Network


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

Hotspot


Dive into the research topics where Geoff Hale is active.

Publication


Featured researches published by Geoff Hale.


Annals of Neurology | 1999

Monoclonal antibody treatment exposes three mechanisms underlying the clinical course of multiple sclerosis

Alasdair Coles; Mark Wing; Paul Molyneux; Andrea Paolillo; Charlie M. Davie; Geoff Hale; David Miller; H Waldmann; Alastair Compston

The elective treatment of patients with multiple sclerosis, using a humanized anti-leukocyte (CD52) monoclonal antibody (Campath-1H), has illuminated mechanisms that underlie the clinical course of the disease. Twenty-seven patients were studied clinically and by magnetic resonance imaging (MRI) before and for 18 months after a single pulse of Campath-1H. The first dose of monoclonal antibody was associated with a transient rehearsal of previous symptoms caused by the release of mediators that impede conduction at previously demyelinated sites; this effect remained despite selective blockade of tumor necrosis factor-alpha. Disease activity persisted for several weeks after treatment but thereafter radiological markers of cerebral inflammation were suppressed for at least 18 months during which there were no new symptoms or signs. However, about half the patients experienced progressive disability and increasing brain atrophy, attributable on the basis of MRI spectroscopy to axonal degeneration, which correlated with the extent of cerebral inflammation in the pretreatment phase. These data support the formulation that inflammation and demyelination are responsible for relapses of multiple sclerosis; that inflammatory mediators, but not tumor necrosis factor-alpha, cause symptomatic reactivation of previously demyelinated lesions; and that axonal degeneration, conditioned by prior inflammation but proceeding despite its suppression, contributes to the progressive phase of disability. These results provide evidence supporting the emerging view that treatment in multiple sclerosis must be given early in the course, before the consequences of inflammation are irretrievably established.


Journal of Neurology | 2006

The window of therapeutic opportunity in multiple sclerosis: evidence from monoclonal antibody therapy.

Alasdair Coles; Amanda L. Cox; E. Le Page; Joanne L. Jones; Sa Trip; J. Deans; S Seaman; David H. Miller; Geoff Hale; H Waldmann; D. A. S. Compston

AbstractFrom 1991–2002, we treated 58 patients with multiple sclerosis (MS) using the humanised monoclonal antibody, Campath–1H, which causes prolonged T lymphocyte depletion. Clinical and surrogate markers of inflammation were suppressed. In both the relapsing–remitting (RR) and secondary progressive (SP) stages of the illness, Campath–1H reduced the annual relapse rate (from 2.2 to 0.19 and from 0.7 to 0.001 respectively; both p < 0.001). Remarkably, MRI scans of patients with SP disease, treated with Campath–1H 7 years previously, showed no new lesion formation. However, despite these effects on inflammation, disability was differently affected depending on the phase of the disease. Patients with SPMS showed sustained accumulation of disability due to uncontrolled progression marked by unrelenting cerebral atrophy, attributable to ongoing axonal loss. The rate of cerebral atrophy was greatest in patients with established cerebral atrophy and highest inflammatory lesion burden before treatment (2.3 versus 0.7 ml/year; p = 0.04). In contrast, patients with RR disease showed an impressive reduction in disability at 6 months after Campath–1H (by a mean of 1.2 EDSS points) perhaps owing to a suppression of on–going inflammation in these patients with unusually active disease. In addition, there was a further significant, albeit smaller, mean improvement in disability up to 36 months after treatment.We speculate that this represents the beneficial effects of early rescue of neurons and axons from a toxic inflammatory environment, and that prevention of demyelination will prevent long–term axonal degeneration. These concepts are currently being tested in a controlled trial comparing Campath–1H and IFN–beta in the treatment of drug–naïve patients with early, active RR MS.


Transplantation | 1999

Campath IH allows low-dose cyclosporine monotherapy in 31 cadaveric renal allograft recipients.

R. Y. Calne; Susan D. Moffatt; Peter J. Friend; Neville V. Jamieson; J. A. Bradley; Geoff Hale; J. Firth; John R. Bradley; Kenneth Smith; Herman Waldmann

BACKGROUND Campath 1H is a depleting, humanized anti-CD52 monoclonal antibody that has now been used in 31 renal allograft recipients. The results have been very encouraging and are presented herein. METHODS Campath 1H was administered, intravenously, in a dose of 20 mg, on day 0 and day 1 after renal transplant. Low-dose cyclosporine (Neoral) was then initiated at 72 hr after transplant. These patients were maintained on low-dose monotherapy with cyclosporine. RESULTS At present, the mean follow-up is 21 months (range: 15-28 months). All but one patient are alive and 29 have intact functioning grafts. There have been six separate episodes of steroid-responsive rejection. One patient has had a recurrence of her original disease. Two patients have suffered from opportunistic infections, which responded to therapy. One patient has died secondary to ischemic cardiac failure. CONCLUSIONS Campath 1H has resulted in acceptable outcomes in this group of renal allograft recipients. This novel therapy is of equal efficacy compared to conventional triple therapy, but allows the patient to be steroid-free and to be maintained on very-low-dose immunosuppressive monotherapy.


The Lancet | 2005

Clinical evidence of a graft-versus-Hodgkin's-lymphoma effect after reduced-intensity allogeneic transplantation

Karl S. Peggs; Ann Hunter; Rajesh Chopra; Anne Parker; Premini Mahendra; Donald Milligan; Charles Craddock; Ruth Pettengell; Ahmet Dogan; Kirsty Thomson; Emma Morris; Geoff Hale; Herman Waldmann; Anthony H. Goldstone; David C. Linch; Stephen Mackinnon

BACKGROUND In patients with multiply relapsed Hodgkins lymphoma allogeneic stem-cell transplantation has been limited by prohibitive non-relapse-related mortality rates and by a lack of definitive evidence for a therapeutic graft-versus-tumour effect. Therefore, we aimed to assess the graft-versus-tumour effect of reduced-intensity allogeneic transplantation. METHODS We undertook reduced-intensity transplantation in 49 patients with multiply relapsed Hodgkins lymphoma, 44 (90%) of whom had progression of disease after previous autologous transplantation (median age 32 years [range 18-51], number of previous treatment courses was five [range 3-8], and time from diagnosis 4.8 years [range 0.6-4.8]). 31 patients had HLA matched donors who were related and 18 had donors who were unrelated. Median follow-up was 967 days (range 102-2232). The primary endpoints were engraftment, toxic effects, non-relapse-related mortality, incidence of graft-versus-host disease (GVHD), and the toxic effects of adjuvant donor-lymphocyte infusion. FINDINGS All patients engrafted. Eight of 49 (16%) had grade II-IV acute GVHD and seven (14%) had chronic GVHD before donor-lymphocyte infusion. 16 (33%) patients received donor-lymphocyte infusion from 3 months after transplantation for residual disease or progression. Six (38%) of the 16 developed grade II-IV acute GVHD and five developed chronic GVHD. Nine (56%) showed disease responses after infusion (eight complete, one partial). Non-relapse-related mortality was 16.3% at 730 days (7.2% for patients who had related donors vs 34.1% for those with unrelated donors, p=0.0206). Projected 4 year overall and progression-free survival were 55.7% and 39.0%, respectively (62.0% and 41.5% for related donors). INTERPRETATION These data show the potential for durable responses in patients who have previously had substantial treatment for Hodgkins lymphoma. The low non-relapse-related mortality suggests the procedure could be undertaken earlier in the course of the disease.


European Journal of Immunology | 2005

Lymphocyte homeostasis following therapeutic lymphocyte depletion in multiple sclerosis

Amanda L. Cox; Sara A. J. Thompson; Joanne L. Jones; Vicki Robertson; Geoff Hale; Herman Waldmann; D Alastair S Compston; Alasdair Coles

Following lymphocyte depletion, homeostatic mechanisms drive the reconstitution of lymphocytes. We prospectively studied this process in 16 patients for 1 year after a single pulse of treatment with Campath‐1H, a humanised anti‐CD52 monoclonal antibody. We observed two phases of lymphocyte reconstitution. In the first 6 months after treatment the precursor frequency and proliferation index of the patients’ autologous mixed lymphocyte reaction increased; the depleted T cell pool was dominated by memory T cells, especially CD4+CD25high T cells, a putative regulatory phenotype; and there was a non‐significant rise in peripheral mononuclear cell FoxP3 mRNA expression and fall in constitutive cytokine mRNA expression. In the later phase, from 6‐to‐12 months after Campath‐1H, these changes reversed and there was a rise in ROG mRNA expression. However, total CD4+ numbers remained below 50% of pre‐treatment levels at 12 months, perhaps reflecting a failure in homeostasis. This was not due to an impaired IL‐7 response, as in rheumatoid arthritis, nor to a lack of IL‐7 receptors, which are found on fewer human CD4+CD25high than naive cells. We speculate that CCL21 and IL‐15 responses to lymphopaenia may be suboptimal in multiple sclerosis.


Journal of Clinical Investigation | 1996

Mechanism of first-dose cytokine-release syndrome by CAMPATH 1-H: involvement of CD16 (FcgammaRIII) and CD11a/CD18 (LFA-1) on NK cells.

Mark Wing; Thibault Moreau; Judith Greenwood; Richard M. Smith; Geoff Hale; John D. Isaacs; Herman Waldmann; Peter Lachmann; Alastair Compston

The administration of the immunosuppressive humanized monoclonal antibody CAMPATH 1-H, which recognizes CD52 on lymphocytes and monocytes, is associated with a first-dose cytokine-release syndrome involving TNFalpha, IFNgamma, and IL-6 clinically. In vitro models have been used to establish the cellular source and mechanism responsible for cytokine release, demonstrating that cytokine release is isotype dependent, with the rat IgG2b and human IgG1 isotype inducing the highest levels of cytokine release, which was inhibited with antibody to CD16, the low affinity Fc-receptor for IgG (FcgammaR). Cross-linking antibody opsonized CD4 T lymphocytes failed to stimulate TNFalpha release, which together with the observation that TNFalpha release by purified natural killer (NK) cells stimulated by fixed autologous CAMPATH 1-H-opsonized targets was inhibited with anti-CD16, indicates that cytokine release results from ligation of CD16 on the NK cells, rather than Fc-receptor (FcR)-dependent cross-linking of CD52 on the targeted cell. Since the hierarchy of isotypes inducing cytokine release in these cultures matches that seen clinically, we conclude that ligation of CD16 on NK cells is also responsible for cytokine release after injection of CAMPATH 1-H in vivo.


Journal of Clinical Oncology | 2005

Allogeneic Stem-Cell Transplantation Using a Reduced-Intensity Conditioning Regimen Has the Capacity to Produce Durable Remissions and Long-Term Disease-Free Survival in Patients With High-Risk Acute Myeloid Leukemia and Myelodysplasia

Sudhir Tauro; Charles Craddock; Karl S. Peggs; Gulnaz Begum; Premini Mahendra; Gordon Cook; Judith Marsh; Donald Milligan; Anthony H. Goldstone; Ann Hunter; Asim Khwaja; Raj Chopra; Timothy Littlewood; Andrew Peniket; Anne Parker; Graham Jackson; Geoff Hale; Mark Cook; Nigel H. Russell; Stephen Mackinnon

PURPOSE The toxicity of allogeneic stem-cell transplantation can be substantially reduced using a reduced-intensity conditioning (RIC) regimen. This has increased the proportion of patients with myeloid malignancies eligible for allogeneic transplantation. However, the capacity of RIC allografts to produce durable remissions in patients with acute myeloid leukemia (AML) and myelodysplasia (MDS) has not yet been defined, and consequently, the role of RIC allografts in the management of these diseases remains conjectural. PATIENTS AND METHODS Seventy-six patients with high-risk AML or MDS received an allograft using a fludarabine/melphalan RIC regimen incorporating alemtuzumab. The median age of the cohort was 52 years (range, 18 to 71 years). RESULTS The 100-day transplantation-related mortality rate was 9%, and no patient developed greater than grade 2 graft-versus-host disease. With a median follow-up of 36 months (range, 13 to 70 months), 27 patients were alive and in remission, with 3-year actuarial overall survival (OS) and disease-free survival (DFS) rates of 41% and 37%, respectively. The 3-year OS and DFS rates of patients with AML in complete remission at the time of transplantation were 48% and 42%, respectively. Disease relapse was the most common cause of treatment failure and occurred at a median time of 6 months after transplantation. All but one patient destined to relapse did so within 24 months of transplantation. CONCLUSION The extended follow-up in this series identifies a high risk of early disease relapse but provides evidence that RIC allografts can produce sustained DFS in a significant number of patients with AML who would be ineligible for allogeneic transplantation with myeloablative conditioning.


American Journal of Transplantation | 2005

Alemtuzumab (CAMPATH 1H) Induction Therapy in Cadaveric Kidney Transplantation—Efficacy and Safety at Five Years

Christopher J. E. Watson; J. Andrew Bradley; Peter J. Friend; John Firth; Craig J. Taylor; John R. Bradley; Kenneth Smith; S. Thiru; Neville V. Jamieson; Geoff Hale; Herman Waldmann; Roy Calne

Alemtuzumab is a powerful lymphocyte depleting antibody currently being evaluated in solid organ transplantation. This paper describes 5‐year results of a single center study of alemtuzumab as induction in renal transplantation.


Transplantation | 1999

Phase I study of an engineered aglycosylated humanized CD3 antibody in renal transplant rejection

Peter J. Friend; Geoff Hale; L Chatenoud; P. R. U. B. Rebello; John R. Bradley; S. Thiru; J M Phillips; H. Waldmann

BACKGROUND The potential therapeutic benefits of CD3 monoclonal antibodies, such as OKT3, have been limited by their immunogenicity and their propensity to activate a severe cytokine release syndrome. This has constrained the clinical use of OKT3 to the treatment of acute rejection episodes of organ allografts. METHODS We have humanized a rat CD3 antibody and created a single amino acid substitution in position 297 of the IgG1 heavy chain to prevent glycosylation and, consequently, binding of the therapeutic antibody to Fc receptors and to complement. This antibody has been given as first line antirejection therapy in nine kidney transplant recipients with biopsy-proven acute rejection episodes. RESULTS None of the patients demonstrated any antiglobulin response nor any significant cytokine release syndrome. Seven of the nine showed evidence of resolution of their rejection, although some patients experienced re-rejection. CONCLUSIONS These findings suggest that CD3 antibodies can be engineered to lose their toxicity while retaining their potency as immunosuppressants. Nonactivating humanized CD3 monoclonal antibodies now merit further investigation in the management of transplant patients and in therapy of autoimmune diseases.


British Journal of Haematology | 1996

Unrelated donor bone marrow transplantation for children with relapsed acute lymphoblastic leukaemia in second complete remission

Anthony Oakhill; D. H. Pamphilon; M. N. Potter; Colin G. Steward; S. Goodman; A. Green; P. Goulden; N. J. Goulden; Geoff Hale; Herman Waldmann; Jacqueline M. Cornish

Allogeneic sibling bone marrow transplantation (BMT) is the recommended treatment for relapsed childhood acute lymphoblastic leukaemia (ALL), but appropriate donors are only available in 30% of cases. Unfortunately, BMT from unrelated donors (UD) has been associated with high rates of severe graft‐versus‐host disease (GvHD) and transplant‐related mortality (TRM). In an attempt to improve outcome in UD‐BMT we have assessed the impact of T‐cell depletion using CAMPATH‐1 (anti‐CD52) monoclonal antibodies in 50 consecutively referred patients with relapsed ALL in second remission. All were previously treated according to MRC protocols UKALL X and XI, and then given chemotherapy on MRC R1 from relapse until UD‐BMT. 19 patients had relapsed on and 31 off therapy.

Collaboration


Dive into the Geoff Hale's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donald Milligan

Heart of England NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

John D. Isaacs

Newcastle upon Tyne Hospitals NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karl S. Peggs

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David C. Linch

University College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge