Network


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

Hotspot


Dive into the research topics where Kathy Gratton is active.

Publication


Featured researches published by Kathy Gratton.


Blood | 2011

Integrin β7-mediated regulation of multiple myeloma cell adhesion, migration, and invasion.

Paola Neri; Li Ren; Abdel Kareem Azab; Matthew Brentnall; Kathy Gratton; Alexander C. Klimowicz; Charles P. Lin; Peter Duggan; Pierfrancesco Tassone; Adnan Mansoor; Douglas A. Stewart; Lawrence H. Boise; Irene M. Ghobrial; Nizar J. Bahlis

Integrin-β7 (ITGB7) mRNA is detected in multiple myeloma (MM) cells and its presence is correlated with MAF gene activation. Although the involvement of several integrin family members in MM-stoma cell interaction is well documented, the specific biologic functions regulated by integrin-β7 in MM are largely unknown. Clinically, we have correlated integrin-β7 expression in MM with poor survival outcomes post autologous stem cell transplantation and postsalvage therapy with bortezomib. Functionally, we have found that shRNA-mediated silencing of ITGB7 reduces MM-cell adhesion to extra-cellular matrix elements (fibronectin, E-cadherin) and reverses cell-adhesion-mediated drug resistance (CAM-DR) sensitizing them to bortezomib and melphalan. In addition, ITGB7 silencing abrogated MM-cell transwell migration in response to SDF1α gradients, reduced vessel density in xenografted tumors, and altered MM cells in vivo homing into the BM. Mechanistically, ITGB7 knockdown inhibited focal adhesion kinase (FAK) and Src phosphorylation, Rac1 activation, and SUMOylation, reduced VEGF production in MM-BM stem cell cocultures and attenuated p65-NF-κB activity. Our findings support a role for integrin-β7 in MM-cell adhesion, migration, and BM homing, and pave the way for a novel therapeutic approach targeting this molecule.


Human Genetics | 1998

Localization of a gene for incomplete X-linked congenital stationary night blindness to the interval between DXS6849 and DXS8023 in Xp11.23

N. T. Bech-Hansen; Kym M. Boycott; Kathy Gratton; Daniel Ross; L. Leigh Field; William Pearce

Congenital stationary night blindness (CSNB) is a nonprogressive retinal disorder characterized by night blindness, nystagmus, myopia, a variable decrease in visual acuity, an abnormal electroretinographic response, and a disturbance in dark adaptation. Two forms of X-linked CSNB have been defined, complete CSNB in which rod function is extinguished, and incomplete CSNB in which rod function is reduced but not extinguished, as seen by electroretinography and dark adaptometry. In studying a large family of Mennonite ancestry, we have confirmed linkage between the locus (CSNB2) for incomplete CSNB and genetic markers in the Xp11 region. In particular, lod scores of 12.25 and 15.26 at zero recombination were observed between CSNB2 and the markers DXS573 and DXS255. Detailed analysis of critical recombinant chromosomes in this extended family have refined the minimal region for the CSNB2 locus to the interval between DXS6849 and DXS8023 in Xp11.23.


Cancer Research | 2016

Abstract 541: Successful oncolytic virotherapy in a bortezomib resistant syngeneic mouse model of multiple myeloma: Implications for translational significance

Chandini M. Thirukkumaran; Zhong Qiao Shi; Joanne Luider; Karen Kopciuk; Marta Chesi; Leif Bergsagel; Yuan Dong; Chunfen Zhang; Ahmed E. Mostafa; Kathy Gratton; Satbir Thakur; Don M. Morris

Introduction: Multiple myeloma (MM) is a malignancy of plasma cells that is still considered incurable due to the 90% relapse rate in patients. Despite the advent of new agents the majority of MM patients relapse secondary to therapy resistance. The potential of reovirus (RV) as a novel therapeutic agent for MM under in vitro, in vivo and ex vivo conditions has been demonstrated by us and a phase I clinical trial of MM with RV therapy has shown indications of efficacy. Recently we have shown that RV could synergize with several standard of care MM drugs such as bortezomib (BTZ), carfilzomib (CFZ) and dexamethasone and importantly, enhance its therapeutic potential in therapy resistant human MM cell lines in vitro. Utilizing the syngeneic Vk*MYC BTZ resistant transplantable MM mouse model, we demonstrate that mice harbouring BTZ insensitive MM tumors significantly respond to RV treatment. Our data indicate that this RV treatment sensitivity is manifested via, direct oncolysis in conjunction with immune modulatory effects. Methods: Vk*MYC myeloma cells (Vk12598) were injected via tail vein (IV) into 3 groups of C57BL/6 wt recipient mice. Seven days post tumor injection, mice were treated with PBS (vehicle) or live (LV) or dead reovirus (DV) (5×10⁁8 PFU) administered IV every 6 days in a total of 5 doses. Serum gamma globulins (M-spike) were assessed weekly by serum protein electrophoresis. Mice were sacrificed on day 35 post tumour injection and their spleens and bone marrow (BM) were harvested. Splenic and BM cells were stained for CD45+/CD45R-/CD138+ (MM tumors), CD4+/CD8+ (T cells), CD3+/NKG2D+ (NKT cells), CD11b+Ly6C+ (monocytic myeloid derived suppressor cells (Mo-MDSC) and CD11b+Ly6G+ (polymorphonuclear (PMN) - MDSCs) and analyzed by flow cytometry. Results: Mice treated with RV demonstrated highly significant (P Conclusions: Our results indicate reovirotherapy is successful in a validated syngeneic BTZ resistant MM model. These results have important implications for future clinical trials. Currently we are conducting further experiments to assess the immune modulatory (such as T - memory and T - regulatory cells) contributions of RV in this model. Citation Format: Chandini M. Thirukkumaran, Zhong Qiao Shi, Joanne Luider, Karen Kopciuk, Marta Chesi, Leif Bergsagel, Yuan Dong, Chunfen Zhang, Ahmed Mostafa, Kathy Gratton, Satbir Thakur, Don Morris. Successful oncolytic virotherapy in a bortezomib resistant syngeneic mouse model of multiple myeloma: Implications for translational significance. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 541.


Cancers | 2018

Oncolytic Reovirus and Immune Checkpoint Inhibition as a Novel Immunotherapeutic Strategy for Breast Cancer

Ahmed E. Mostafa; Daniel E. Meyers; Chandini M. Thirukkumaran; Peter Liu; Kathy Gratton; Jason C. L. Spurrell; Qiao Shi; Satbir Thakur; Don M. Morris

As the current efficacy of oncolytic viruses (OVs) as monotherapy is limited, exploration of OVs as part of a broader immunotherapeutic treatment strategy for cancer is necessary. Here, we investigated the ability for immune checkpoint blockade to enhance the efficacy of oncolytic reovirus (RV) for the treatment of breast cancer (BrCa). In vitro, oncolysis and cytokine production were assessed in human and murine BrCa cell lines following RV exposure. Furthermore, RV-induced upregulation of tumor cell PD-L1 was evaluated. In vivo, the immunocompetent, syngeneic EMT6 murine model of BrCa was employed to determine therapeutic and tumor-specific immune responses following treatment with RV, anti-PD-1 antibodies or in combination. RV-mediated oncolysis and cytokine production were observed following BrCa cell infection and RV upregulated tumor cell expression of PD-L1. In vivo, RV monotherapy significantly reduced disease burden and enhanced survival in treated mice, and was further enhanced by PD-1 blockade. RV therapy increased the number of intratumoral regulatory T cells, which was reversed by the addition of PD-1 blockade. Finally, dual treatment led to the generation of a systemic adaptive anti-tumor immune response evidenced by an increase in tumor-specific IFN-γ producing CD8+ T cells, and immunity from tumor re-challenge. The combination of PD-1 blockade and RV appears to be an efficacious immunotherapeutic strategy for the treatment of BrCa, and warrants further investigation in early-phase clinical trials.


Cancer immunology research | 2015

Abstract A12: PDL-1 blockade and Sunitinib enhance the efficiency of oncolytic viral therapy

Ahmed E. Mostafa; Kathy Gratton; Keith A. Lawson; Zhong-Qiao Shi; Chandini M. Thirukkumaran; Don Morris

Purpose: Oncolytic viruses (OV) are attractive for use against multiple tumours given their ability to prime therapeutic immune responses in addition to their direct cytotoxic effects. We investigated the utility of reovirus, an OV currently in phase III clinical trials, as a novel immunotherapeutic against renal cell (RCC), non small cell lung (NSCLC) and breast carcinomas (BrCa) as both a monotherapy and in combination with sunitinib, a first line metastatic RCC agent. In addition the utility of adding an immune checkpoint inhibitor was investigated. Experimental Design: In-vitro, cytotoxicity, viral replication and chemokine production were assessed in a panel of human and murine cancer cell lines following exposure to reovirus, sunitinib or their combination. In-vivo, RENCA (murine renal cell carcinoma) cells were implanted subcutaneously into female balb/c mice to establish a syngeneic immunocompetent model of RCC. Tumor growth and overall survival were assessed following treatment with reovirus, sunitinib or their combination. IFN-γ, myeloid-derived suppressor cells (MDSC), and protective immunity were assessed by ELISA, flow cytometry (Gr1+/CD11b+) and adoptive transfer experiments, respectively. PDL-1 and PD-1 expression on both tumour and immune cells were assayed. Results: In vitro, exposure of RCC cell lines to reovirus resulted in an inflammatory cytotoxic response that was augmented through combination with sunitinib. In vivo, reovirus significantly reduced RENCA tumor burden and generated an anti-tumor immune response that was augmented by sunitinib. Combinations with sunitinib also lead to a decrease in myeloid-derived suppressor cells (MDSC) and resulted in improved overall survival and enhanced protective immunity. Preliminary results in NSCLC and BrCa are suggestive that this strategy maybe applicable to many cancer histologies. Both PDL-1 and PD-1 expression were up regulated by RV (similar to IFN gamma) in the majority of cell lines tested suggesting a potential role for checkpoint blockade (CB). Conclusions: Reovirus has both direct oncolytic and immunotherapeutic activity against RCC, which is augmented through combination with sunitinib. The role of immune CB is ongoing. These proof-of-principle investigations provide clear rationale to investigate RV/sunitinib/immune CB as a promising approach for early phase clinical trials as a novel treatment strategy for cancer. Citation Format: Ahmed Mostafa, Kathy Gratton, Keith A. Lawson, Zhong-Qiao Shi, Chandini Thirukkumaran, Don G. Morris. PDL-1 blockade and Sunitinib enhance the efficiency of oncolytic viral therapy. [abstract]. In: Proceedings of the AACR Special Conference: Tumor Immunology and Immunotherapy: A New Chapter; December 1-4, 2014; Orlando, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2015;3(10 Suppl):Abstract nr A12.


Blood | 2011

Bortezomib induced BRCAness sensitizes multiple myeloma cells to PARP inhibitors

Paola Neri; Li Ren; Kathy Gratton; Erin Stebner; Jordan Johnson; Alexander C. Klimowicz; Peter Duggan; Pierfrancesco Tassone; Adnan Mansoor; Douglas A. Stewart; Sagar Lonial; Lawrence H. Boise; Nizar J. Bahlis


Clinical Cancer Research | 2016

Repurposing Sunitinib with Oncolytic Reovirus as a Novel Immunotherapeutic Strategy for Renal Cell Carcinoma

Keith A. Lawson; Ahmed A. Mostafa; Zhong Qiao Shi; Jason C. L. Spurrell; Wenqian Chen; Jun Kawakami; Kathy Gratton; Satbir Thakur; Donald G. Morris


Blood | 2012

High Cereblon Protein Expression Correlates with Improved Response and Survival in Myeloma Patients Treated with Lenalidomide

Alexander C. Klimowicz; Paola Neri; Andrew R. Belch; Michelle Dean; Li Ren; Kathy Gratton; Jiri Slaby; Jordan Johnson; Peter Duggan; Douglas A. Stewart; Nizar J. Bahlis


Blood | 2011

A miRNA Risk Score for the Prediction of Response to Lenalidomide in Multiple Myeloma (MM) Patients

Paola Neri; Andrew R. Belch; Jordan Johnson; Kathy Gratton; Li Ren; Peter Duggan; Adnan Mansoor; Douglas A. Stewart; Nizar J. Bahlis


Blood | 2011

Mir-34a Sensitizes Multiple Myeloma (MM) Cells to the Proteasome Inhibitor Bortezomib

Erin Stebner; Paola Neri; Jordan Johnson; Kathy Gratton; Li Ren; Peter Duggan; Douglas A. Stewart; Nizar J. Bahlis

Collaboration


Dive into the Kathy Gratton's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li Ren

University of Calgary

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge