Network


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

Hotspot


Dive into the research topics where Bruce W. S. Robinson is active.

Publication


Featured researches published by Bruce W. S. Robinson.


Nature Reviews Cancer | 2005

Immunotherapy and chemotherapy--a practical partnership.

Richard A. Lake; Bruce W. S. Robinson

This article discusses how recent data have altered the way we understand how dying tumour cells, particularly those killed by chemotherapy, engage with antitumour immune responses. These data have significant implications for the development of new protocols combining chemotherapy with immunotherapy, indicating an exciting potential for therapeutic synergy with general applicability to many cancer types.


The Lancet | 2003

Mesothelin-family proteins and diagnosis of mesothelioma

Bruce W. S. Robinson; Jenette Creaney; Richard A. Lake; Anna K. Nowak; A. William Musk; Nicholas de Klerk; Pernilla Winzell; Karl Erik Hellström; Ingegerd Hellström

BACKGROUND Mesothelioma is a highly aggressive tumour for which there are no reliable serum tumour markers. Identification of such a marker would be useful in diagnosis of mesothelioma and for monitoring responses to treatment and screening at-risk individuals. METHODS We assayed serum concentrations of soluble mesothelin-related proteins (SMR) using a double determinant (sandwich) ELISA in a blinded study of serum samples from 44 patients with histologically proven mesothelioma; 68 matched healthy controls, 40 of whom had been exposed to asbestos; and 160 patients with other inflammatory or malignant lung and pleural diseases. FINDINGS 37 (84%) of 44 patients with mesothelioma had raised concentrations of SMR at a serum dilution of 1/80, compared with three (2%) of 160 patients with other cancers or other inflammatory lung or pleural diseases, and with none of 28 controls who had not been exposed to asbestos. SMR concentrations correlated with tumour size and increased during tumour progression. Seven of the 40 asbestos-exposed individuals had increased serum concentrations of SMR; three of those seven developed mesothelioma and one developed lung carcinoma within 1-5 years. None of the 33 asbestos-exposed participants whose serum samples had normal concentrations of SMR and who were followed up over 8 years developed mesothelioma. INTERPRETATION Determination of SMR in serum could be a useful marker for diagnosis of mesothelioma and to monitor disease progression. It might also prove helpful for screening asbestos-exposed individuals for early evidence of mesothelioma.


Journal of Immunology | 2003

Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells.

Anna K. Nowak; Richard A. Lake; Amanda L. Marzo; Bernadette Scott; William R. Heath; Edward J. Collins; Jeffrey A. Frelinger; Bruce W. S. Robinson

Cross-presentation of cell-bound Ags from established, solid tumors to CD8 cells is efficient and likely to have a role in determining host response to tumor. A number of investigators have predicted that when tumor Ags are derived from apoptotic cells either no response, due to Ag “sequestration,” or CD8 cross-tolerance would ensue. Because the crucial issue of whether this happens in vivo has never been addressed, we induced apoptosis of established hemagglutinin (HA)-transfected AB1 tumors in BALB/c mice using the apoptosis-inducing reagent gemcitabine. This shrank the tumor by ∼80%. This induction of apoptosis increased cross-presentation of HA to CD8 cells yet neither gross deletion nor functional tolerance of HA-specific CD8 cells were observed, based on tetramer analysis, proliferation of specific CD8 T cells, and in vivo CTL activity. Interestingly, apoptosis primed the host for a strong antitumor response to a second, virus-generated HA-specific signal in that administration of an HA-expressing virus after gemcitabine administration markedly decreased tumor growth compared with viral administration without gemcitabine. Thus tumor cell apoptosis in vivo neither sequesters tumor Ags nor cross-tolerizes tumor-specific CD8 cells. This observation has fundamental consequences for the development of tumor immunotherapy protocols and for understanding T cell reactivity to tumors and the in vivo immune responses to apoptotic cells.


Journal of Clinical Oncology | 1999

Cisplatin and Gemcitabine Treatment for Malignant Mesothelioma: A Phase II Study

Michael J. Byrne; J. A. Davidson; Arthur W. Musk; Joanna Dewar; G. Van Hazel; Martin Buck; N. H. de Klerk; Bruce W. S. Robinson

PURPOSE We performed a phase II study of combined cisplatin 100 mg/m2, given intravenously on day 1, and gemcitabine 1,000 mg/m2, given intravenously on days 1, 8, and 15 of a 28-day cycle for six cycles among patients with advanced measurable pleural mesothelioma. PATIENTS AND METHODS Pleural tumor was measured at three levels on computed tomographic scans at study entry and before the second, fourth, and sixth cycles and every 2 months thereafter to disease progression. Of the 21 patients treated, 19 were male; the median age was 62 years (range, 46 to 74 years); 62% had epithelial tumors; and 18 were classified as tumor-node-metastasis system stage III or IV. Ninety-four cycles were given (median, six; mean, 4.5 per patient), with a mean relative dose intensity of cisplatin 96.7% and gemcitabine 82.5%. RESULTS Best objective responses achieved were as follows: complete response, no patients; partial response, 10 patients (complete response + partial response, 47.6% [95% confidence interval, 26.2% to 69.0%]); no change, nine patients; and progressive disease, two patients. Median response duration was 25 weeks, progression-free survival was 25 weeks, and overall survival was 41 weeks. Nine of the 10 responders (90%) and three of nine patients with no change had significant symptom improvement. Serial measurements of vital capacity were performed on three of the responders; all showed a significant increase during the time of remission. Toxicity was mainly gastroenterologic and hematologic. Grade 3 nausea and vomiting occurred in 33% of patients, grade 3 leukopenia in 38%, grade 3 thrombocytopenia in 14%, and grade 4 thrombocytopenia in 19%. CONCLUSION Combined cisplatin and gemcitabine is an active combination in malignant mesothelioma and produces symptomatic benefit in responding patients.


Journal of Immunology | 2000

Tumor-Specific CD4+ T Cells Have a Major “Post-Licensing” Role in CTL Mediated Anti-Tumor Immunity

Amanda L. Marzo; Beverley F. Kinnear; Richard A. Lake; Jeffrey J. Frelinger; Edward J. Collins; Bruce W. S. Robinson; Bernadette Scott

A number of tumor studies have indicated a link between CD4 help and the magnitude and persistence of CTL activity; however, the mechanisms underlying this have been largely unclear. To evaluate and determine the mechanisms by which CD4+ T cells synergize with CD8+ T cells to prevent tumor growth, we used the novel technique of monitoring in vivo CTL by labeling target cells with CFSE. This approach was supported by the direct visualization of CTL using peptide-MHC tetramers to follow tumor-specific T cells. The data presented demonstrate that while cotransfer of Ag-specific CD4+ T cells was not required for the generation of CTLs, because adoptive transfer of CD8+ T cells alone was sufficient, CD4+ T cells were required for the maintenance of CD8+ T cell numbers. Our data suggest that there is a correlation among the number of CD8+ T cells, in vivo CTL function, and IFN-γ production, with no evidence of a partial or nonresponsive phenotype among tetramer-positive cells. We also show that CD4+ T cells are required for CD8+ T cell infiltration of the tumor.


Nature Reviews Immunology | 2010

Cross-priming in health and disease

Christian Kurts; Bruce W. S. Robinson; Percy A. Knolle

Cross-priming is an important mechanism to activate cytotoxic T lymphocytes (CTLs) for immune defence against viruses and tumours. Although it was discovered more than 25 years ago, we have only recently gained insight into the underlying cellular and molecular mechanisms, and we are just beginning to understand its physiological importance in health and disease. Here we summarize current concepts on the cross-talk between the immune cells involved in CTL cross-priming and on its role in antimicrobial and antitumour defence, as well as in immune-mediated diseases.


Clinical & Experimental Allergy | 1992

Genetic ‘risk’ for atopy is associated with delayed postnatal maturation of T‐cell competence

Patrick G. Holt; J.B. Clough; B.J. Holt; M.J. Baron-Hay; Alison H. Rose; Bruce W. S. Robinson; W.R. Thomas

Recent in vitro studies suggest that IgE production in adults is co‐ordinately regulated by negative signals from γIFN‐producing CD4+ T‐helper‐1 (TH‐1) and positive signals from IL‐4 producing (TH‐2) T‐cells. Additionally, seroepidemiological evidence has pinpointed infancy as the period of maximum lifetime risk for T‐cell sensitization to ubiquitous environmental antigens. The present study sought to elucidate the relationship between these observations, by examination of CD4+ T‐cell function in normal children and those genetically at‘high risk’ for atopy, spanning the age range (up to 4 years) in which IgE responses to environmental allergens is typically manifest. Immunocompetent T‐cell precursor frequencies (determined by cloning at limiting dilution) were markedly reduced in ‘high risk’ children relative to normals (0.53.0.29 vs 0.26.0.19; P= 0.0025). Consistent with reports from other laboratories employing bulk T‐cell culture techniques, the γIFN producing capacity of CD4+ T‐cell clones from both groups of children were markedly reduced relative to adults, and was lowest in the high risk group (P<0.02). IL‐4 production by CD4+ T‐cell clones from the normal children was within the adult range, but again was significantly lower in the high risk group (P<0.00005). This indicates that initial immune responses to environmental allergens in early childhood occur against a background of maturational ‘deficiency’ in CD4+ T‐cell function, and suggests the possibility that variations in the rate of postnatal maturation of T‐cell competence may be a contributing factor in the development of differing patterns of immunological responsiveness to environmental allergens.


British Journal of Cancer | 2002

A multicentre phase II study of cisplatin and gemcitabine for malignant mesothelioma

Anna K. Nowak; Michael J. Byrne; R Williamson; G Ryan; Amanda Segal; D Fielding; Paul Mitchell; Arthur W. Musk; Bruce W. S. Robinson

Our previous phase II study of cisplatin and gemcitabine in malignant mesothelioma showed a 47.6% (95% CI 26.2–69.0%) response rate with symptom improvement in responding patients. Here we confirm these findings in a multicentre setting, and assess the effect of this treatment on quality of life and pulmonary function. Fifty-three patients with pleural malignant mesothelioma received cisplatin 100 mg m−2 i.v. day 1 and gemcitabine 1000 mg m−2 i.v. days 1, 8, and 15 of a 28 day cycle for a maximum of six cycles. Quality of life and pulmonary function were assessed at each cycle. The best response achieved in 52 assessable patients was: partial response, 17 (33%, 95% CI 20–46%); stable disease, 31 (60%); and progressive disease, four (8%). The median time to disease progression was 6.4 months, median survival from start of treatment 11.2 months, and median survival from diagnosis 17.3 months. Vital capacity and global quality of life remained stable in all patients and improved significantly in responding patients. Major toxicities were haematological, limiting the mean relative dose intensity of gemcitabine to 75%. This schedule of cisplatin and gemcitabine is active in malignant mesothelioma in a multicentre setting. Investigation of alternative scheduling is needed to decrease haematological toxicity and increase the relative dose intensity of gemcitabine whilst maintaining response rate and quality of life.


Clinical & Developmental Immunology | 2012

Programmed Death Ligand 2 in Cancer-Induced Immune Suppression

Esdy N. Rozali; Stanleyson V. Hato; Bruce W. S. Robinson; Richard A. Lake; W. Joost Lesterhuis

Inhibitory molecules of the B7/CD28 family play a key role in the induction of immune tolerance in the tumor microenvironment. The programmed death-1 receptor (PD-1), with its ligands PD-L1 and PD-L2, constitutes an important member of these inhibitory pathways. The relevance of the PD-1/PD-L1 pathway in cancer has been extensively studied and therapeutic approaches targeting PD-1 and PD-L1 have been developed and are undergoing human clinical testing. However, PD-L2 has not received as much attention and its role in modulating tumor immunity is less clear. Here, we review the literature on the immunobiology of PD-L2, particularly on its possible roles in cancer-induced immune suppression and we discuss the results of recent studies targeting PD-L2 in cancer.


Journal of Immunology | 2003

IL-2 Intratumoral Immunotherapy Enhances CD8+ T Cells That Mediate Destruction of Tumor Cells and Tumor-Associated Vasculature: A Novel Mechanism for IL-2

Connie Jackaman; Christine Bundell; Beverley F. Kinnear; Alison M. Smith; Pierre Filion; Deborah van Hagen; Bruce W. S. Robinson; Delia J. Nelson

Therapeutic use of IL-2 can generate antitumor immunity; however, a variety of different mechanisms have been reported. We injected IL-2 intratumorally (i.t.) at different stages of growth, using our unique murine model of mesothelioma (AE17; and AE17 transfected with secretory OVA (AE17-sOVA)), and systematically analyzed real-time events as they occurred in vivo. The majority of mice with small tumors when treatment commenced displayed complete tumor regression, remained tumor free for >2 mo, and survived rechallenge with AE17 tumor cells. However, mice with large tumors at the start of treatment failed to respond. Timing experiments showed that IL-2-mediated responses were dependent upon tumor size, not on the duration of disease. Although i.t. IL-2 did not alter tumor Ag presentation in draining lymph nodes, it did enhance a previously primed, endogenous, tumor-specific in vivo CTL response that coincided with regressing tumors. Both CD4+ and CD8+ cells were required for IL-2-mediated tumor eradication, because IL-2 therapy failed in CD4+-depleted, CD8+-depleted, and both CD4+- and CD8+-depleted C57BL/6J animals. Tumor-infiltrating CD8+ T cells, but not CD4+ T cells, increased in association with a marked reduction in tumor-associated vascularity. Destruction of blood vessels required CD8+ T cells, because this did not occur in nude mice or in CD8+-depleted C57BL/6J mice. These results show that repeated doses of i.t. (but not systemic) IL-2 mediates tumor regression via an enhanced endogenous tumor-specific CTL response concomitant with reduced vasculature, thereby demonstrating a novel mechanism for IL-2 activity.

Collaboration


Dive into the Bruce W. S. Robinson's collaboration.

Top Co-Authors

Avatar

Jenette Creaney

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Richard A. Lake

Sir Charles Gairdner Hospital

View shared research outputs
Top Co-Authors

Avatar

Arthur W. Musk

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Anna K. Nowak

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alison H. Rose

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Ian M. Dick

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Amanda Segal

Sir Charles Gairdner Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge