Network


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

Hotspot


Dive into the research topics where Karen Scott is active.

Publication


Featured researches published by Karen Scott.


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.


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 | 2013

Measles virus causes immunogenic cell death in human melanoma

Oliver Donnelly; Fiona Errington-Mais; Lynette Steele; Elizabeth M. Hadac; Victoria Jennings; Karen Scott; Howard Peach; Roger M. Phillips; Jacquelyn Bond; Hardev Pandha; Kevin J. Harrington; Richard G. Vile; Stephen J. Russell; Peter Selby; Alan Melcher

Oncolytic viruses (OV) are promising treatments for cancer, with several currently undergoing testing in randomised clinical trials. Measles virus (MV) has not yet been tested in models of human melanoma. This study demonstrates the efficacy of MV against human melanoma. It is increasingly recognised that an essential component of therapy with OV is the recruitment of host antitumour immune responses, both innate and adaptive. MV-mediated melanoma cell death is an inflammatory process, causing the release of inflammatory cytokines including type-1 interferons and the potent danger signal HMGB1. Here, using human in vitro models, we demonstrate that MV enhances innate antitumour activity, and that MV-mediated melanoma cell death is capable of stimulating a melanoma-specific adaptive immune response.


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.


International Journal of Cancer | 2013

Cytotoxic and immune-mediated killing of human colorectal cancer by reovirus-loaded blood and liver mononuclear cells

Robert A. Adair; Karen Scott; Sheila Fraser; Fiona Errington-Mais; Hardev Pandha; Matt Coffey; Peter Selby; Graham P. Cook; Richard Vile; Kevin J. Harrington; Giles Toogood; Alan Melcher

Reovirus is a promising oncolytic virus, acting by both direct and immune‐mediated mechanisms, although its potential may be limited by inactivation after systemic delivery. Our study addressed whether systemically delivered reovirus might be shielded from neutralising antibodies by cell carriage and whether virus‐loaded blood or hepatic innate immune effector cells become activated to kill colorectal cancer cells metastatic to the liver in human systems. We found that reovirus was directly cytotoxic against tumour cells but not against fresh hepatocytes. Although direct tumour cell killing by neat virus was significantly reduced in the presence of neutralising serum, reovirus was protected when loaded onto peripheral blood mononuclear cells, which may carry virus after intravenous administration in patients. As well as handing off virus for direct oncolytic killing, natural killer (NK) cells within reovirus‐treated blood mononuclear cells were stimulated to kill tumour targets, but not normal hepatocytes, in a Type I interferon‐dependent manner. Similarly, NK cells within liver mononuclear cells became selectively cytotoxic towards tumour cells when activated by reovirus. Hence, intravenous reovirus may evade neutralisation by serum via binding to circulating mononuclear cells, and this blood cell carriage has the potential to investigate both direct and innate immune‐mediated therapy against human colorectal or other cancers metastatic to the liver.


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.


Clinical Cancer Research | 2013

Perioperative Influenza Vaccination Reduces Postoperative Metastatic Disease by Reversing Surgery-Induced Dysfunction in Natural Killer Cells

Lee-Hwa Tai; Jiqing Zhang; Karen Scott; C. T. de Souza; Almohanad A. Alkayyal; Abhirami A. Ananth; Shalini Sahi; Robert A. Adair; Ahmad Bakur Mahmoud; Subash Sad; John C. Bell; Andrew P. Makrigiannis; Alan Melcher; Rebecca C. Auer

Purpose: Surgical removal of solid primary tumors is an essential component of cancer treatment. Surgery-induced dysfunction in natural killer (NK) cells has been linked to the development of metastases in animal models and patients with cancer. We investigated the activation of NK cells using influenza vaccine in the perioperative period to eradicate micrometastatic disease. Experimental Design: Both the B16lacZ and 4T1 tumor models in immunocompetent mice were used to assess the in vivo efficacy of perioperative influenza vaccine administration. In healthy human donors and cancer surgery patients, we assessed NK cell function pre- and post-influenza vaccination using both in vivo and ex vivo assays. Results: Using the TLR3 agonist poly(I:C), we showed as proof-of-principle that perioperative administration of a nonspecific innate immune stimulant can inhibit surgery-induced dysfunction in NK cells and attenuate metastases. Next, we assessed a panel of prophylactic vaccines for NK cell activation and determined that inactivated influenza vaccine was the best candidate for perioperative administration. Perioperative influenza vaccine significantly reduced tumor metastases and improved NK cytotoxicity in preclinical tumor models. Significantly, IFNα is the main cytokine mediator for the therapeutic effect of influenza vaccination. In human studies, influenza vaccine significantly enhanced NK cell activity in healthy human donors and cancer surgery patients. Conclusion: These results provide the preclinical rationale to pursue future clinical trials of perioperative NK cell activation, using vaccination in cancer surgery patients. Research into perioperative immune therapy is warranted to prevent immune dysfunction following surgery and eradicate metastatic disease. Clin Cancer Res; 19(18); 5104–15. ©2013 AACR.


International Journal of Cancer | 2014

Lymphokine-activated killer and dendritic cell carriage enhances oncolytic reovirus therapy for ovarian cancer by overcoming antibody neutralization in ascites

Victoria Jennings; Elizabeth Ilett; Karen Scott; Emma West; Richard Vile; Hardev Pandha; Kevin J. Harrington; A. Young; Geoff Hall; Matt Coffey; Peter Selby; Fiona Errington-Mais; Alan Melcher

Reovirus is an oncolytic virus (OV), which acts by both direct tumor cell killing and priming of antitumor immunity. A major obstacle for effective oncolytic virotherapy is effective delivery of OV to tumor cells. Ovarian cancer is often confined to the peritoneal cavity and therefore i.p. delivery of reovirus may provide the ideal locoregional delivery, avoiding systemic dissemination. However, ovarian cancer is associated with an accumulation of ascitic fluid, which may interfere with oncolytic viral therapy. Here, we investigated the effect of ascites on reovirus‐induced oncolysis against primary ovarian cancer cells and ovarian cancer cell lines. In the absence of ascites, reovirus was cytotoxic against ovarian cancer cells; however, cytotoxicity was abrogated in the presence of ascitic fluid. Neutralizing antibodies (NAb) were identified as the cause of this inhibition. Loading OV onto cell carriers may facilitate virus delivery in the presence of NAb and immune cells which have their own antitumor effector activity are particularly appealing. Immature dendritic cells (iDC), Lymphokine‐activated killer (LAK) cells and LAKDC cocultures were tested as potential carriers for reovirus for tumor cell killing and immune cell priming. Reovirus‐loaded LAKDC, and to a lesser degree iDC, were able to: (i) protect from NAb and hand‐off reovirus for tumor cell killing; (ii) induce a proinflammatory cytokine milieu (IFNɣ, IL‐12, IFNα and TNFα) and (iii) generate an innate and specific antitumor adaptive immune response. Hence, LAKDC pulsed with reovirus represent a novel, clinically practical treatment for ovarian cancer to maximise both direct and innate/adaptive immune‐mediated tumor cell killing.


Viruses | 2015

Evidence for Oncolytic Virotherapy: Where Have We Got to and Where Are We Going?

Samantha Turnbull; Emma West; Karen Scott; Elizabeth S. Appleton; Alan Melcher; Christy Ralph

The last few years have seen an increased interest in immunotherapy in the treatment of malignant disease. In particular, there has been significant enthusiasm for oncolytic virotherapy, with a large amount of pre-clinical data showing promise in animal models in a wide range of tumour types. How do we move forward into the clinical setting and translate something which has such potential into meaningful clinical outcomes? Here, we review how the field of oncolytic virotherapy has developed thus far and what the future may hold.

Collaboration


Dive into the Karen Scott's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emma West

St James's University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Selby

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christy Ralph

St James's University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge