patients were mixed chimeras in one or more lineages. At a median follow-up of 9 months (range, 3-29 months) 33 patients remain alive in CR, or with no evidence of disease progression. Seven patients relapsed or progressed post-transplant and four of them subsequently died. Four patients died from regimen-related complications. There were no cases of Grades III-IV acute GvHD. Only two patients developed Grade II acute GvHD and only one had chronic GvHD. The estimated probability of non-relapse mortality at 1 year was 11%.Results: Although longer follow-up is needed to establish the long-term remission rates, this study demonstrates that this nonmyeloablative preparative regimen is associated with durable engraftment, minimal toxicity and low incidence of GvHD.


British Journal of Haematology | 2005

Analysis of outcome following allogeneic haemopoietic stem cell transplantation for myeloma using myeloablative conditioning – evidence for a superior outcome using melphalan combined with total body irradiation

Hannah M. Hunter; Karl S. Peggs; R. Powles; Amin Rahemtulla; Premini Mahendra; Jamie Cavenagh; Timothy Littlewood; Michael N. Potter; Ann Hunter; Antonio Pagliuca; Catherine Williams; Gordon Cook; Keiren Towlson; David I. Marks; Nigel H. Russell

We have undertaken a retrospective multicentre analysis of 139 patients (median age 44·4 years) undergoing allogeneic haematopoietic stem cell transplantation (HSCT) for multiple myeloma using myeloablative conditioning. The majority of patients received total body irradiation (TBI) combined with either melphalan (56·9%) or cyclophosphamide (28·5%). Overall, transplant‐related mortality (TRM) was 37·9% at 1 year and was not significantly different for patients receiving melphalan/TBI compared with cyclophosphamide/TBI. The overall complete remission (CR) rate, including patients in CR at the time of transplant, was greater for patients receiving melphalan/TBI (64·7%) compared with cyclophosphamide/TBI (47·2%)(P = 0·085). A significantly higher proportion of patients with continuing disease at the time of transplant achieved CR post‐transplant following melphalan/TBI conditioning compared with cyclophosphamide/TBI (52·9% and 33·4% respectively, P = 0·009). Relapse/progression rates at 5 years were significantly lower for melphalan/TBI (36·7%) compared with cyclophosphamide/TBI (80·8%, P < 0·0001) and remained significant in multivariate analysis. This resulted in an overall survival at 5 years of 44·1% and 28·1% for melphalan/TBI and cyclophosphamide/TBI, respectively (P = 0·059). These results demonstrate that the type of conditioning for sibling allogeneic HSCT for myeloma has a major effect on transplant outcome.


British Journal of Haematology | 2009

Bortezomib, low‐dose intravenous melphalan, and dexamethasone for patients with relapsed multiple myeloma

Rakesh Popat; Heather Oakervee; Catherine Williams; Mark Cook; Charles Craddock; Supratik Basu; Charles Singer; Stephen Harding; Nicola Foot; Simon Hallam; Liz Odeh; Simon Joel; Jamie Cavenagh

This multicenter phase I/II study investigated the maximum tolerated dose (MTD), safety, and efficacy of low dose intravenous (IV) melphalan in combination with bortezomib for patients with relapsed multiple myeloma (MM). Patients received bortezomib 1·3 mg/m2 on days 1, 4, 8, and 11 and escalating doses of IV melphalan (2·5–10·0 mg/m2) on day 2 of a 28‐day cycle for a maximum of eight cycles. Dexamethasone 20 mg was added for progressive or stable disease. Fifty‐three patients were enrolled. The MTD was defined at melphalan 7·5 mg/m2 and bortezomib 1·3 mg/m2. The overall response rate (ORR) was 68% (23% complete or near‐complete responses [CR/nCR]) whilst at the MTD (n = 33) the ORR was 76% (34% CR/nCR). After median follow‐up of 17 months, the median progression free survival was 10 months, rising to 12 months at the MTD (P < 0·05 vs. non‐MTD regimens). The median overall survival was 28 months, but was not yet reached at the MTD. Grade 3/4 adverse events included thrombocytopenia (62%), neutropenia (57%), infection (21%), and neuropathy (15%). Bortezomib and low‐dose IV melphalan combination therapy is a safe and highly effective regimen for patients with relapsed MM. These data suggest further investigation of this combination is warranted.


Bone Marrow Transplantation | 1998

A case of EBV-associated lymphoproliferative disease following high-dose therapy and CD34-purified autologous peripheral blood progenitor cell transplantation.

Andrew Peniket; Ar Perry; Catherine Williams; A MacMillan; Mj Watts; Pg Isaacson; Ah Goldstone; David C. Linch

A fatal case of EBV-associated lymphoproliferative disorder arising after a CD34-selected autologous peripheral blood stem cell transplant is reported in a patient with multiple myeloma in first plateau phase. It is suggested that this is likely to be a consequence of the accessory cell depletion associated with the CD34+ cell purification and it is recommended that a source of autologous T cells is stored before transplantation to be used if a severe opportunistic infection or EBV lymphoma arises post-transplantation.


Bone Marrow Transplantation | 1997

Evaluation of clinical scale CD34+ cell purification: experience of 71 immunoaffinity column procedures.

Michael J. Watts; Am Sullivan; Stuart J. Ings; D Leverett; Peniket Aj; Ar Perry; Catherine Williams; S Devereux; Anthony H. Goldstone; David C. Linch

Seventy-one mobilised PBSC collections were subject to CD34+ cell purification using the CEPRATE SC® stem cell concentration system. The overall median purity of CD34+ cells was 69% (6–93%). CD34+ cell, and GM-CFC recoveries were 52% (8–107%) and 36% (3–118%). Purity was logarithmically related to the input percentage of CD34+ cells and starting requirements were established of 1% CD34 cell content for optimal purity and a minimum of 2 × 106/kg CD34+ cells to ensure recovery of our minimum engraftment threshold of 1 × 106/kg CD34+ cells. Reduction of the washing steps reduced non-specific cell losses and shortened the procedure but did not affect progenitor cell recovery. Purified CD34+ cells were reinfused following high-dose therapy in 35 patients. The median time to neutrophil recovery of 0.5 × 109/l was 12 (10–23) days and to the attainment of platelet independence was 13 (7–100) days. The risks of delayed platelet recovery were related to the CD34+ cell dose infused and were identical to the risks when non-purified PBSC collections were used. In conclusion, purification of CD34+ cells using the CEPRATE device is reliable and the purified product results in prompt engraftment. The cell losses that occur do however restrict its use in many patients.


British Journal of Haematology | 1995

Accessory cells do not contribute to G‐CSF or IL‐6 production nor to rapid haematological recovery following peripheral blood stem cell transplantation

Michael J. Watts; H. M. Jones; A. M. Sullivan; Stephen E. Langabeer; E. Jamieson; Adele K. Fielding; Catherine Williams; R. J. Berenson; Ah Goldstone; David C. Linch

Summary. Haemopoietic recovery is more rapid after peripheral blood stem cell (PBSC) transplantation than after autologous bone marrow transplantation, and the aim of this study was to assess the role of the large number of lymphocytes and monocytes (accessory cells) in a PBSC leukapheresis product in this rapid regeneration. Haematological recovery was therefore assessed in 10 PBSC recipients with lymphoma or myeloma in whom monocytes and T cells were depleted by a median of 2.3 and 3.3 logs by CD34+ cell selection using the CEPRATE® SC stem cell concentration system and compared with recovery in 59 recipients who received whole PBSC. After allowing for the number of progenitor cells reinfused, there was no significant delay in engraftment induced by accessory cell depletion. Plasma levels of granulocyte‐colony stimulating factor (G‐CSF), granulocyte/monocyte‐colony stimulating factor (GM‐CSF), interleukin‐6 (IL‐6), stem cell factor (SCF) and macrophage‐inhibition factor‐alpha (MIP‐1‐alpha) during the transplant procedure were similar whether or not accessory cells were given. The G‐CSF and IL‐6 levels rose between days 5 and 14 post transplantation to approximately 1 ng/ml and 50pg/ml respectively. This study indicates that accessory cells reinfused with PBSC collections are not responsible for the subsequent cytokine profile or rapid haematological recovery.

Collaboration


Dive into the Catherine Williams's collaboration.

Top Co-Authors

Avatar

David C. Linch

University College London

View shared research outputs
Top Co-Authors

Avatar

Karl S. Peggs

University College London

View shared research outputs
Top Co-Authors

Avatar

Donald Milligan

Heart of England NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Jamie Cavenagh

St Bartholomew's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony H. Goldstone

University College London Hospitals NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Premini Mahendra

Queen Elizabeth Hospital Birmingham

View shared research outputs
Top Co-Authors

Avatar

Charles Craddock

University Hospitals Birmingham NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Ronjon Chakraverty

Royal Free London NHS Foundation Trust

View shared research outputs
Researchain Logo
Decentralizing Knowledge