Network


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

Hotspot


Dive into the research topics where Ailsa Rose is active.

Publication


Featured researches published by Ailsa Rose.


Science Translational Medicine | 2012

Cell Carriage, Delivery, and Selective Replication of an Oncolytic Virus in Tumor in Patients

R. A. Adair; Roulstone; Karen Scott; Ruth Morgan; Gerard J. Nuovo; M Fuller; Debbie Beirne; Emma West; V. A Jennings; Ailsa Rose; Joan Kyula; Sheila Fraser; R. Dave; D. A Anthoney; Alison Merrick; Robin Prestwich; A Aldouri; Oliver Donnelly; Hardev Pandha; Matt Coffey; Peter Selby; Richard Vile; G. J. Toogood; Kevin J. Harrington; Alan Melcher

Oncolytic reovirus is carried by cells to tumors and protected from neutralizing antibodies in the circulation. Therapeutic Virus Hide-and-Seek Oncolytic viruses (OVs) selectively kill cancer cells by direct lysis as well as by stimulating an antitumor immune response. However, the lack of a method for widespread delivery of OVs to tumor cells hangs like an enthusiasm-squelching dark cloud over the field. Direct intratumoral injection is an option but limits this therapy to easily accessible tumors. Mouse studies suggest that the intravenous route would be blocked by preexisting neutralizing antibodies to the virus—the fast immune response that prevents recurrent infection would block the virus from getting to the tumor. Adair et al. now show in human patients with colorectal cancer that, after intravenous injection, reovirus can be escorted to the tumor by immune cells, which protect it from neutralizing antibodies in the plasma. The authors performed a window-of-opportunity clinical trial in 10 colorectal cancer patients scheduled to have surgery to remove liver metastases. Before the planned surgery, the patients were injected with oncolytic reovirus. Replication-competent cytotoxic reovirus was recovered from blood cells, but not from plasma taken from these patients, and reovirus protein was identified preferentially in malignant cells compared with nonmalignant liver tissue from surgical specimens. These data suggest that in contrast to observations in mice, human immune cells may shield reovirus from neutralizing antibodies and deliver the oncolytic reovirus to tumors in patients. Although the mechanism behind the delivery process and the efficacy of the OVs remain to be determined, these potentially cloud-lifting studies support intravenous administration of reovirus for cancer therapy. Oncolytic viruses, which preferentially lyse cancer cells and stimulate an antitumor immune response, represent a promising approach to the treatment of cancer. However, how they evade the antiviral immune response and their selective delivery to, and replication in, tumor over normal tissue has not been investigated in humans. Here, we treated patients with a single cycle of intravenous reovirus before planned surgery to resect colorectal cancer metastases in the liver. Tracking the viral genome in the circulation showed that reovirus could be detected in plasma and blood mononuclear, granulocyte, and platelet cell compartments after infusion. Despite the presence of neutralizing antibodies before viral infusion in all patients, replication-competent reovirus that retained cytotoxicity was recovered from blood cells but not plasma, suggesting that transport by cells could protect virus for potential delivery to tumors. Analysis of surgical specimens demonstrated greater, preferential expression of reovirus protein in malignant cells compared to either tumor stroma or surrounding normal liver tissue. There was evidence of viral factories within tumor, and recovery of replicating virus from tumor (but not normal liver) was achieved in all four patients from whom fresh tissue was available. Hence, reovirus could be protected from neutralizing antibodies after systemic administration by immune cell carriage, which delivered reovirus to tumor. These findings suggest new preclinical and clinical scheduling and treatment combination strategies to enhance in vivo immune evasion and effective intravenous delivery of oncolytic viruses to patients in vivo.


Gene Therapy | 2008

Inflammatory tumour cell killing by oncolytic reovirus for the treatment of melanoma

Fiona Errington; Christine L. White; Katie Twigger; Ailsa Rose; Karen Scott; Lynette Steele; Liz Ilett; Robin Prestwich; Hardev Pandha; Matt Coffey; Peter Selby; Richard G. Vile; Kevin J. Harrington; Alan Melcher

Reovirus is a promising unmodified double-stranded RNA (dsRNA) anti-cancer oncolytic virus, which is thought to specifically target cells with activated Ras. Although reovirus has been tested in a wide range of preclinical models and has entered early clinical trials, it has not previously been tested for the treatment of human melanoma. Here, we show that reovirus effectively kills and replicates in both human melanoma cell lines and freshly resected tumour; intratumoural injection also causes regression of melanoma in a xenograft in vivo model. Reovirus-induced melanoma death is blocked by caspase inhibition and is dependent on constituents of the Ras/RalGEF/p38 pathway. Reovirus melanoma killing is more potent than, and distinct from, chemotherapy or radiotherapy-induced cell death; a range of inflammatory cytokines and chemokines are released by infected tumour cells, while IL-10 secretion is abrogated. Furthermore, the inflammatory response generated by reovirus-infected tumour cells causes bystander toxicity against reovirus-resistant tumour cells and activates human myeloid dendritic cells (DC) in vitro. Hence, reovirus is suitable for clinical testing in melanoma, and may provide a useful danger signal to reverse the immunologically suppressive environment characteristic of this tumour.


Modern Pathology | 2012

Reovirus-associated reduction of microRNA-let-7d is related to the increased apoptotic death of cancer cells in clinical samples

Gerard J. Nuovo; Michela Garofalo; Nicola Valeri; Vicki Roulstone; Stefano Volinia; David E. Cohn; Mitch A. Phelps; Kevin J. Harrington; Richard G. Vile; Alan Melcher; Evanthia Galanis; Sarah Sehl; Rob Adair; Karen Scott; Ailsa Rose; Giles Toogood; Matt Coffey

We analyzed the in situ molecular correlates of infection from cancer patients treated with reovirus. Melanoma, colorectal, and ovarian cancer samples from such patients showed variable infection of the cancer cells but not the intermingled benign cells. RT in situ PCR showed most cancer cells contained the viral genome with threefold less having productive viral infection as documented by either tubulin or reoviral protein co-expression. Productive infection in the cancer cells was strongly correlated with co-expression of p38 and caspase-3 as well as apoptosis-related death (P<0.001). The cancer cell apoptotic death was due to a marked viral-induced inhibition of microRNA-let-7d that, in turn, upregulated caspase-3 activity. In summary, reovirus shows a striking tropism to cancer cells in clinical samples. A rate-limiting factor of reovirus-induced cancer cell death is productive viral infection that operates via the marked reduction of microRNA-let-7d and concomitant elevated caspase-3 expression.


Science Translational Medicine | 2018

Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade

Adel Samson; Karen Scott; David Taggart; Emma West; Erica B. Wilson; Gerard J. Nuovo; Simon Thomson; Robert Corns; Ryan K. Mathew; Martin J. Fuller; Timothy Kottke; Jill Thompson; Elizabeth Ilett; Julia Cockle; Philip van Hille; Gnanamurthy Sivakumar; Euan S. Polson; Samantha Turnbull; Elizabeth S. Appleton; Gemma Migneco; Ailsa Rose; Matt Coffey; Deborah A. Beirne; Fiona Collinson; Christy Ralph; D. Alan Anthoney; Chris Twelves; Andrew J.S. Furness; Sergio A. Quezada; Heiko Wurdak

Intravenous infusion of oncolytic reovirus in patients leads to infection of brain tumors, infiltration by cytotoxic T cells, and up-regulation of PD-L1. Viruses team up with cancer immunotherapy Immune checkpoint inhibitors have shown great promise for cancer therapy, but they do not treat all cancers, and neither breast nor brain tumors are usually treatable with these drugs. However, Bourgeois-Daigneault et al. discovered a way to address this for breast cancer, and Samson et al. discovered a way to address this for brain tumors. In both cases, the authors found that oncolytic virus treatment given early, before surgical resection, alters the antitumor immune response and potentiates the effects of subsequent treatment with immune checkpoint inhibitors. Although these studies differ in the details of their methods and the immune effects induced by the oncolytic viruses, they indicate the potential of such viruses for enhancing the potential of checkpoint therapy and expanding it to new types of cancer. Immune checkpoint inhibitors, including those targeting programmed cell death protein 1 (PD-1), are reshaping cancer therapeutic strategies. Evidence suggests, however, that tumor response and patient survival are determined by tumor programmed death ligand 1 (PD-L1) expression. We hypothesized that preconditioning of the tumor immune microenvironment using targeted, virus-mediated interferon (IFN) stimulation would up-regulate tumor PD-L1 protein expression and increase cytotoxic T cell infiltration, improving the efficacy of subsequent checkpoint blockade. Oncolytic viruses (OVs) represent a promising form of cancer immunotherapy. For brain tumors, almost all studies to date have used direct intralesional injection of OV, because of the largely untested belief that intravenous administration will not deliver virus to this site. We show, in a window-of-opportunity clinical study, that intravenous infusion of oncolytic human Orthoreovirus (referred to herein as reovirus) leads to infection of tumor cells subsequently resected as part of standard clinical care, both in high-grade glioma and in brain metastases, and increases cytotoxic T cell tumor infiltration relative to patients not treated with virus. We further show that reovirus up-regulates IFN-regulated gene expression, as well as the PD-1/PD-L1 axis in tumors, via an IFN-mediated mechanism. Finally, we show that addition of PD-1 blockade to reovirus enhances systemic therapy in a preclinical glioma model. These results support the development of combined systemic immunovirotherapy strategies for the treatment of both primary and secondary tumors in the brain.


BioResearch Open Access | 2012

Reovirus-Mediated Cytotoxicity and Enhancement of Innate Immune Responses Against Acute Myeloid Leukemia

Kathryn Hall; Karen Scott; Ailsa Rose; Michael Desborough; Kevin J. Harrington; Hardev Pandha; Christopher Parrish; Richard G. Vile; Matt Coffey; David Bowen; Fiona Errington-Mais; Alan Melcher

Abstract Reovirus is a naturally occurring oncolytic virus that has shown preclinical efficacy in the treatment of a wide range of tumor types and has now reached phase III testing in clinical trials. The anti-cancer activity of reovirus has been attributed to both its direct oncolytic activity and the enhancement of anti-tumor immune responses. In this study, we have investigated the direct effect of reovirus on acute myeloid leukemia (AML) cells and its potential to enhance innate immune responses against AML, including the testing of primary samples from patients. Reovirus was found to replicate in and kill AML cell lines, and to reduce cell viability in primary AML samples. The pro-inflammatory cytokine interferon alpha (IFNα) and the chemokine (C-C motif) ligand 5 (known as RANTES [regulated upon activation, normal T-cell expressed, and secreted]) were also secreted from AML cells in response to virus treatment. In addition, reovirus-mediated activation of natural killer (NK) cells, within the context of peripheral blood mononuclear cells, stimulated their anti-leukemia response, with increased NK degranulation and IFNγ production and enhanced killing of AML targets. These data suggest that reovirus has the potential as both a direct cytotoxic and an immunotherapeutic agent for the treatment of AML.


Molecular Therapy - Oncolytics | 2017

Oncolytic Herpes Simplex Virus Inhibits Pediatric Brain Tumor Migration and Invasion

Julia Cockle; Anke Brüning-Richardson; Karen Scott; Jill Thompson; Timothy Kottke; Ewan E. Morrison; Azam Ismail; Angel M. Carcaboso; Ailsa Rose; Peter Selby; Joe Conner; Susan Picton; Susan Short; Richard Vile; Alan Melcher; Elizabeth Ilett

Pediatric high-grade glioma (pHGG) and diffuse intrinsic pontine glioma (DIPG) are invasive tumors with poor survival. Oncolytic virotherapy, initially devised as a direct cytotoxic treatment, is now also known to act via immune-mediated mechanisms. Here we investigate a previously unreported mechanism of action: the inhibition of migration and invasion in pediatric brain tumors. We evaluated the effect of oncolytic herpes simplex virus 1716 (HSV1716) on the migration and invasion of pHGG and DIPG both in vitro using 2D (scratch assay, live cell imaging) and 3D (spheroid invasion in collagen) assays and in vivo using an orthotopic xenograft model of DIPG invasion. HSV1716 inhibited migration and invasion in pHGG and DIPG cell lines. pHGG cells demonstrated reduced velocity and changed morphology in the presence of virus. HSV1716 altered pHGG cytoskeletal dynamics by stabilizing microtubules, inhibiting glycogen synthase kinase-3, and preventing localized clustering of adenomatous polyposis coli (APC) to the leading edge of cells. HSV1716 treatment also reduced tumor infiltration in a mouse orthotopic xenograft DIPG model. Our results demonstrate that HSV1716 targets the migration and invasion of pHGG and DIPG and indicates the potential of an oncolytic virus (OV) to be used as a novel anti-invasive treatment strategy for pediatric brain tumors.


International Journal of Oncology | 2012

A Herpesvirus saimiri-based vector expressing TRAIL induces cell death in human carcinoma cell lines and multicellular spheroid cultures

Susan J. Turrell; Stuart Macnab; Ailsa Rose; Alan Melcher; Adrian Whitehouse

Herpesvirus saimiri (HVS) is capable of infecting a range of human carcinoma cell types with high efficiency and the viral genome persists as high copy number, circular, non-integrated episomes which segregate to progeny upon cell division. This allows HVS-based vectors to stably transduce a dividing cell population and provide sustained transgene expression for an extended period of time both in vitro and in vivo. Moreover, the insertion of a bacterial artificial chromosome cassette into the HVS genome simplifies the incorporation of large amounts of heterologous DNA for gene delivery. Herein we have produced a recombinant HVS-based vector containing full-length human TRAIL under the control of the α-survivin promoter, and subsequently challenged a variety of cancer cell lines with this vector. The TRAIL transgene was expressed in infected colorectal SW480 cells, causing considerable apoptosis induction. Apoptosis was also observed when several other cancer cell lines derived from different tissues were infected. Moreover, co-treatment with Jak inhibitor AG490 led to the disruption of spheroid cultures grown from the melanoma Mel888 line. These data suggest that an HVS gene therapy vector expressing TRAIL could be an effective treatment against cancer.


Journal of Clinical Oncology | 2014

Oncolytic wild-type reovirus infection in brain tumors following intravenous administration in patients.

Adel Jebar; Emma West; Karen Scott; Simon Thomson; Robert Corns; Matt Coffey; Ailsa Rose; Gerard J. Nuovo; Matthew Ryan; Fiona Errington-Mais; Christy Ralph; Chris Twelves; Stephen Griffin; Kevin J. Harrington; Hardev Pandha; Olly Donnely; Peter Selby; Richard G. Vile; Susan Short; Alan Melcher


Neuro-oncology | 2016

HG-08ONCOLYTIC HERPES SIMPLEX VIRUS: AN ANTI-INVASIVE THERAPEUTIC STRATEGY FOR PAEDIATRIC HIGH GRADE GLIOMA AND DIFFUSE INTRINSIC PONTINE GLIOMA

Julia Cockle; Elizabeth Ilett; Anke Brüning-Richardson; Jill Thompson; Tim Kottke; Karen Scott; Ewan E. Morrison; Azam Ismail; Ailsa Rose; Susan Picton; Richard Vile; Peter Selby; Susan Short; Alan Melcher


Neuro-oncology | 2015

OP25INTRAVENOUS DELIVERY OF ONCOLYTIC REOLYSIN® TO PRIMARY AND SECONDARY BRAIN TUMOURS

Adel Jebar; Emma West; Karen Scott; G. Nuovo; M. Fuller; Simon Thomson; R. Corns; Ryan K. Mathew; G. SivaKumar; Julia Cockle; P. Van Hille; F. Errington; Elizabeth S. Appleton; G. Migneco; David Taggart; Matt Coffey; Ailsa Rose; Stephen Griffin; Ralph C. Anth

Collaboration


Dive into the Ailsa Rose's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emma West

St James's University Hospital

View shared research outputs
Top Co-Authors

Avatar

Kevin J. Harrington

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Peter Selby

St James's University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge