Network


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

Hotspot


Dive into the research topics where Robin Prestwich is active.

Publication


Featured researches published by Robin Prestwich.


Human Gene Therapy | 2009

The Case of Oncolytic Viruses Versus the Immune System: Waiting on the Judgment of Solomon

Robin Prestwich; Fiona Errington; Rosa Maria Diaz; Hardev Pandha; Kevin J. Harrington; Alan Melcher; Richard Vile

The three-way interaction between oncolytic viruses, the tumor microenvironment, and the immune system is critical to the outcome of antitumor therapy. Classically, the immune system is thought to limit the efficacy of therapy, leading to viral clearance. However, preclinical and clinical data suggest that in some cases virotherapy may in fact act as cancer immunotherapy. In this review we discuss the ability of oncolytic viruses to alter the immunogenic milieu of the tumor microenvironment, and the role of innate and adaptive immunity in both restricting and augmenting therapy. Strategies to improve virotherapy by immunomodulation, including suppression or enhancement of the innate and adaptive responses, are discussed.


Journal of Immunology | 2008

Reovirus activates human dendritic cells to promote innate antitumor immunity

Fiona Errington; Lynette Steele; Robin Prestwich; Kevin J. Harrington; Hardev Pandha; L. Vidal; Johann S. de Bono; Peter Selby; Matt Coffey; Richard Vile; Alan Melcher

Oncolytic viruses can exert their antitumor activity via direct oncolysis or activation of antitumor immunity. Although reovirus is currently under clinical investigation for the treatment of localized or disseminated cancer, any potential immune contribution to its efficacy has not been addressed. This is the first study to investigate the ability of reovirus to activate human dendritic cells (DC), key regulators of both innate and adaptive immune responses. Reovirus induced DC maturation and stimulated the production of the proinflammatory cytokines IFN-α, TNF-α, IL-12p70, and IL-6. Activation of DC by reovirus was not dependent on viral replication, while cytokine production (but not phenotypic maturation) was inhibited by blockade of PKR and NF-κB signaling. Upon coculture with autologous NK cells, reovirus-activated DC up-regulated IFN-γ production and increased NK cytolytic activity. Moreover, short-term coculture of reovirus-activated DC with autologous T cells also enhanced T cell cytokine secretion (IL-2 and IFN-γ) and induced non-Ag restricted tumor cell killing. These data demonstrate for the first time that reovirus directly activates human DC and that reovirus-activated DC stimulate innate killing by not only NK cells, but also T cells, suggesting a novel potential role for T cells in oncolytic virus-induced local tumor cell death. Hence reovirus recognition by DC may trigger innate effector mechanisms to complement the virus’s direct cytotoxicity, potentially enhancing the efficacy of reovirus as a therapeutic agent.


Expert Review of Anticancer Therapy | 2008

Oncolytic viruses: a novel form of immunotherapy

Robin Prestwich; Kevin J. Harrington; Hardev Pandha; Richard Vile; Alan Melcher; Fiona Errington

Oncolytic viruses are novel anticancer agents, currently under investigation in Phase I–III clinical trials. Until recently, most studies have focused on the direct antitumor properties of these viruses, although there is now an increasing body of evidence that the host immune response may be critical to the efficacy of oncolytic virotherapy. This may be mediated via innate immune effectors, adaptive antiviral immune responses eliminating infected cells or adaptive antitumor immune responses. This report summarizes preclinical and clinical evidence for the importance of immune interactions, which may be finely balanced between viral and tumor elimination. On this basis, oncolytic viruses represent a promising novel immunotherapy strategy, which may be optimally combined with existing therapeutic modalities.


Clinical Cancer Research | 2009

Immune-Mediated Antitumor Activity of Reovirus Is Required for Therapy and Is Independent of Direct Viral Oncolysis and Replication

Robin Prestwich; Elizabeth Ilett; Fiona Errington; Rosa Maria Diaz; Lynette Steele; Tim Kottke; Jill Thompson; Feorillo Galivo; Kevin J. Harrington; Hardev Pandha; Peter Selby; Richard G. Vile; Alan Melcher

Purpose: Reovirus is a naturally occurring oncolytic virus in clinical trials. Although tumor infection by reovirus can generate adaptive antitumor immunity, its therapeutic importance versus direct viral oncolysis is undefined. This study addresses the requirement for viral oncolysis and replication, and the relative importance of antitumor immunity and direct oncolysis in therapy. Experimental Design: Nonantigen specific T cells loaded with reovirus were delivered i.v. to C57BL/6 and severe combined immunodeficient mice bearing lymph node and splenic metastases from the murine melanoma, B16ova, with assessment of viral replication, metastatic clearance by tumor colony outgrowth, and immune priming. Human cytotoxic lymphocyte priming assays were done with dendritic cells loaded with Mel888 cells before the addition of reovirus. Results: B16ova was resistant to direct oncolysis in vitro, and failed to support reovirus replication in vitro or in vivo. Nevertheless, reovirus purged lymph node and splenic metastases in C57BL/6 mice and generated antitumor immunity. In contrast, reovirus failed to reduce tumor burden in severe combined immunodeficient mice bearing either B16ova or reovirus-sensitive B16tk metastases. In the human system, reovirus acted solely as an adjuvant when added to dendritic cells already loaded with Mel888, supporting priming of specific antitumor cytotoxic lymphocyte, in the absence of significant direct tumor oncolysis; UV-treated nonreplicating reovirus was similarly immunogenic. Conclusion: The immune response is critical in mediating the efficacy of reovirus, and does not depend upon direct viral oncolysis or replication. The findings are of direct relevance to fulfilling the potential of this novel anticancer agent.


Clinical Cancer Research | 2008

Tumor Infection by Oncolytic Reovirus Primes Adaptive Antitumor Immunity

Robin Prestwich; Fiona Errington; Elizabeth Ilett; Ruth Morgan; Karen Scott; Timothy Kottke; Jill Thompson; Ewan E. Morrison; Kevin J. Harrington; Hardev Pandha; Peter Selby; Richard G. Vile; Alan Melcher

Purpose: Early clinical trials are under way exploring the direct oncolytic potential of reovirus. This study addresses whether tumor infection by reovirus is also able to generate bystander, adaptive antitumor immunity. Experimental Design: Reovirus was delivered intravenously to C57BL/6 mice bearing lymph node metastases from the murine melanoma, B16-tk, with assessment of nodal metastatic clearance, priming of antitumor immunity against the tumor-associated antigen tyrosinase-related protein-2, and cytokine responses. In an in vitro human system, the effect of reovirus infection on the ability of Mel888 melanoma cells to activate and load dendritic cells for cytotoxic lymphocyte (CTL) priming was investigated. Results: In the murine model, a single intravenous dose of reovirus reduced metastatic lymph node burden and induced antitumor immunity (splenocyte response to tyrosinase-related protein-2 and interleukin-12 production in disaggregated lymph nodes). In vitro human assays revealed that uninfected Mel888 cells failed to induce dendritic cell maturation or support priming of an anti-Mel888 CTL response. In contrast, reovirus-infected Mel888 cells (reo-Mel) matured dendritic cells in a reovirus dose-dependent manner. When cultured with autologous peripheral blood lymphocytes, dendritic cells loaded with reo-Mel induced lymphocyte expansion, IFN-γ production, specific anti-Mel888 cell cytotoxicity, and cross-primed CD8+ T cells specific against the human tumor-associated antigen MART-1. Conclusion: Reovirus infection of tumor cells reduces metastatic disease burden and primes antitumor immunity. Future clinical trials should be designed to explore both direct cytotoxic and immunotherapeutic effects of reovirus.


Molecular Therapy | 2009

Cell carriers for oncolytic viruses: Fed Ex for cancer therapy.

Candice Willmon; Kevin J. Harrington; Timothy Kottke; Robin Prestwich; Alan Melcher; Richard Vile

Oncolytic viruses delivered directly into the circulation face many hazards that impede their localization to, and infection of, metastatic tumors. Such barriers to systemic delivery could be overcome if couriers, which confer both protection, and tumor localization, to their viral cargoes, could be found. Several preclincal studies have shown that viruses can be loaded into, or onto, different types of cells without losing the biological activity of either virus or cell carrier. Importantly, such loading can significantly protect the viruses from immune-mediated virus-neutralizing activities, including antiviral antibody. Moreover, an impressive portfolio of cellular vehicles, which have some degree of tropism for tumor cells themselves, or for the biological properties associated with the tumor stroma, is already available. Therefore, it will soon be possible to initiate clinical protocols to test the hypopthesis that cell-mediated delivery can permit efficient shipping of oncolytic viruses from the loading bay (the production laboratory) directly to the tumor in immune-competent patients with metastatic disease.


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.


Clinical Cancer Research | 2010

Two-Stage Phase I Dose-Escalation Study of Intratumoral Reovirus Type 3 Dearing and Palliative Radiotherapy in Patients with Advanced Cancers

Kevin J. Harrington; Eleni M. Karapanagiotou; Victoria Roulstone; Katie Twigger; Christine L. White; L. Vidal; Debbie Beirne; Robin Prestwich; Kate Newbold; Merina Ahmed; Khin Thway; Christopher M. Nutting; Matt Coffey; Dean Harris; Richard Vile; Hardev Pandha; Johann S. DeBono; Alan Melcher

Purpose: To determine the safety and feasibility of combining intratumoral reovirus and radiotherapy in patients with advanced cancer and to assess viral biodistribution, reoviral replication in tumors, and antiviral immune responses. Experimental Design: Patients with measurable disease amenable to palliative radiotherapy were enrolled. In the first stage, patients received radiotherapy (20 Gy in five fractions) plus two intratumoral injections of RT3D at doses between 1 × 108 and 1 × 1010 TCID50. In the second stage, the radiotherapy dose was increased (36 Gy in 12 fractions) and patients received two, four, or six doses of RT3D at 1 × 1010 TCID50. End points were safety, viral replication, immunogenicity, and antitumoral activity. Results: Twenty-three patients with various solid tumors were treated. Dose-limiting toxicity was not seen. The most common toxicities were grade 2 (or lower) pyrexia, influenza-like symptoms, vomiting, asymptomatic lymphopenia, and neutropenia. There was no exacerbation of the acute radiation reaction. Reverse transcription-PCR (RT-PCR) studies of blood, urine, stool, and sputum were negative for viral shedding. In the low-dose (20 Gy in five fractions) radiation group, two of seven evaluable patients had a partial response and five had stable disease. In the high-dose (36 Gy in 12 fractions) radiation group, five of seven evaluable patients had partial response and two stable disease. Conclusions: The combination of intratumoral RT3D and radiotherapy was well tolerated. The favorable toxicity profile and lack of vector shedding means that this combination should be evaluated in newly diagnosed patients receiving radiotherapy with curative intent. Clin Cancer Res; 16(11); 3067–77. ©2010 AACR.


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.


Gene Therapy | 2009

Dendritic cells and T cells deliver oncolytic reovirus for tumour killing despite pre-existing anti-viral immunity.

Elizabeth Ilett; Robin Prestwich; Timothy Kottke; Fiona Errington; Jill Thompson; Kevin J. Harrington; Hardev Pandha; Matt Coffey; Peter Selby; Richard G. Vile; Alan Melcher

Reovirus is a naturally occurring oncolytic virus currently in early clinical trials. However, the rapid induction of neutralizing antibodies represents a major obstacle to successful systemic delivery. This study addresses, for the first time, the ability of cellular carriers in the form of T cells and dendritic cells (DC) to protect reovirus from systemic neutralization. In addition, the ability of these cellular carriers to manipulate the subsequent balance of anti-viral versus anti-tumour immune response is explored. Reovirus, either neat or loaded onto DC or T cells, was delivered intravenously into reovirus-naive or reovirus-immune C57Bl/6 mice bearing lymph node B16tk melanoma metastases. Three and 10 days after treatment, reovirus delivery, carrier cell trafficking, metastatic clearance and priming of anti-tumour/anti-viral immunity were assessed. In naive mice, reovirus delivered either neat or through cell carriage was detectable in the tumour-draining lymph nodes 3 days after treatment, though complete clearance of metastases was only obtained when the virus was delivered on T cells or mature DC (mDC); neat reovirus or loaded immature DC (iDC) gave only partial early tumour clearance. Furthermore, only T cells carrying reovirus generated anti-tumour immune responses and long-term tumour clearance; reovirus-loaded DC, in contrast, generated only an anti-viral immune response. In reovirus-immune mice, however, the results were different. Neat reovirus was completely ineffective as a therapy, whereas mDC—though not iDC—as well as T cells, effectively delivered reovirus to melanoma in vivo for therapy and anti-tumour immune priming. Moreover, mDC were more effective than T cells over a range of viral loads. These data show that systemically administered neat reovirus is not optimal for therapy, and that DC may be an appropriate vehicle for carriage of significant levels of reovirus to tumours. The pre-existing immune status against the virus is critical in determining the balance between anti-viral and anti-tumour immunity elicited when reovirus is delivered by cell carriage, and the viral dose and mode of delivery, as well as the immune status of patients, may profoundly affect the success of any clinical anti-tumour viral therapy. These findings are therefore of direct translational relevance for the future design of clinical trials.

Collaboration


Dive into the Robin Prestwich's collaboration.

Top Co-Authors

Avatar

Mehmet Sen

Dokuz Eylül University

View shared research outputs
Top Co-Authors

Avatar

Andrew Scarsbrook

St James's University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin J. Harrington

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fiona Errington

St James's University Hospital

View shared research outputs
Top Co-Authors

Avatar

K.E. Dyker

St James's University Hospital

View shared research outputs
Top Co-Authors

Avatar

Peter Selby

St James's University Hospital

View shared research outputs
Top Co-Authors

Avatar

R. Speight

Leeds Teaching Hospitals NHS Trust

View shared research outputs
Top Co-Authors

Avatar

M. Sen

St James's University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge