Network


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

Hotspot


Dive into the research topics where Fiona Thistlethwaite is active.

Publication


Featured researches published by Fiona Thistlethwaite.


Molecular Cell | 1998

Involvement of Brca2 in DNA Repair

Ketan J. Patel; Veronica Yu; Hyunsook Lee; Anne E. Corcoran; Fiona Thistlethwaite; Martin J. Evans; William H. Colledge; Lori Friedman; Bruce A.J. Ponder; Ashok R. Venkitaraman

Abnormalities precipitated by a targeted truncation in the murine gene Brca2 define its involvement in DNA repair. In culture, cells harboring truncated Brca2 exhibit a proliferative impediment that worsens with successive passages. Arrest in the G1 and G2/M phases is accompanied by elevated p53 and p21 expression. Increased sensitivity to genotoxic agents, particularly ultraviolet light and methylmethanesulfonate, shows that Brca2 function is essential for the ability to survive DNA damage. But checkpoint activation and apoptotic mechanisms are largely unaffected, thereby implicating Brca2 in repair. This is substantiated by the spontaneous accumulation of chromosomal abnormalities, including breaks and aberrant chromatid exchanges. These findings define a function of Brca2 in DNA repair, whose loss precipitates replicative failure, mutagen sensitivity, and genetic instability reminiscent of Bloom syndrome and Fanconi anemia.


Molecular Cell | 1999

Mitotic Checkpoint Inactivation Fosters Transformation in Cells Lacking the Breast Cancer Susceptibility Gene, Brca2

Hyunsook Lee; Alison H Trainer; Lori Friedman; Fiona Thistlethwaite; Martin J. Evans; Bruce A.J. Ponder; Ashok R. Venkitaraman

The murine Brca2 gene encodes a nuclear protein implicated in DNA repair. Brca2 behaves as a tumor suppressor, but paradoxically, its truncation causes proliferative arrest and spontaneous chromosomal damage. Here, we report that inactivation of cell cycle checkpoints responsive to mitotic spindle disruption, by mutant forms of p53 or Bub1, relieves growth arrest and initiates neoplastic transformation in primary cells homozygous for truncated Brca2. Tumors from Brca2-deficient animals exhibit dysfunction of the spindle assembly checkpoint, accompanied by mutations in p53, Bub1, and Mad3L. The chromosomal aberrations precipitated by Brca2 truncation can be suppressed by mutant forms of Bub1 and p53. Thus, inactivating mutations in mitotic checkpoint genes likely cooperate with BRCA2 deficiency in the pathogenesis of inherited breast cancer, with important implications for treatment.


Clinical Cancer Research | 2010

Modulation of lymphocyte regulation for cancer therapy: a phase II trial of tremelimumab in advanced gastric and esophageal adenocarcinoma.

Christy Ralph; Eyad Elkord; Deborah J. Burt; Jackie F. O'Dwyer; Eric Austin; Peter L. Stern; Robert E. Hawkins; Fiona Thistlethwaite

Purpose: Cytotoxic T lymphocyte antigen 4 (CTLA4), a key negative regulator of T-cell activation, is targeted by the antibody tremelimumab to release potentially useful antitumor activity. Experimental Design: This phase II trial investigated tremelimumab as a second-line treatment for patients with metastatic gastric and esophageal adenocarcinomas. Tremelimumab was given every 3 months until symptomatic disease progression. Safety, clinical efficacy, and immunologic activity were evaluated. Results: Eighteen patients received tremelimumab. Most drug-related toxicity was mild; however, there was a single death due to bowel perforation that complicated colitis. Four patients had stable disease with clinical benefit; one patient achieved a partial response after eight cycles (25.4 months) and remains well on study at 32.7 months. Markers of regulatory phenotype, forkhead box protein 3 and CTLA4, doubled transiently in CD4+CD25high lymphocytes in the first month after tremelimumab before returning to baseline. In contrast, CTLA4 increased in CD4+CD25low/negative lymphocytes throughout the cycle of treatment. De novo proliferative responses to tumor-associated antigens 5T4 (8 of 18 patients) and carcinoembryonic antigen (5 of 13) were detected. Patients with a posttreatment carcinoembryonic antigen proliferative response had median survival of 17.1 months compared with 4.7 months for nonresponders (P = 0.004). Baseline interleukin-2 release after T-cell activation was higher in patients with clinical benefit and toxicity. Conclusion: Despite the disappointing response rate of tremelimumab, one patient had a remarkably durable benefit for this poor-prognosis disease. In vitro evidence of enhanced proliferative responses to relevant tumor-associated antigens suggests that combining CTLA4 blockade with antigen-targeted therapy may warrant further investigation. Clin Cancer Res; 16(5); 1662–72


Clinical Immunology | 2011

Tremelimumab (anti-CTLA4) mediates immune responses mainly by direct activation of T effector cells rather than by affecting T regulatory cells

Sameena Khan; Deborah J. Burt; Christy Ralph; Fiona Thistlethwaite; Robert E. Hawkins; Eyad Elkord

Cytotoxic T Lymphocyte Antigen 4 (CTLA4) blockade has shown antitumor activity against common cancers. However, the exact mechanism of immune mediation by anti-CTLA4 remains to be elucidated. Further understanding of how CTLA4 blockade with tremelimumab mediates immune responses may allow a more effective selection of responsive patients. Our results show that tremelimumab enhanced the proliferative response of T effector cells (Teff) upon TCR stimulation, and abrogated Treg suppressive ability. In the presence of tremelimumab, frequencies of IL-2-secreting CD4(+) T cells and IFN-γ-secreting CD4(+) and CD8(+) T cells were increased in response to polyclonal activation and tumor antigens. Importantly, Treg frequency was not reduced in the presence of tremelimumab, and expanded Tregs in cancer patients treated with tremelimumab expressed FoxP3 with no IL-2 release, confirming them as bona fide Tregs. Taken together, this data indicates that tremelimumab induces immune responses mainly by direct activation of Teff rather than by affecting Tregs.


Journal of Immunotherapy | 2009

Vaccination of Patients with Metastatic Renal Cancer with Modified Vaccinia Ankara Encoding the Tumor Antigen 5T4 (TroVax) Given Alongside Interferon-α

Robert E. Hawkins; Catriona Macdermott; Alaaeldin Shablak; Caroline Hamer; Fiona Thistlethwaite; Noel Drury; Priscilla Chikoti; William Shingler; Stuart Naylor; Richard Harrop

Approximately 90% of renal cell tumors overexpress the tumor antigen 5T4. The attenuated strain of vaccinia virus, modified vaccinia Ankara, has been engineered to express 5T4 (TroVax). We conducted an open-label phase 1/2 trial in which TroVax was administered alongside interferon-α (IFNα) to 11 patients with metastatic renal cell carcinoma. Antigen-specific cellular and humoral responses were monitored throughout the study, and clinical responses were assessed by measuring the changes in tumor burden by computed tomography scan (Response Evaluation Criteria In Solid Tumors). The primary objective was to assess the safety, immunogenicity, and efficacy of TroVax when given alongside IFNα. Treatment with TroVax plus IFNα was well tolerated with no serious adverse events attributed to TroVax. All 11 patients mounted 5T4-specific antibody responses and 5 (45%) mounted cellular responses. No objective tumor responses were seen, but the overall median time to progression (TTP) of 9 months (range: 2.1 to 26+ mo) was longer than expected for IFNα alone. For the 10 clear cell patients the TTP ranged from 3.9 to 26+ months, with a median TTP of 10.4 months. The high frequency of 5T4-specific immune responses and prolonged median TTP for clear cell patients compared with that expected for IFNα alone is encouraging and warrants further investigation.


British Journal of Cancer | 2005

Engineering T cells for cancer therapy

Was Mansoor; David E. Gilham; Fiona Thistlethwaite; Robert E. Hawkins

It is generally accepted that the immune system plays an important role in controlling tumour development. However, the interplay between tumour and immune system is complex, as demonstrated by the fact that tumours can successfully establish and develop despite the presence of T cells in tumour. An improved understanding of how tumours evade T-cell surveillance, coupled with technical developments allowing the culture and manipulation of T cells, has driven the exploration of therapeutic strategies based on the adoptive transfer of tumour-specific T cells. The isolation, expansion and re-infusion of large numbers of tumour-specific T cells generated from tumour biopsies has been shown to be feasible. Indeed, impressive clinical responses have been documented in melanoma patients treated with these T cells. These studies and others demonstrate the potential of T cells for the adoptive therapy of cancer. However, the significant technical issues relating to the production of natural tumour-specific T cells suggest that the application of this approach is likely to be limited at the moment. With the advent of retroviral gene transfer technology, it has become possible to efficiently endow T cells with antigen-specific receptors. Using this strategy, it is potentially possible to generate large numbers of tumour reactive T cells rapidly. This review summarises the current gene therapy approaches in relation to the development of adoptive T-cell-based cancer treatments, as these methods now head towards testing in the clinical trial setting.


Journal of Immunotherapy | 2011

High dose interleukin-2 can produce a high rate of response and durable remissions in appropriately selected patients with metastatic renal cancer

Alaaeldin Shablak; Kanwal A Sikand; Jonathan Shanks; Fiona Thistlethwaite; Andrea Spencer-Shaw; Robert E. Hawkins

Metastatic renal cancer remains hard to treat and the treatment is generally palliative. However, high-dose interleukin-2 (HD IL-2) produced 5% to 10% complete remissions and most of these were durable. With the advent of newer treatments with less toxicity, the role of HD IL-2 is uncertain. We present here a case series of 72 patients with metastatic renal cancer given first-line treatment with HD IL-2. From 2003 to 2006, the patients were offered treatment with HD IL-2 irrespective of their histologic features (retrospective cohort). From 2006 to 2008, the treatment was only offered to patients after stratification into risk groups based on histologic criteria (prospective cohort). In the early series, the response rate to HD IL-2 was 27% (8/30), but with prospective stratification of patients by histology the response rate was 52% (21/40) in the group with favorable histologic features. Combining outcome for all patients with the favorable histology (including those identified retrospectively) 49% (28/57) responded with 25% (14/57) achieving a complete remission and these seem durable. Patients with metastatic renal cancer should be carefully assessed for their suitability to undergo treatment with first-line systemic therapy with HD IL-2 as in carefully selected patients it has a high-rate response and durable remissions.


British Journal of Haematology | 2005

Killing of non-Hodgkin lymphoma cells by autologous CD19 engineered T cells.

Eleanor J. Cheadle; David E. Gilham; Fiona Thistlethwaite; John Radford; Robert E. Hawkins

Adoptive immunotherapy with tumour‐specific T cells is an emerging technology that may be applicable to a broad range of cancers. However, tumours can avoid T cell‐mediated attack through multiple mechanisms including downregulation of major histocompatability complex (MHC). Consequently, engineering T cells to target intact protein antigen directly, thus bypassing the need for MHC presentation, can facilitate T cell targeting of tumour cells. Peripheral blood lymphocytes from nine of nine patients with non‐Hodgkin lymphoma (NHL) were successfully gene‐modified to express a receptor consisting of a CD19 single chain variable fragment (scFv) fused to the T cell CD3ζ signalling molecule. These T cells were functionally active against the CD19+ Raji Burkitts lymphoma cell line. Importantly, engineered T cells from seven of nine NHL patients efficiently lysed autologous lymph node tumour biopsy cells. There was a clear correlation between levels of CD19 expression on the tumour and effective killing by the engineered T cells. For two patients with a low or absent CD19+ cells within the biopsy, no significant killing was observed. These results demonstrate that patients with CD19+ NHL would be suitable candidates for this form of therapy in the setting of a phase I clinical trial.


Lancet Oncology | 2018

Preliminary results for avelumab plus axitinib as first-line therapy in patients with advanced clear-cell renal-cell carcinoma (JAVELIN Renal 100): an open-label, dose-finding and dose-expansion, phase 1b trial

Toni K. Choueiri; James Larkin; Mototsugu Oya; Fiona Thistlethwaite; Marcella Martignoni; Paul Nathan; Thomas Powles; David F. McDermott; Paul B. Robbins; David D. Chism; Daniel Cho; Michael B. Atkins; Michael S. Gordon; Sumati Gupta; Hirotsugu Uemura; Yoshihiko Tomita; Anna Compagnoni; Camilla Fowst; Alessandra di Pietro; Brian I. Rini

BACKGROUND The combination of an immune checkpoint inhibitor and a VEGF pathway inhibitor to treat patients with advanced renal-cell carcinoma might increase the clinical benefit of these drugs compared with their use alone. Here, we report preliminary results for the combination of avelumab, an IgG1 monoclonal antibody against the programmed cell death protein ligand PD-L1, and axitinib, a VEGF receptor inhibitor approved for second-line treatment of advanced renal-cell carcinoma, in treatment-naive patients with advanced renal-cell carcinoma. METHODS The JAVELIN Renal 100 study is an ongoing open-label, multicentre, dose-finding, and dose-expansion, phase 1b study, done in 14 centres in the USA, UK, and Japan. Eligible patients were aged 18 years or older (≥20 years in Japan) and had histologically or cytologically confirmed advanced renal-cell carcinoma with clear-cell component, life expectancy of at least 3 months, an Eastern Cooperative Oncology Group performance status of 1 or less, received no previous systemic treatment for advanced renal cell carcinoma, and had a resected primary tumour. Patients enrolled into the dose-finding phase received 5 mg axitinib orally twice daily for 7 days, followed by combination therapy with 10 mg/kg avelumab intravenously every 2 weeks and 5 mg axitinib orally twice daily. Based on the pharmacokinetic data from the dose-finding phase, ten additional patients were enrolled into the dose-expansion phase and assigned to this regimen. The other patients in the dose-expansion phase started taking combination therapy directly. The primary endpoint was dose-limiting toxicities in the first 4 weeks (two cycles) of treatment with avelumab plus axitinib. Safety and antitumour activity analyses were done in all patients who received at least one dose of avelumab or axitinib. This trial is registered with ClinicalTrials.gov, number NCT02493751. FINDINGS Between Oct 30, 2015, and Sept 30, 2016, we enrolled six patients into the dose-finding phase and 49 into the dose-expansion phase of the study. One dose-limiting toxicity of grade 3 proteinuria due to axitinib was reported among the six patients treated during the dose-finding phase. At the cutoff date (April 13, 2017), six (100%, 95% CI 54-100) of six patients in the dose-finding phase and 26 (53%, 38-68) of 49 patients in the dose-expansion phase had confirmed objective responses (32 [58%, 44-71] of all 55 patients). 32 (58%) of 55 patients had grade 3 or worse treatment-related adverse events, the most frequent being hypertension in 16 (29%) patients and increased concentrations of alanine aminotransferase, amylase, and lipase, and palmar-plantar erythrodysaesthesia syndrome in four (7%) patients each. Six (11%) of 55 patients died before data cutoff, five (9%) due to disease progression and one (2%) due to treatment-related autoimmune myocarditis. At the end of the dose-finding phase, the maximum tolerated dose established for the combination was avelumab 10 mg/kg every 2 weeks and axitinib 5 mg twice daily. INTERPRETATION The safety profile of the combination avelumab plus axitinib in treatment-naive patients with advanced renal-cell carcinoma seemed to be manageable and consistent with that of each drug alone, and the preliminary data on antitumour activity are encouraging. A phase 3 trial is assessing avelumab and axitinib compared with sunitinib monotherapy. FUNDING Pfizer and Merck.


Cancer Immunology, Immunotherapy | 2014

Definition and application of good manufacturing process-compliant production of CEA-specific chimeric antigen receptor expressing T-cells for phase I/II clinical trial

Ryan D. Guest; Natalia Kirillova; Sam Mowbray; Hannah Gornall; Dominic G. Rothwell; Eleanor J. Cheadle; Eric Austin; Keith Smith; Suzanne M. Watt; Klaus Kühlcke; Nigel Westwood; Fiona Thistlethwaite; Robert E. Hawkins; David E. Gilham

Adoptive cell therapy employing gene-modified T-cells expressing chimeric antigen receptors (CARs) has shown promising preclinical activity in a range of model systems and is now being tested in the clinical setting. The manufacture of CAR T-cells requires compliance with national and European regulations for the production of medicinal products. We established such a compliant process to produce T-cells armed with a first-generation CAR specific for carcinoembryonic antigen (CEA). CAR T-cells were successfully generated for 14 patients with advanced CEA+ malignancy. Of note, in the majority of patients, the defined procedure generated predominantly CD4+ CAR T-cells with the general T-cell population bearing an effector–memory phenotype and high in vitro effector function. Thus, improving the process to generate less-differentiated T-cells would be more desirable in the future for effective adoptive gene-modified T-cell therapy. However, these results confirm that CAR T-cells can be generated in a manner compliant with regulations governing medicinal products in the European Union.

Collaboration


Dive into the Fiona Thistlethwaite's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James Larkin

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Louise Carter

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Matthew Krebs

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Natalie Cook

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Austin

NHS Blood and Transplant

View shared research outputs
Researchain Logo
Decentralizing Knowledge