Network


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

Hotspot


Dive into the research topics where Shane Zaidi is active.

Publication


Featured researches published by Shane Zaidi.


Molecular Therapy | 2016

Combination Therapy With Reovirus and Anti-PD-1 Blockade Controls Tumor Growth Through Innate and Adaptive Immune Responses.

Karishma Rajani; Christopher Parrish; Timothy Kottke; Jill Thompson; Shane Zaidi; Liz Ilett; Kevin G. Shim; Rosa Maria Diaz; Hardev Pandha; Kevin J. Harrington; Matt Coffey; Alan Melcher; Richard Vile

Oncolytic reovirus can be delivered both systemically and intratumorally, in both preclinical models and in early phase clinical trials. Reovirus has direct oncolytic activity against a variety of tumor types and antitumor activity is directly associated with immune activation by virus replication in tumors. Immune mechanisms of therapy include both innate immune activation against virally infected tumor cells, and the generation of adaptive antitumor immune responses as a result of in vivo priming against tumor-associated antigens. We tested the combination of local oncolytic reovirus therapy with systemic immune checkpoint inhibition. We show that treatment of subcutaneous B16 melanomas with a combination of intravenous (i.v.) anti-PD-1 antibody and intratumoral (i.t.) reovirus significantly enhanced survival of mice compared to i.t. reovirus (P < 0.01) or anti-PD-1 therapy alone. In vitro immune analysis demonstrated that checkpoint inhibition improved the ability of NK cells to kill reovirus-infected tumor cells, reduced T(reg) activity, and increased the adaptive CD8(+) T-cell-dependent antitumor T-cell response. PD-1 blockade also enhanced the antiviral immune response but through effector mechanisms which overlapped with but also differed from those affecting the antitumor response. Therefore, combination with checkpoint inhibition represents a readily translatable next step in the clinical development of reovirus viroimmunotherapy.


Clinical Cancer Research | 2008

Radiation-Mediated Up-Regulation of Gene Expression from Replication-Defective Adenoviral Vectors: Implications for Sodium Iodide Symporter Gene Therapy

Mohan Hingorani; Christine L. White; Shane Zaidi; Andrew Merron; Inge Peerlinck; Martin Gore; Christopher M. Nutting; Hardev Pandha; Alan Melcher; Richard G. Vile; Georges Vassaux; Kevin J. Harrington

Purpose: To assess the effects of external beam radiotherapy (EBRT) on adenoviral-mediated transgene expression in vitro and in vivo and to define an optimal strategy for combining sodium iodide symporter (NIS)–mediated 131I therapy with EBRT. Experimental Design: Expression of reporter genes [NIS, green fluorescent protein (GFP), β-galactosidase (lacZ), and luciferase (Luc)] from replication-deficient adenoviruses was assessed in tumor cell lines under basal conditions and following irradiation. The effects of viral multiplicity of infection (MOI) and EBRT dose on the magnitude and duration of gene expression were determined. In vivo studies were done with Ad-CMV-GFP and Ad-RSV-Luc. Results: EBRT increased NIS, GFP, and β-galactosidase expression in colorectal, head and neck, and lung cancer cells. Radiation dose and MOI were important determinants of response to EBRT, with greatest effects at higher EBRT doses and lower MOIs. Radiation exerted both transductional (through increased coxsackie-adenoviral receptor and integrin αv) and nontransductional effects, irrespective of promoter sequence (CMV, RSV, hTR, or hTERT). Analysis of the schedule of EBRT followed by viral infection revealed maximal transduction at 24 hours. Radiation maintained increasing radioiodide uptake from Ad-hTR-NIS over 6 days, in direct contrast to reducing levels in unirradiated cells. The effects of EBRT in increasing and maintaining adenovirus-mediated transgene expression were also seen in vivo using GFP- and luciferase-expressing adenoviral vectors. Conclusions: Radiation increased the magnitude and duration of NIS gene expression from replication-deficient adenoviruses. The transductional effect is maximal at 24 hours, but radioiodide uptake is maintained at an elevated level over 6 days after infection.


Radiotherapy and Oncology | 2012

Dose–response analysis of acute oral mucositis and pharyngeal dysphagia in patients receiving induction chemotherapy followed by concomitant chemo-IMRT for head and neck cancer

Shreerang A. Bhide; S. Gulliford; Ulrike Schick; Aisha Miah; Shane Zaidi; K. Newbold; Christopher M. Nutting; Kevin J. Harrington

Dose-response curves (DRCs) and the quantitative parameters describing these curves were generated for grade 3 oral mucositis and dysphagia in 144 patients using individual patient DVHs. Curve fits to the oral mucositis clinical data yielded parameter values of mean dose in 2 Gy equivalent, MD(50) = 51 Gy (95% CI 40-61), slope of the curve, k = 1(95% CI 0.6-1.5). R(2) value for the goodness of fit was 0.80. Fits to the grade 3 dysphagia clinical data yielded parameter values of MD(50) = 44.5 Gy (95% CI 36-53), k = 2.6 (95% CI 0.8-4.5). R(2) value for the goodness of fit was 0.65. This is the first study to derive DRCs in patients receiving induction chemotherapy followed by chemo-radiation (IC-C-IMRT) for head and neck cancer. The dose-response model described in this study could be useful for comparing acute mucositis rates for different dose-fractionation schedules when using IMRT for head and neck cancer.


Gene Therapy | 2013

Synergistic cytotoxicity of oncolytic reovirus in combination with cisplatin-paclitaxel doublet chemotherapy

Victoria Roulstone; Katie Twigger; Shane Zaidi; T. Pencavel; Joan Kyula; Christine L. White; Martin McLaughlin; Rohit Seth; Eleni M. Karapanagiotou; David Mansfield; Matt Coffey; Gerard J. Nuovo; Richard G. Vile; Hardev Pandha; Alan Melcher; Kevin J. Harrington

Oncolytic reovirus is currently under active investigation in a range of tumour types. Early phase studies have shown that this agent has modest monotherapy efficacy and its future development is likely to focus on combination regimens with cytotoxic chemotherapy. Indeed, phase I/II clinical trials have confirmed that reovirus can be safely combined with cytotoxic drugs, including a platin–taxane doublet regimen, which is currently being tested in a phase III clinical trial in patients with relapsed/metastatic head and neck cancer. Therefore, we have tested this triple (reovirus, cisplatin, paclitaxel) combination therapy in a panel of four head and neck cancer cell lines. Using the combination index (CI) method, the triple therapy demonstrated synergistic cytotoxicity in vitro in both malignant and non-malignant cell lines. In head and neck cancer cell lines, this was associated with enhanced caspase 3 and 7 cleavage, but no increase in viral replication. In vitro analyses confirmed colocalisation of markers of reovirus infection and caspase 3. Triple therapy was significantly more effective than reovirus or cisplatin–paclitaxel in athymic nude mice. These data suggest that the combination of reovirus plus platin–taxane doublet chemotherapy has significant activity in head and neck cancer and underpin the current phase III study in this indication.


Current Drug Discovery Technologies | 2009

Novel Targeted Radiosensitisers in Cancer Treatment

Shane Zaidi; Robert Huddart; Kevin J. Harrington

The last few years have seen a significant increase in our understanding of the molecular pathways governing cell function in cancer. This has led to an explosive interest in novel molecularly-targeted agents and, until recently, the focus of research effort has been to combine these agents with conventional cytotoxic chemotherapy. However, following a recent trial of an anti-EGFR targeted antibody in combination with radiation, a new paradigm is emerging in which these novel agents will be combined with external beam radiotherapy (RT). In this article we review classes of novel targeted radiosensitisers that are directed at specific aspects of cell function. Such agents are aimed at either single or multiple targets (the latter is a more attractive approach in view of cross-talk between different cell signaling pathways). We review available preclinical and clinical literature with a particular focus on novel agents targeting components of the ErbB and IGF-1R family cell signaling pathways. In this model, radiosensitisers can exert their effects at the cell membrane surface by preventing receptor activation or by interfering with the function of second messengers such as the Ras/PI3K/mTOR pathway. In addition, the effects of novel DNA repair inhibitors will be considered in the context of combination strategies with signal transduction pathway blockade. Other small molecule inhibitors, such as HSP90 inhibitors, that can disrupt signaling in a number of different pathways, will also be discussed. Ultimately, through the synergistic use of these innovative molecules and RT, the therapeutic index may be enhanced by modulating cellular metabolism, proliferation, repair, angiogenesis, and apoptosis. The rapid proliferation of available targeted agents and their entry into phase I clinical trials means that this is an extremely interesting area for research in radiation oncology.


Molecular Therapy | 2014

Cytokine Conditioning Enhances Systemic Delivery and Therapy of an Oncolytic Virus

Elizabeth Ilett; Timothy Kottke; Oliver Donnelly; Jill Thompson; Candice Willmon; Rosa Maria Diaz; Shane Zaidi; Matt Coffey; Peter Selby; Kevin J. Harrington; Hardev Pandha; Alan Melcher; Richard Vile

Optimum clinical protocols require systemic delivery of oncolytic viruses in the presence of an intact immune system. We show that preconditioning with immune modulators, or loading virus onto carrier cells ex vivo, enhances virus-mediated antitumor activity. Our early trials of systemic reovirus delivery showed that after infusion reovirus could be recovered from blood cells--but not from plasma--suggesting that rapid association with blood cells may protect virus from neutralizing antibody. We therefore postulated that stimulation of potential carrier cells directly in vivo before intravenous viral delivery would enhance delivery of cell-associated virus to tumor. We show that mobilization of the CD11b(+) cell compartment by granulocyte macrophage-colony stimulating factor immediately before intravenous reovirus, eliminated detectable tumor in mice with small B16 melanomas, and achieved highly significant therapy in mice bearing well-established tumors. Unexpectedly, cytokine conditioning therapy was most effective in the presence of preexisting neutralizing antibody. Consistent with this, reovirus bound by neutralizing antibody effectively accessed monocytes/macrophages and was handed off to tumor cells. Thus, preconditioning with cytokine stimulated recipient cells in vivo for enhanced viral delivery to tumors. Moreover, preexisting neutralizing antibody to an oncolytic virus may, therefore, even be exploited for systemic delivery to tumors in the clinic.


Neuro-oncology | 2016

Combination viroimmunotherapy with checkpoint inhibition to treat glioma, based on location-specific tumor profiling.

Julia Cockle; Karishma Rajani; Shane Zaidi; Timothy Kottke; Jill Thompson; Rosa Maria Diaz; Kevin G. Shim; Tim Peterson; Ian F. Parney; Susan Short; Peter Selby; Elizabeth Ilett; Alan Melcher; Richard G. Vile

BACKGROUND Systemic delivery of a complementary cDNA library expressed from the vesicular stomatitis virus (VSV) treats tumors by vaccinating against a wide range of tumor associated antigens (TAAs). For subcutaneous B16 melanomas, therapy was achieved using a specific combination of self-TAAs (neuroblastoma-Ras, cytochrome c, and tyrosinase-related protein 1) expressed from VSV. However, for intracranial B16 tumors, a different combination was therapeutic (consisting of VSV-expressed hypoxia-inducible factor [HIF]-2α, Sox-10, c-Myc, and tyrosinase-related protein 1). Therefore, we tested the hypothesis that tumors of different histological types growing in the brain share a common immunogenic signature which can be exploited for immunotherapy. METHODS Syngeneic tumors, including GL261 gliomas, in the brains of immune competent mice were analyzed for their antigenic profiles or were treated with systemic viroimmunotherapy. RESULTS Several different histological types of tumors growing intracranially, as well as freshly resected human brain tumor explants, expressed a HIF-2α(Hi) phenotype imposed by brain-derived CD11b+ cells. This location-specific antigen expression was exploited therapeutically against intracranial GL261 gliomas using systemically delivered VSV expressing HIF-2α, Sox-10, and c-Myc. Viroimmunotherapy was enhanced by immune checkpoint inhibitors, associated with the de-repression of antitumor T-helper cell type 1 (Th1) interferon-γ and Th17 T cell responses. CONCLUSIONS Since different tumor types growing in the same location in the brain share a location-specific phenotype, we suggest that antigen-specific immunotherapies should be based upon expression of both histological type-specific tumor antigens and location-specific antigens. Our findings support clinical application of VSV-TAA therapy with checkpoint inhibition for aggressive brain tumors and highlight the importance of the intracranial microenvironment in sculpting a location-specific profile of tumor antigen expression.


Advances in Anatomic Pathology | 2016

Dedifferentiated Liposarcoma: Updates on Morphology, Genetics, and Therapeutic Strategies

Khin Thway; Robin L. Jones; Jonathan Noujaim; Shane Zaidi; Aisha Miah; Cyril Fisher

Well-differentiated liposarcoma (WDL) and dedifferentiated liposarcoma (DDL) form the largest subgroup of liposarcomas, and represent a morphologic and behavioral spectrum of 1 disease entity, which arises typically in middle to late adult life, most frequently within the retroperitoneum or extremities. DDL is defined as nonlipogenic sarcoma that is juxtaposed to WDL, occurs as a recurrence of WDL or which can arise de novo, and typically has the appearance of undifferentiated pleomorphic or spindle cell sarcoma. DDL have a propensity for local recurrence, whereas distant metastasis is rarer, and behavior is related to anatomic site, with retroperitoneal neoplasms showing a significantly worse prognosis. Surgical resection remains the mainstay of treatment, and medical options for patients with aggressive recurrent or metastatic disease are limited. DDL share similar genetic abnormalities to WDL, with high-level amplifications of chromosome 12q14-15, including the MDM2 and CDK4 cell cycle oncogenes, and DDL harbor additional genetic changes, particularly coamplifications of 6q23 and 1p32. Novel therapies targeted at the gene products of chromosome 12 are being tested in clinical trials. We review the pathology and genetics of DDL, discussing morphologic patterns, immunohistochemical and genetic findings, the differential diagnosis, and future therapeutic strategies.


Molecular Therapy | 2010

Therapeutic Effect of Sodium Iodide Symporter Gene Therapy Combined With External Beam Radiotherapy and Targeted Drugs That Inhibit DNA Repair

Mohan Hingorani; Christine L. White; Shane Zaidi; Hardev Pandha; Alan Melcher; Shreerang A. Bhide; Christopher M. Nutting; Konstantinos Syrigos; Richard Vile; Georges Vassaux; Kevin J. Harrington

Adenoviral (AdV) transfer of sodium iodide symporter (NIS) gene has translational potential, but relatively low levels of transduction and subsequent radioisotope uptake limit the efficacy of the approach. In previous studies, we showed that combining NIS gene delivery with external beam radiotherapy (EBRT) and DNA damage repair inhibitors increased viral gene expression and radioiodide uptake. Here, we report the therapeutic efficacy of this strategy. An adenovirus expressing NIS from a telomerase promoter (Ad-hTR-NIS) was cytotoxic combined with relatively high-dose (50 microCi) (131)I therapy and enhanced the efficacy of EBRT combined with low-dose (10 and 25 microCi) (131)I therapy in colorectal and head and neck cancer cells. Combining this approach with ataxia-telangiectasia mutated (ATM) or DNA-dependent protein kinase (DNA-PK) inhibition caused maintenance of double-stranded DNA breaks (DSBs) at 24 hours and increased cytotoxicity on clonogenic assay. When the triplet of NIS-mediated (131)I therapy, EBRT, and DNA-PKi was used in vivo, 90% of mice were tumor-free at 5 weeks. Acute radiation toxicity in the EBRT field was not exacerbated. In contrast, DNA-PKi did not enhance the therapeutic efficacy of EBRT plus adenovirus-mediated HSVtk/ganciclovir (GCV). Therefore, combining NIS gene therapy and EBRT represents an ideal strategy to exploit the therapeutic benefits of novel radiosensitizers.


Oncogene | 2014

Synergistic cytotoxicity of radiation and oncolytic Lister strain vaccinia in V600D/E BRAF mutant melanoma depends on JNK and TNF-α signaling

Joan Kyula; Aadil A. Khan; David Mansfield; Eleni M. Karapanagiotou; Martin McLaughlin; Roulstone; Shane Zaidi; T. Pencavel; Yann Touchefeu; Rohit Seth; Nanhai G. Chen; Y. A Yu; Q Zhang; Alan Melcher; Richard G. Vile; Hardev Pandha; M Ajaz; Aladar A. Szalay; Kevin J. Harrington

Melanoma is an aggressive skin cancer that carries an extremely poor prognosis when local invasion, nodal spread or systemic metastasis has occurred. Recent advances in melanoma biology have revealed that RAS-RAF-MEK-ERK signaling has a pivotal role in governing disease progression and treatment resistance. Proof-of-concept clinical studies have shown that direct BRAF inhibition yields impressive responses in advanced disease but these are short-lived as treatment resistance rapidly emerges. Therefore, there is a pressing need to develop new targeted strategies for BRAF mutant melanoma. As such, oncolytic viruses represent a promising cancer-specific approach with significant activity in melanoma. This study investigated interactions between genetically-modified vaccinia virus (GLV-1h68) and radiotherapy in melanoma cell lines with BRAF mutant, Ras mutant or wild-type genotype. Preclinical studies revealed that GLV-1h68 combined with radiotherapy significantly increased cytotoxicity and apoptosis relative to either single agent in V600DBRAF/V600EBRAF mutant melanoma in vitro and in vivo. The mechanism of enhanced cytotoxicity with GLV-1h68/radiation (RT) was independent of viral replication and due to attenuation of JNK, p38 and ERK MAPK phosphorylation specifically in BRAF mutant cells. Further studies showed that JNK pathway inhibition sensitized BRAF mutant cells to GLV-1h68-mediated cell death, mimicking the effect of RT. GLV-1h68 infection activated MAPK signaling in V600DBRAF/V600EBRAF mutant cell lines and this was associated with TNF-α secretion which, in turn, provided a prosurvival signal. Combination GLV-1h68/RT (or GLV-1h68/JNK inhibition) caused abrogation of TNF-α secretion. These data provide a strong rationale for combining GLV-1h68 with irradiation in V600D/EBRAF mutant tumors.

Collaboration


Dive into the Shane Zaidi's collaboration.

Top Co-Authors

Avatar

Kevin J. Harrington

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Aisha Miah

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Khin Thway

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Robin L. Jones

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Selby

St James's University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge