277 results on '"Robert W. Wilkinson"'
Search Results
2. Small gene networks delineate immune cell states and characterize immunotherapy response in melanoma
- Author
-
Donagh Egan, Martina Kreileder, Myriam Nabhan, Luis F. Iglesias-Martinez, Simon J. Dovedi, Viia Valge-Archer, Amit Grover, Robert W. Wilkinson, Timothy Slidel, Claus Bendtsen, Ian P. Barrett, Donal J. Brennan, Walter Kolch, and Vadim Zhernovkov
- Subjects
Cancer Research ,Immunology - Abstract
Single-cell technologies have elucidated mechanisms responsible for immune checkpoint inhibitor (ICI) response, but are not amenable to a clinical diagnostic setting. In contrast, bulk RNA sequencing (RNA-seq) is now routine for research and clinical applications. Our workflow uses transcription factor (TF)-directed co-expression networks (regulons) inferred from single-cell RNA-seq data to deconvolute immune functional states from bulk RNA-seq data. Regulons preserve the phenotypic variation in CD45+ immune cells from metastatic melanoma samples (n=19, discovery dataset) treated with ICIs, despite reducing dimensionality by > 100-fold. Four cell states, termed exhausted T cells, monocyte lineage cells, memory T cells, and B cells were associated with therapy response; and were characterized by differentially active and cell-state specific regulons. Clustering of bulk RNA-seq melanoma samples from four independent studies (n=209, validation dataset) according to regulon-inferred scores identified four groups with significantly different response outcomes (p < 0.001). An intercellular link was established between exhausted T cells and monocyte lineage cells, whereby their cell numbers were correlated, and exhausted T cells predicted prognosis as a function of monocyte lineage cell number. The ligand–receptor expression analysis suggested that monocyte lineage cells drive exhausted T cells into terminal exhaustion through programs that regulate antigen presentation, chronic inflammation, and negative co-stimulation. Together, our results demonstrate how regulon-based characterization of cell states provide robust and functionally informative markers that can deconvolve bulk RNA-seq data to identify ICI responders.
- Published
- 2023
- Full Text
- View/download PDF
3. MEDI5752FINALSupplementaryMaterial.docx from Design and Efficacy of a Monovalent Bispecific PD-1/CTLA4 Antibody That Enhances CTLA4 Blockade on PD-1+ Activated T Cells
- Author
-
Yariv Mazor, Robert W. Wilkinson, Daniel J. Freeman, Ikbel Achour, William Dall'Acqua, Aleksandra D. Toloczko, Thomas V. Murray, Frances Neal, Gareth J. Browne, Godfrey J. Rainey, Michelle Morrow, Asis Palazon, Yanli Wu, James Dodgson, Michael G. Overstreet, Yaya Wang, Kathy Mulgrew, Stacy Kentner, Xiaofang Jin, Arthur Lewis, Kapil Vashisht, Shelby D. Gainer, Deepa S. Subramaniam, Ben Tran, Seock-Ah Im, Bo Wang, Sumati Hasani, Des C. Jones, James Hair, Anna Hansen, Lorraine Irving, Suzanne I. Sitnikova, Chunning Yang, Matthew J. Elder, and Simon J. Dovedi
- Abstract
Supplementary Materials and Methods, and Supplementary Figures
- Published
- 2023
- Full Text
- View/download PDF
4. Data from Design and Efficacy of a Monovalent Bispecific PD-1/CTLA4 Antibody That Enhances CTLA4 Blockade on PD-1+ Activated T Cells
- Author
-
Yariv Mazor, Robert W. Wilkinson, Daniel J. Freeman, Ikbel Achour, William Dall'Acqua, Aleksandra D. Toloczko, Thomas V. Murray, Frances Neal, Gareth J. Browne, Godfrey J. Rainey, Michelle Morrow, Asis Palazon, Yanli Wu, James Dodgson, Michael G. Overstreet, Yaya Wang, Kathy Mulgrew, Stacy Kentner, Xiaofang Jin, Arthur Lewis, Kapil Vashisht, Shelby D. Gainer, Deepa S. Subramaniam, Ben Tran, Seock-Ah Im, Bo Wang, Sumati Hasani, Des C. Jones, James Hair, Anna Hansen, Lorraine Irving, Suzanne I. Sitnikova, Chunning Yang, Matthew J. Elder, and Simon J. Dovedi
- Abstract
The clinical benefit of PD-1 blockade can be improved by combination with CTLA4 inhibition but is commensurate with significant immune-related adverse events suboptimally limiting the doses of anti-CTLA4 mAb that can be used. MEDI5752 is a monovalent bispecific antibody designed to suppress the PD-1 pathway and provide modulated CTLA4 inhibition favoring enhanced blockade on PD-1+ activated T cells. We show that MEDI5752 preferentially saturates CTLA4 on PD-1+ T cells versus PD-1− T cells, reducing the dose required to elicit IL2 secretion. Unlike conventional PD-1/CTLA4 mAbs, MEDI5752 leads to the rapid internalization and degradation of PD-1. Moreover, we show that MEDI5752 preferentially localizes and accumulates in tumors providing enhanced activity when compared with a combination of mAbs targeting PD-1 and CTLA4 in vivo. Following treatment with MEDI5752, robust partial responses were observed in two patients with advanced solid tumors. MEDI5752 represents a novel immunotherapy engineered to preferentially inhibit CTLA4 on PD-1+ T cells.Significance:The unique characteristics of MEDI5752 represent a novel immunotherapy engineered to direct CTLA4 inhibition to PD-1+ T cells with the potential for differentiated activity when compared with current conventional mAb combination strategies targeting PD-1 and CTLA4. This molecule therefore represents a step forward in the rational design of cancer immunotherapy.See related commentary by Burton and Tawbi, p. 1008.This article is highlighted in the In This Issue feature, p. 995
- Published
- 2023
- Full Text
- View/download PDF
5. Data from Rational Selection of Syngeneic Preclinical Tumor Models for Immunotherapeutic Drug Discovery
- Author
-
Robert W. Wilkinson, Simon J. Dovedi, Ross Stewart, Viia Valge-Archer, Michelle Morrow, James A. Harper, Hazel Jones, Matthew McCourt, Brandon W. Higgs, Philip Brohawn, Jane Coates Ulrichsen, Amanda Watkins, Judith Anderton, Danielle Marcus, Luciano Pacelli, Stefanie Mullins, Rebecca Leyland, Miika J. Ahdesmaki, Danielle M. Greenawalt, Dennis Y.Q. Wang, Jens-Oliver Koopmann, Richard C.A. Sainson, John E. Prime, and Suzanne I.S. Mosely
- Abstract
Murine syngeneic tumor models are critical to novel immuno-based therapy development, but the molecular and immunologic features of these models are still not clearly defined. The translational relevance of differences between the models is not fully understood, impeding appropriate preclinical model selection for target validation, and ultimately hindering drug development. Across a panel of commonly used murine syngeneic tumor models, we showed variable responsiveness to immunotherapies. We used array comparative genomic hybridization, whole-exome sequencing, exon microarray analysis, and flow cytometry to extensively characterize these models, which revealed striking differences that may underlie these contrasting response profiles. We identified strong differential gene expression in immune-related pathways and changes in immune cell–specific genes that suggested differences in tumor immune infiltrates between models. Further investigation using flow cytometry showed differences in both the composition and magnitude of the tumor immune infiltrates, identifying models that harbor “inflamed” and “non-inflamed” tumor immune infiltrate phenotypes. We also found that immunosuppressive cell types predominated in syngeneic mouse tumor models that did not respond to immune-checkpoint blockade, whereas cytotoxic effector immune cells were enriched in responsive models. A cytotoxic cell–rich tumor immune infiltrate has been correlated with increased efficacy of immunotherapies in the clinic, and these differences could underlie the varying response profiles to immunotherapy between the syngeneic models. This characterization highlighted the importance of extensive profiling and will enable investigators to select appropriate models to interrogate the activity of immunotherapies as well as combinations with targeted therapies in vivo. Cancer Immunol Res; 5(1); 29–41. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figures 1 through 5, Supplementary Tables 1 through 4, and Supplementary Methods from Rational Selection of Syngeneic Preclinical Tumor Models for Immunotherapeutic Drug Discovery
- Author
-
Robert W. Wilkinson, Simon J. Dovedi, Ross Stewart, Viia Valge-Archer, Michelle Morrow, James A. Harper, Hazel Jones, Matthew McCourt, Brandon W. Higgs, Philip Brohawn, Jane Coates Ulrichsen, Amanda Watkins, Judith Anderton, Danielle Marcus, Luciano Pacelli, Stefanie Mullins, Rebecca Leyland, Miika J. Ahdesmaki, Danielle M. Greenawalt, Dennis Y.Q. Wang, Jens-Oliver Koopmann, Richard C.A. Sainson, John E. Prime, and Suzanne I.S. Mosely
- Abstract
Supplementary Figure S1: Summary of experiments Supplementary Figure S2: Gating strategy Supplementary Figure S3:Profiling by array CGH, whole-exome and targeted sequencing Supplementary Figure S4: Comparison of mutational profiles of murine syngeneic tumor cell lines and TCGA patient tumors Supplementary Figure S5: Differentially-expressed gene-sets in lymph node and spleen Supplementary Table S1: Cell line details Supplementary Table S2: Copy Number Variation Supplementary Table S3: List of 64 genes investigated by targeted sequencing Supplementary Table S4: Fluorescent antibodies used Supplementary Methods: Linear mixed-effect model
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Dataset from Rational Selection of Syngeneic Preclinical Tumor Models for Immunotherapeutic Drug Discovery
- Author
-
Robert W. Wilkinson, Simon J. Dovedi, Ross Stewart, Viia Valge-Archer, Michelle Morrow, James A. Harper, Hazel Jones, Matthew McCourt, Brandon W. Higgs, Philip Brohawn, Jane Coates Ulrichsen, Amanda Watkins, Judith Anderton, Danielle Marcus, Luciano Pacelli, Stefanie Mullins, Rebecca Leyland, Miika J. Ahdesmaki, Danielle M. Greenawalt, Dennis Y.Q. Wang, Jens-Oliver Koopmann, Richard C.A. Sainson, John E. Prime, and Suzanne I.S. Mosely
- Abstract
Supplementary dataset containing raw data from targeted sequencing, whole exome sequencing, array CGH and transcriptomic analysis
- Published
- 2023
- Full Text
- View/download PDF
8. Data from The Mitogen-Activated Protein/Extracellular Signal-Regulated Kinase Kinase 1/2 Inhibitor AZD6244 (ARRY-142886) Enhances the Radiation Responsiveness of Lung and Colorectal Tumor Xenografts
- Author
-
Stephen R. Wedge, Kaye J. Williams, Ian J. Stratford, Camille Debray, Robert W. Wilkinson, Armelle Logie, Muhammed Babur, Paul D. Smith, Brian A. Telfer, and Aoife M. Shannon
- Abstract
Purpose: Novel molecularly targeted agents, given in combination with radiotherapy, have the potential to increase tumor response rates and the survival of patients with lung cancer. AZD6244 is a potent and selective inhibitor of mitogen-activated protein kinase (MAPK) kinase 1/2 (MEK1/2), a critical enzyme within the MAPK/extracellular signal-regulated kinase (ERK) signaling pathway that regulates the proliferation and survival of tumor cells.Experimental Design: This study examined the potential benefit of combining AZD6244 with fractionated radiotherapy using human lung and colon carcinoma xenograft models.Results: AZD6244 reduced ERK phosphorylation in Calu-6 lung cancer cells in vitro. Administration of AZD6244 for 10 days (25 mg/kg twice daily p.o.) inhibited the tumor growth of Calu-6 xenografts, with regrowth occurring on cessation of drug treatment. When fractionated tumor-localized radiotherapy (5 × 2 Gy) was combined with AZD6244 treatment, the tumor growth delay was enhanced significantly when compared with either modality alone, and this effect was also seen in a colon tumor model. We examined the effect of inhibiting MEK1/2 on the molecular responses to hypoxia, a potential interaction that could contribute to radioresponsiveness. AZD6244 reduced hypoxia-inducible factor–specific transactivation in vivo, shown using Calu-6 dual clone cells that stably express a Firefly luciferase gene under the control of a hypoxia-driven promoter. Furthermore, hypoxia-inducible factor-1α, GLUT-1, and vascular endothelial growth factor levels were reduced by AZD6244, and there was a significant decrease in vascular perfusion in the tumors given combination treatment when compared with the other treatment groups.Conclusions: These data provide support for the clinical development of AZD6244 in combination with radiotherapy and indicate a potential role for AZD6244 in inhibiting the tumor hypoxia response. (Clin Cancer Res 2009;15(21):6619–29)
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental Figure 4 from Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Timothy M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Harlan S. Robins, Ryan O. Emerson, Marissa Vignali, Catherine M. Sanders, Erik C. Yusko, Ross Stewart, Michelle Morrow, Edmund Poon, Amy L. Popple, Eleanor J. Cheadle, and Simon J. Dovedi
- Abstract
Concurrent but not sequential blockade of PD-1 is required to augment the efficacy of fractionated RT
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S1-S11 from Intratumoral IL12 mRNA Therapy Promotes TH1 Transformation of the Tumor Microenvironment
- Author
-
Nadia Luheshi, Joshua P. Frederick, Ronald Herbst, Robert W. Wilkinson, Maja Sedic, Chris Morehouse, Han Si, Jean-Martin Lapointe, Philip Martin, Chris Bagnall, Michal Sulikowski, Steve Novick, David A. Leinster, Gordon Moody, Darren Potz, Katharina Deschler, Grace Adjei, Kathy Mulgrew, Eleanor Clancy-Thompson, James Moynihan, Kristen Arnold, Amanda Watkins, Sushma Gurumurthy, Ankita Mishra, Fabien Garcon, Shannon Burke, Russell Karp, Faith Musenge, Ameya Apte, John Zielinski, Dyane Bailey, and Susannah L. Hewitt
- Abstract
Supplementary Figures S1-S11
- Published
- 2023
- Full Text
- View/download PDF
11. Video - from Optoacoustic Detection of Early Therapy-Induced Tumor Cell Death Using a Targeted Imaging Agent
- Author
-
Kevin M. Brindle, Robert W. Wilkinson, Sarah E. Bohndiek, Richard C.A. Sainson, David Tice, Stefanie R. Mullins, Sarah McGuire, De-En Hu, Susana Ros, André A. Neves, Michal R. Tomaszewski, and Bangwen Xie
- Abstract
Supplementary videos. Representative reconstructed 3D rendering of a Colo205 tumor 3 h after injection of 0.2 µmole/kg of C2Am-750. Signals from C2Am-750, oxygenated hemoglobin (HbO2), deoxygenated hemoglobin (Hb) are shown in green, red and blue respectively.
- Published
- 2023
- Full Text
- View/download PDF
12. Supporting information tracked changes from A Novel Murine GITR Ligand Fusion Protein Induces Antitumor Activity as a Monotherapy That Is Further Enhanced in Combination with an OX40 Agonist
- Author
-
Ross Stewart, Robert W. Wilkinson, Ching Ching Leow, Athula Herath, Philip Mallinder, Scott A. Hammond, Michelle Morrow, Lesley Young, Kelly McGlinchey, David A. Leinster, Jane Coates Ulrichsen, Michael D. Oberst, Stefanie Mullins, Li Yan, John Andrews, Emily Offer, Natalie J. Tigue, Lisa Bamber, Nicholas Holoweckyj, Kathy A. Mulgrew, Amanda Watkins, and Rebecca Leyland
- Abstract
Additional materials and methods and supplementary figure legends with tracked changes
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figures from Optoacoustic Detection of Early Therapy-Induced Tumor Cell Death Using a Targeted Imaging Agent
- Author
-
Kevin M. Brindle, Robert W. Wilkinson, Sarah E. Bohndiek, Richard C.A. Sainson, David Tice, Stefanie R. Mullins, Sarah McGuire, De-En Hu, Susana Ros, André A. Neves, Michal R. Tomaszewski, and Bangwen Xie
- Abstract
Supplementary figures 1 - 4. Caption for video file.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplemental Figure Legends from Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Timothy M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Harlan S. Robins, Ryan O. Emerson, Marissa Vignali, Catherine M. Sanders, Erik C. Yusko, Ross Stewart, Michelle Morrow, Edmund Poon, Amy L. Popple, Eleanor J. Cheadle, and Simon J. Dovedi
- Abstract
Figure legends for Supplemental Figures 1-6
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Optoacoustic Detection of Early Therapy-Induced Tumor Cell Death Using a Targeted Imaging Agent
- Author
-
Kevin M. Brindle, Robert W. Wilkinson, Sarah E. Bohndiek, Richard C.A. Sainson, David Tice, Stefanie R. Mullins, Sarah McGuire, De-En Hu, Susana Ros, André A. Neves, Michal R. Tomaszewski, and Bangwen Xie
- Abstract
Purpose: The development of new treatments and their deployment in the clinic may be assisted by imaging methods that allow an early assessment of treatment response in individual patients. The C2A domain of Synaptotagmin-I (C2Am), which binds to the phosphatidylserine (PS) exposed by apoptotic and necrotic cells, has been developed as an imaging probe for detecting cell death. Multispectral optoacoustic tomography (MSOT) is a real-time and clinically applicable imaging modality that was used here with a near infrared (NIR) fluorophore-labeled C2Am to image tumor cell death in mice treated with a TNF-related apoptosis-inducing ligand receptor 2 (TRAILR2) agonist and with 5-fluorouracil (5-FU).Experimental Design: C2Am was labeled with a NIR fluorophore and injected intravenously into mice bearing human colorectal TRAIL-sensitive Colo205 and TRAIL-resistant HT-29 xenografts that had been treated with a potent agonist of TRAILR2 and in Colo205 tumors treated with 5-FU.Results: Three-dimensional (3D) MSOT images of probe distribution showed development of tumor contrast within 3 hours of probe administration and a signal-to-background ratio in regions containing dead cells of >10 after 24 hours. A site-directed mutant of C2Am that is inactive in PS binding showed negligible binding. Tumor retention of the active probe was strongly correlated (R2 = 0.97, P value < 0.01) with a marker of apoptotic cell death measured in histologic sections obtained post mortem.Conclusions: The rapid development of relatively high levels of contrast suggests that NIR fluorophore-labeled C2Am could be a useful optoacoustic imaging probe for detecting early therapy-induced tumor cell death in the clinic. Clin Cancer Res; 23(22); 6893–903. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplemental Figure 6 from Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Timothy M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Harlan S. Robins, Ryan O. Emerson, Marissa Vignali, Catherine M. Sanders, Erik C. Yusko, Ross Stewart, Michelle Morrow, Edmund Poon, Amy L. Popple, Eleanor J. Cheadle, and Simon J. Dovedi
- Abstract
Treatment with FTY-720 has no direct effect on tumor growth and leads to deletion of non-LN resident and circulating T cells
- Published
- 2023
- Full Text
- View/download PDF
17. Supplemental Figure 2 from Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Timothy M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Harlan S. Robins, Ryan O. Emerson, Marissa Vignali, Catherine M. Sanders, Erik C. Yusko, Ross Stewart, Michelle Morrow, Edmund Poon, Amy L. Popple, Eleanor J. Cheadle, and Simon J. Dovedi
- Abstract
Out-of-field tumor regression is a rare event following local single dose or fractionated RT.
- Published
- 2023
- Full Text
- View/download PDF
18. Data from Intratumoral IL12 mRNA Therapy Promotes TH1 Transformation of the Tumor Microenvironment
- Author
-
Nadia Luheshi, Joshua P. Frederick, Ronald Herbst, Robert W. Wilkinson, Maja Sedic, Chris Morehouse, Han Si, Jean-Martin Lapointe, Philip Martin, Chris Bagnall, Michal Sulikowski, Steve Novick, David A. Leinster, Gordon Moody, Darren Potz, Katharina Deschler, Grace Adjei, Kathy Mulgrew, Eleanor Clancy-Thompson, James Moynihan, Kristen Arnold, Amanda Watkins, Sushma Gurumurthy, Ankita Mishra, Fabien Garcon, Shannon Burke, Russell Karp, Faith Musenge, Ameya Apte, John Zielinski, Dyane Bailey, and Susannah L. Hewitt
- Abstract
Purpose:While immune checkpoint inhibitors such as anti–PD-L1 are rapidly becoming the standard of care in the treatment of many cancers, only a subset of treated patients have long-term responses. IL12 promotes antitumor immunity in mouse models; however, systemic recombinant IL12 had significant toxicity and limited efficacy in early clinical trials.Experimental Design:We therefore designed a novel intratumoral IL12 mRNA therapy to promote local IL12 tumor production while mitigating systemic effects.Results:A single intratumoral dose of mouse (m)IL12 mRNA induced IFNγ and CD8+ T-cell–dependent tumor regression in multiple syngeneic mouse models, and animals with a complete response demonstrated immunity to rechallenge. Antitumor activity of mIL12 mRNA did not require NK and NKT cells. mIL12 mRNA antitumor activity correlated with TH1 tumor microenvironment (TME) transformation. In a PD-L1 blockade monotherapy-resistant model, antitumor immunity induced by mIL12 mRNA was enhanced by anti–PD-L1. mIL12 mRNA also drove regression of uninjected distal lesions, and anti–PD-L1 potentiated this response. Importantly, intratumoral delivery of mRNA encoding membrane-tethered mIL12 also drove rejection of uninjected lesions with very limited circulating IL12p70, supporting the hypothesis that local IL12 could induce a systemic antitumor immune response against distal lesions. Furthermore, in ex vivo patient tumor slice cultures, human IL12 mRNA (MEDI1191) induced dose-dependent IL12 production, downstream IFNγ expression and TH1 gene expression.Conclusions:These data demonstrate the potential for intratumorally delivered IL12 mRNA to promote TH1 TME transformation and robust antitumor immunity.See related commentary by Cirella et al., p. 6080
- Published
- 2023
- Full Text
- View/download PDF
19. Data from Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Timothy M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Harlan S. Robins, Ryan O. Emerson, Marissa Vignali, Catherine M. Sanders, Erik C. Yusko, Ross Stewart, Michelle Morrow, Edmund Poon, Amy L. Popple, Eleanor J. Cheadle, and Simon J. Dovedi
- Abstract
Purpose: Radiotherapy is a highly effective anticancer treatment forming part of the standard of care for the majority of patients, but local and distal disease recurrence remains a major cause of mortality. Radiotherapy is known to enhance tumor immunogenicity; however, the contribution and mechanisms of radiotherapy-induced immune responses are unknown.Experimental Design: The impact of low-dose fractionated radiotherapy (5 × 2 Gy) alone and in combination with αPD-1 mAb on the tumor microenvironment was evaluated by flow cytometry and next-generation sequencing of the T-cell receptor (TCR) repertoire. A dual-tumor model was used, with fractionated radiotherapy delivered to a single tumor site to enable evaluation of the local and systemic response to treatment and ability to induce abscopal responses outside the radiation field.Results: We show that fractionated radiotherapy leads to T-cell infiltration at the irradiated site; however, the TCR landscape remains dominated by polyclonal expansion of preexisting T-cell clones. Adaptive resistance via the PD-1/PD-L1 pathway restricts the generation of systemic anticancer immunity following radiotherapy, which can be overcome through combination with αPD-1 mAb leading to improved local and distal tumor control. Moreover, we show that effective clearance of tumor following combination therapy is dependent on both T cells resident in the tumor at the time of radiotherapy and infiltrating T cells.Conclusions: These data provide evidence that radiotherapy can enhance T-cell trafficking to locally treated tumor sites and augment preexisting anticancer T-cell responses with the capacity to mediate regression of out-of-field tumor lesions when delivered in combination with αPD-1 mAb therapy. Clin Cancer Res; 23(18); 5514–26. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary figures from A Novel Murine GITR Ligand Fusion Protein Induces Antitumor Activity as a Monotherapy That Is Further Enhanced in Combination with an OX40 Agonist
- Author
-
Ross Stewart, Robert W. Wilkinson, Ching Ching Leow, Athula Herath, Philip Mallinder, Scott A. Hammond, Michelle Morrow, Lesley Young, Kelly McGlinchey, David A. Leinster, Jane Coates Ulrichsen, Michael D. Oberst, Stefanie Mullins, Li Yan, John Andrews, Emily Offer, Natalie J. Tigue, Lisa Bamber, Nicholas Holoweckyj, Kathy A. Mulgrew, Amanda Watkins, and Rebecca Leyland
- Abstract
Supplementary figures S1-4
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Data from AZD8931, an Equipotent, Reversible Inhibitor of Signaling by Epidermal Growth Factor Receptor, ERBB2 (HER2), and ERBB3: A Unique Agent for Simultaneous ERBB Receptor Blockade in Cancer
- Author
-
Donald Ogilvie, Robert W. Wilkinson, Bernard Barlaam, Paul Smith, Konstantina Grosios, Elizabeth Mills, Rowena Callis, Sara Davenport, Gayle Marshall, Sarah Beck, Judith Anderton, Cath Trigwell, John Vincent, Georgina Speake, Teresa Klinowska, and D. Mark Hickinson
- Abstract
Supplementary Data from AZD8931, an Equipotent, Reversible Inhibitor of Signaling by Epidermal Growth Factor Receptor, ERBB2 (HER2), and ERBB3: A Unique Agent for Simultaneous ERBB Receptor Blockade in Cancer
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Data from Intratumoral IL12 mRNA Therapy Promotes TH1 Transformation of the Tumor Microenvironment
- Author
-
Nadia Luheshi, Joshua P. Frederick, Ronald Herbst, Robert W. Wilkinson, Maja Sedic, Chris Morehouse, Han Si, Jean-Martin Lapointe, Philip Martin, Chris Bagnall, Michal Sulikowski, Steve Novick, David A. Leinster, Gordon Moody, Darren Potz, Katharina Deschler, Grace Adjei, Kathy Mulgrew, Eleanor Clancy-Thompson, James Moynihan, Kristen Arnold, Amanda Watkins, Sushma Gurumurthy, Ankita Mishra, Fabien Garcon, Shannon Burke, Russell Karp, Faith Musenge, Ameya Apte, John Zielinski, Dyane Bailey, and Susannah L. Hewitt
- Abstract
Supplementary Methods
- Published
- 2023
- Full Text
- View/download PDF
23. Data from AZD5438, a potent oral inhibitor of cyclin-dependent kinases 1, 2, and 9, leads to pharmacodynamic changes and potent antitumor effects in human tumor xenografts
- Author
-
Robert W. Wilkinson, Andy Barker, Jim Growcott, Stephen Green, Nicholas Newcombe, Mike Walker, Clive Green, Sandra Oakes, Catherine Geh, Alexandra McGregor, Cheryl Forder, Gareth Hughes, Andrew Thomas, and Kate F. Byth
- Abstract
Deregulation of the cell cycle has long been recognized as an essential driver of tumorigenesis, and agents that selectively target key cell cycle components continue to hold promise as potential therapeutics. We have developed AZD5438, a 4-(1-isopropyl-2-methylimidazol-5-yl)-2-(4-methylsulphonylanilino) pyrimidine, as a potent inhibitor of cyclin-dependent kinase (cdk) 1, 2, and 9 (IC50, 16, 6, and 20 nmol/L, respectively). In vitro, AZD5438 showed significant antiproliferative activity in human tumor cell lines (IC50 range, 0.2–1.7 μmol/L), causing inhibition of the phosphorylation of cdk substrates pRb, nucleolin, protein phosphatase 1a, and RNA polymerase II COOH-terminal domain and blocking cell cycling at G2-M, S, and G1 phases. In vivo, when orally administered at either 50 mg/kg twice daily or 75 mg/kg once daily, AZD5438 inhibited human tumor xenograft growth (maximum percentage tumor growth inhibition, range, 38–153; P < 0.05). In vivo, AZD5438 reduced the proportion of actively cycling cells. Further pharmacodynamic analysis of AZD5438-treated SW620 xenografts showed that efficacious doses of AZD5438 (>40% tumor growth inhibition) maintained suppression of biomarkers, such as phospho-pRbSer249/Thr252, for up to 16 hours following a single oral dose. A comparison of different schedules indicated that chronic daily oral dosing provided optimal cover to ensure antitumor efficacy. These data indicate that broad cdk inhibition may provide an effective method to impair the dysregulated cell cycle that drives tumorigenesis and AZD5438 has the pharmacologic profile that provides an ideal probe to test this premise. [Mol Cancer Ther 2009;8(7):1856–66]
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Data from The Mitogen-Activated Protein/Extracellular Signal-Regulated Kinase Kinase 1/2 Inhibitor AZD6244 (ARRY-142886) Enhances the Radiation Responsiveness of Lung and Colorectal Tumor Xenografts
- Author
-
Stephen R. Wedge, Kaye J. Williams, Ian J. Stratford, Camille Debray, Robert W. Wilkinson, Armelle Logie, Muhammed Babur, Paul D. Smith, Brian A. Telfer, and Aoife M. Shannon
- Abstract
Supplementary Data from The Mitogen-Activated Protein/Extracellular Signal-Regulated Kinase Kinase 1/2 Inhibitor AZD6244 (ARRY-142886) Enhances the Radiation Responsiveness of Lung and Colorectal Tumor Xenografts
- Published
- 2023
- Full Text
- View/download PDF
25. Supplemental Figure 3 from Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Timothy M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Harlan S. Robins, Ryan O. Emerson, Marissa Vignali, Catherine M. Sanders, Erik C. Yusko, Ross Stewart, Michelle Morrow, Edmund Poon, Amy L. Popple, Eleanor J. Cheadle, and Simon J. Dovedi
- Abstract
The relative frequency of the top 25 clones from tumor 1 tracked in pre-therapy blood, post-therapy blood, and in tumor 2 post-therapy
- Published
- 2023
- Full Text
- View/download PDF
26. Data from A Novel Murine GITR Ligand Fusion Protein Induces Antitumor Activity as a Monotherapy That Is Further Enhanced in Combination with an OX40 Agonist
- Author
-
Ross Stewart, Robert W. Wilkinson, Ching Ching Leow, Athula Herath, Philip Mallinder, Scott A. Hammond, Michelle Morrow, Lesley Young, Kelly McGlinchey, David A. Leinster, Jane Coates Ulrichsen, Michael D. Oberst, Stefanie Mullins, Li Yan, John Andrews, Emily Offer, Natalie J. Tigue, Lisa Bamber, Nicholas Holoweckyj, Kathy A. Mulgrew, Amanda Watkins, and Rebecca Leyland
- Abstract
Purpose: To generate and characterize a murine GITR ligand fusion protein (mGITRL-FP) designed to maximize valency and the potential to agonize the GITR receptor for cancer immunotherapy.Experimental Design: The EC50 value of the mGITRL-FP was compared with an anti-GITR antibody in an in vitro agonistic cell–based reporter assay. We assessed the impact of dose, schedule, and Fc isotype on antitumor activity and T-cell modulation in the CT26 tumor model. The activity of the mGITRL-FP was compared with an agonistic murine OX40L-FP targeting OX40, in CT26 and B16F10-Luc2 tumor models. Combination of the mGITRL-FP with antibodies targeting PD-L1, PD-1, or CTLA-4 was analyzed in mice bearing CT26 tumors.Results: The mGITRL-FP had an almost 50-fold higher EC50 value compared with an anti-murine GITR antibody. Treatment of CT26 tumor-bearing mice with mGITRL-FP–mediated significant antitumor activity that was dependent on isotype, dose, and duration of exposure. The antitumor activity could be correlated with the increased proliferation of peripheral CD8+ and CD4+ T cells and a significant decrease in the frequency of intratumoral Tregs. The combination of mGITRL-FP with mOX40L-FP or checkpoint inhibitor antagonists enhanced antitumor immunity above that of monotherapy treatment.Conclusions: These results suggest that therapeutically targeting GITR represents a unique approach to cancer immunotherapy and suggests that a multimeric fusion protein may provide increased agonistic potential versus an antibody. In addition, these data provide, for the first time, early proof of concept for the potential combination of GITR targeting agents with OX40 agonists and PD-L1 antagonists. Clin Cancer Res; 23(13); 3416–27. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
27. Supplemental Figure 5 from Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Timothy M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Harlan S. Robins, Ryan O. Emerson, Marissa Vignali, Catherine M. Sanders, Erik C. Yusko, Ross Stewart, Michelle Morrow, Edmund Poon, Amy L. Popple, Eleanor J. Cheadle, and Simon J. Dovedi
- Abstract
Representative staining for PD-L1 on tumor cells (CD45-) and CD11b+Gr1+ cells
- Published
- 2023
- Full Text
- View/download PDF
28. Supplemental Figure 1 from Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Timothy M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Harlan S. Robins, Ryan O. Emerson, Marissa Vignali, Catherine M. Sanders, Erik C. Yusko, Ross Stewart, Michelle Morrow, Edmund Poon, Amy L. Popple, Eleanor J. Cheadle, and Simon J. Dovedi
- Abstract
Gating strategy for tumor cells and tumor infiltrating immune cells
- Published
- 2023
- Full Text
- View/download PDF
29. Supporting information from A Novel Murine GITR Ligand Fusion Protein Induces Antitumor Activity as a Monotherapy That Is Further Enhanced in Combination with an OX40 Agonist
- Author
-
Ross Stewart, Robert W. Wilkinson, Ching Ching Leow, Athula Herath, Philip Mallinder, Scott A. Hammond, Michelle Morrow, Lesley Young, Kelly McGlinchey, David A. Leinster, Jane Coates Ulrichsen, Michael D. Oberst, Stefanie Mullins, Li Yan, John Andrews, Emily Offer, Natalie J. Tigue, Lisa Bamber, Nicholas Holoweckyj, Kathy A. Mulgrew, Amanda Watkins, and Rebecca Leyland
- Abstract
Additional materials and methods and supplementary figure legends.
- Published
- 2023
- Full Text
- View/download PDF
30. 3D reconstruction of optoacoustic images of a MEDI3039-treated Colo205 tumor before injection of C2Am-750 from Optoacoustic Detection of Early Therapy-Induced Tumor Cell Death Using a Targeted Imaging Agent
- Author
-
Kevin M. Brindle, Robert W. Wilkinson, Sarah E. Bohndiek, Richard C.A. Sainson, David Tice, Stefanie R. Mullins, Sarah McGuire, De-En Hu, Susana Ros, André A. Neves, Michal R. Tomaszewski, and Bangwen Xie
- Abstract
Supplementary video. Representative reconstructed 3D rendering of a Colo205 tumor before injection of 0.2 µmole/kg of C2Am-750. Signals from C2Am-750, oxygenated hemoglobin (HbO2), deoxygenated hemoglobin (Hb) are shown in green, red and blue respectively.
- Published
- 2023
- Full Text
- View/download PDF
31. Data from AZD8931, an Equipotent, Reversible Inhibitor of Signaling by Epidermal Growth Factor Receptor, ERBB2 (HER2), and ERBB3: A Unique Agent for Simultaneous ERBB Receptor Blockade in Cancer
- Author
-
Donald Ogilvie, Robert W. Wilkinson, Bernard Barlaam, Paul Smith, Konstantina Grosios, Elizabeth Mills, Rowena Callis, Sara Davenport, Gayle Marshall, Sarah Beck, Judith Anderton, Cath Trigwell, John Vincent, Georgina Speake, Teresa Klinowska, and D. Mark Hickinson
- Abstract
Purpose: To test the hypothesis that simultaneous, equipotent inhibition of epidermal growth factor receptor (EGFR; erbB1), erbB2 (human epidermal growth factor receptor 2), and erbB3 receptor signaling, using the novel small-molecule inhibitor AZD8931, will deliver broad antitumor activity in vitro and in vivo.Experimental Design: A range of assays was used to model erbB family receptor signaling in homodimers and heterodimers, including in vitro evaluation of erbB kinase activity, erbB receptor phosphorylation, proliferation in cells, and in vivo testing in a human tumor xenograft panel, with ex vivo evaluation of erbB phosphorylation and downstream biomarkers. Gefitinib and lapatinib were used to compare the pharmacological profile of AZD8931 with other erbB family inhibitors.Results: In vitro, AZD8931 showed equipotent, reversible inhibition of EGFR (IC50, 4 nmol/L), erbB2 (IC50, 3 nmol/L), and erbB3 (IC50, 4 nmol/L) phosphorylation in cells. In proliferation assays, AZD8931 was significantly more potent than gefitinib or lapatinib in specific squamous cell carcinoma of the head and neck and non–small cell lung carcinoma cell lines. In vivo, AZD8931 inhibited xenograft growth in a range of models while significantly affecting EGFR, erbB2, and erbB3 phosphorylation and downstream signaling pathways, apoptosis, and proliferation.Conclusions: AZD8931 has a unique pharmacologic profile providing equipotent inhibition of EGFR, erbB2, and erbB3 signaling and showing greater antitumor activity than agents with a narrower spectrum of erbB receptor inhibition in specific preclinical models. AZD8931 provides the opportunity to investigate whether simultaneous inhibition of erbB receptor signaling could be of utility in the clinic, particularly in the majority of solid tumors that do not overexpress erbB2. Clin Cancer Res; 16(4); 1159–69
- Published
- 2023
- Full Text
- View/download PDF
32. Supplemental Figures 1 - 7 from Acquired Resistance to Fractionated Radiotherapy Can Be Overcome by Concurrent PD-L1 Blockade
- Author
-
Tim M. Illidge, Jamie Honeychurch, Robert W. Wilkinson, Hazel Jones, Ross Stewart, Michelle Morrow, Edmund Poon, Ian J. Stratford, Eleanor J. Cheadle, Sherrie Jones, Conor McKenna, Grazyna Lipowska-Bhalla, Amy L. Adlard, and Simon J. Dovedi
- Abstract
Supplemental figure 1: Gating strategy for determining tumor cell PD-L1 expression. Supplemental figure 2: Impact of low-dose fractionated RT on PD-L1 expression by CD11b+GR1Hi cells. Supplemental figure 3: Dual blockade of PD-1 and PD-L1 does not further enhance efficacy of RT combination. Supplemental figure 4: Blockade of PD-1 / PD-L1 in combination with local RT is well tolerated in mice. Supplemental figure 5: PD-1 / PD-L1 blockade does not directly sensitise tumor cells to RT. Supplemental figure 6: Scheduling of RT and anti-PD-L1 is critical for anti-tumor activity. Supplemental figure 7: Only acute administration of anti-PD-L1 mAb may be required to achieve efficacy when delivered in combination with RT.
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary Methods from Enhanced Apoptosis and Tumor Growth Suppression Elicited by Combination of MEK (Selumetinib) and mTOR Kinase Inhibitors (AZD8055)
- Author
-
Paul D. Smith, Sylvie M. Guichard, Robert W. Wilkinson, Yi-Long Wu, Jingchuan Zhang, Yi Gu, Christine M. Chresta, Sarah Fenton, Sarah Runswick, Denis Alferez, Barry R. Davies, Armelle Logie, and Sarah V. Holt
- Abstract
PDF file - 78K
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Figures 1-3, Table 1 from AZD1152 Rapidly and Negatively Affects the Growth and Survival of Human Acute Myeloid Leukemia Cells In vitro and In vivo
- Author
-
Dominique Bonnet, Simon Joel, Andrew T. Lister, Jude Fitzgibbon, Jamie Cavenagh, Rajesh Odedra, Claire Crafter, Robert W. Wilkinson, Daniel Pearce, and Adedayo Oke
- Abstract
Supplementary Figures 1-3, Table 1 from AZD1152 Rapidly and Negatively Affects the Growth and Survival of Human Acute Myeloid Leukemia Cells In vitro and In vivo
- Published
- 2023
- Full Text
- View/download PDF
35. Targeting DNA damage response components induces enhanced STING-dependent type-I IFN response in ATM deficient cancer cells and drives dendritic cell activation
- Author
-
Marta, Lopez-Pelaez, Lucy, Young, Mercedes, Vazquez-Chantada, Nadine, Nelson, Steve, Durant, Robert W, Wilkinson, Edmund, Poon, Miguel, Gaspar, Viia, Valge-Archer, Paul, Smith, and Simon J, Dovedi
- Subjects
Sulfonamides ,Indoles ,Morpholines ,Immunology ,Membrane Proteins ,DNA ,Dendritic Cells ,Nucleotidyltransferases ,Exodeoxyribonucleases ,Pyrimidines ,Oncology ,Neoplasms ,Interferon Type I ,Immunology and Allergy ,DNA Damage - Abstract
The concept of exploiting tumor intrinsic deficiencies in DNA damage repair mechanisms by inhibiting compensatory DNA repair pathways is well established. For example, ATM-deficient cells show increased sensitivity to the ATR inhibitor ceralasertib. DNA damage response (DDR)-deficient cells are also more sensitive to DNA damaging agents like the DNA crosslinker pyrrolobenzodiazepine (PBD) SG-3199. However, additional antitumor benefits from targeting the DDR pathways, which could operate through the activation of the innate immune system are less well studied. DNA accumulation in the cytosol acts as an immunogenic danger signal, inducing the expression of type-I interferon (IFN) stimulated genes (ISGs) by the activation of the cGAS-STING pathway. Here, we demonstrate that ATM
- Published
- 2022
- Full Text
- View/download PDF
36. Recombinant Newcastle Disease Virus Immunotherapy Drives Oncolytic Effects and Durable Systemic Antitumor Immunity
- Author
-
Nicholas M. Durham, Ruth Franks, Christel Navarro, Kelly McGlinchey, Hong Jin, Nicola Rath, Robert W. Wilkinson, James Harper, Andrew Leinster, Rebecca Leyland, Xing Cheng, Simon J. Dovedi, Kathy Mulgrew, Lee Brown, Jon Travers, Jens-Oliver Koopmann, Shannon Burke, Danielle Carroll, and Jim Eyles
- Subjects
Cancer Research ,animal diseases ,medicine.medical_treatment ,T cell ,Newcastle disease virus ,Apoptosis ,Biology ,Virus ,Immunomodulation ,Mice ,Immune system ,In vivo ,Tumor Cells, Cultured ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Cell Proliferation ,Oncolytic Virotherapy ,Mice, Inbred BALB C ,Granulocyte-Macrophage Colony-Stimulating Factor ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,Acquired immune system ,Xenograft Model Antitumor Assays ,Oncolytic virus ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Colonic Neoplasms ,Cancer research ,Female ,Ex vivo - Abstract
A recombinant Newcastle Disease Virus (NDV), encoding either a human (NDVhuGM-CSF, MEDI5395) or murine (NDVmuGM-CSF) GM-CSF transgene, combined broad oncolytic activity with the ability to significantly modulate genes related to immune functionality in human tumor cells. Replication in murine tumor lines was significantly diminished relative to human tumor cells. Nonetheless, intratumoral injection of NDVmuGM-CSF conferred antitumor effects in three syngeneic models in vivo; with efficacy further augmented by concomitant treatment with anti–PD-1/PD-L1 or T-cell agonists. Ex vivo immune profiling, including T-cell receptor sequencing, revealed profound immune-contexture changes consistent with priming and potentiation of adaptive immunity and tumor microenvironment (TME) reprogramming toward an immune-permissive state. CRISPR modifications rendered CT26 tumors significantly more permissive to NDV replication, and in this setting, NDVmuGM-CSF confers immune-mediated effects in the noninjected tumor in vivo. Taken together, the data support the thesis that MEDI5395 primes and augments cell-mediated antitumor immunity and has significant utility as a combination partner with other immunomodulatory cancer treatments.
- Published
- 2021
- Full Text
- View/download PDF
37. Design and Efficacy of a Monovalent Bispecific PD-1/CTLA4 Antibody That Enhances CTLA4 Blockade on PD-1+ Activated T Cells
- Author
-
Bo Wang, Frances Neal, Arthur Lewis, Kapil Vashisht, Deepa S. Subramaniam, Des C. Jones, Sumati Hasani, Daniel J. Freeman, Chunning Yang, Lorraine Irving, Michael G. Overstreet, Gareth J. Browne, Suzanne I. Sitnikova, James Hair, Robert W. Wilkinson, Yaya Wang, Ben Tran, Ikbel Achour, James Dodgson, Shelby D. Gainer, Xiaofang Jin, Seock-Ah Im, William F Dall'Acqua, Yariv Mazor, Godfrey Rainey, Asis Palazon, Anna Hansen, Yanli Wu, Matthew J. Elder, Stacy Kentner, Aleksandra D. Toloczko, Michelle Morrow, Murray Thomas Vincent, Simon J. Dovedi, and Kathy Mulgrew
- Subjects
0301 basic medicine ,biology ,medicine.drug_class ,Chemistry ,medicine.medical_treatment ,media_common.quotation_subject ,Rational design ,chemical and pharmacologic phenomena ,Immunotherapy ,Monoclonal antibody ,Blockade ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,In vivo ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Cancer research ,Secretion ,Antibody ,Internalization ,media_common - Abstract
The clinical benefit of PD-1 blockade can be improved by combination with CTLA4 inhibition but is commensurate with significant immune-related adverse events suboptimally limiting the doses of anti-CTLA4 mAb that can be used. MEDI5752 is a monovalent bispecific antibody designed to suppress the PD-1 pathway and provide modulated CTLA4 inhibition favoring enhanced blockade on PD-1+ activated T cells. We show that MEDI5752 preferentially saturates CTLA4 on PD-1+ T cells versus PD-1− T cells, reducing the dose required to elicit IL2 secretion. Unlike conventional PD-1/CTLA4 mAbs, MEDI5752 leads to the rapid internalization and degradation of PD-1. Moreover, we show that MEDI5752 preferentially localizes and accumulates in tumors providing enhanced activity when compared with a combination of mAbs targeting PD-1 and CTLA4 in vivo. Following treatment with MEDI5752, robust partial responses were observed in two patients with advanced solid tumors. MEDI5752 represents a novel immunotherapy engineered to preferentially inhibit CTLA4 on PD-1+ T cells. Significance: The unique characteristics of MEDI5752 represent a novel immunotherapy engineered to direct CTLA4 inhibition to PD-1+ T cells with the potential for differentiated activity when compared with current conventional mAb combination strategies targeting PD-1 and CTLA4. This molecule therefore represents a step forward in the rational design of cancer immunotherapy. See related commentary by Burton and Tawbi, p. 1008. This article is highlighted in the In This Issue feature, p. 995
- Published
- 2021
- Full Text
- View/download PDF
38. Intratumoral IL12 mRNA Therapy Promotes TH1 Transformation of the Tumor Microenvironment
- Author
-
Kristen Arnold, Chris Morehouse, Eleanor Clancy-Thompson, Katharina Deschler, Susannah Hewitt, Russell Karp, Ameya Apte, Chris Bagnall, Faith Musenge, Darren Potz, David A. Leinster, Kathy Mulgrew, Ankita Mishra, Gordon Moody, Maja Sedic, Joshua Frederick, John Zielinski, Fabien Garcon, Sushma Gurumurthy, Nadia Luheshi, Michal Sulikowski, Steve Novick, Han Si, Philip Martin, Grace Adjei, James Moynihan, Jean-Martin Lapointe, Amanda Watkins, Shannon Burke, Ronald Herbst, Dyane Bailey, and Robert W. Wilkinson
- Subjects
0301 basic medicine ,Cancer Research ,Mice, Nude ,Apoptosis ,Mice, SCID ,CD8-Positive T-Lymphocytes ,B7-H1 Antigen ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Immunity ,Gene expression ,Tumor Cells, Cultured ,Tumor Microenvironment ,Animals ,Humans ,Medicine ,RNA, Messenger ,Melanoma ,Cell Proliferation ,Mice, Inbred BALB C ,Tumor microenvironment ,business.industry ,Antibodies, Monoclonal ,Th1 Cells ,Natural killer T cell ,Interleukin-12 ,Xenograft Model Antitumor Assays ,Mice, Inbred C57BL ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Interleukin 12 ,Female ,Colorectal Neoplasms ,business ,Ex vivo ,CD8 - Abstract
Purpose: While immune checkpoint inhibitors such as anti–PD-L1 are rapidly becoming the standard of care in the treatment of many cancers, only a subset of treated patients have long-term responses. IL12 promotes antitumor immunity in mouse models; however, systemic recombinant IL12 had significant toxicity and limited efficacy in early clinical trials. Experimental Design: We therefore designed a novel intratumoral IL12 mRNA therapy to promote local IL12 tumor production while mitigating systemic effects. Results: A single intratumoral dose of mouse (m)IL12 mRNA induced IFNγ and CD8+ T-cell–dependent tumor regression in multiple syngeneic mouse models, and animals with a complete response demonstrated immunity to rechallenge. Antitumor activity of mIL12 mRNA did not require NK and NKT cells. mIL12 mRNA antitumor activity correlated with TH1 tumor microenvironment (TME) transformation. In a PD-L1 blockade monotherapy-resistant model, antitumor immunity induced by mIL12 mRNA was enhanced by anti–PD-L1. mIL12 mRNA also drove regression of uninjected distal lesions, and anti–PD-L1 potentiated this response. Importantly, intratumoral delivery of mRNA encoding membrane-tethered mIL12 also drove rejection of uninjected lesions with very limited circulating IL12p70, supporting the hypothesis that local IL12 could induce a systemic antitumor immune response against distal lesions. Furthermore, in ex vivo patient tumor slice cultures, human IL12 mRNA (MEDI1191) induced dose-dependent IL12 production, downstream IFNγ expression and TH1 gene expression. Conclusions: These data demonstrate the potential for intratumorally delivered IL12 mRNA to promote TH1 TME transformation and robust antitumor immunity. See related commentary by Cirella et al., p. 6080
- Published
- 2020
- Full Text
- View/download PDF
39. Extensive sequence and structural evolution of Arginase 2 inhibitory antibodies enabled by an unbiased approach to affinity maturation
- Author
-
Ellen Gowans, Stuart W. Haynes, Mark D. Carr, Stephanie Ryman, Maria A T Groves, Daniel Burschowsky, Tristan J Vaughan, Alexandra Addyman, Vincenzo Cerundolo, Agata Diamandakis, Chitra Seewooruthun, Yoko Shibata, Louise H. Slater, Sarah V. Holt, Denice T Y Chan, Sebastian Fiedler, Jessica Whitehouse, Robert W. Wilkinson, Lesley Jenkinson, Mark Austin, and Michelle Barnard
- Subjects
inhibitory antibodies ,Antibody Affinity ,Computational biology ,Biochemistry ,Antibodies ,Affinity maturation ,Antigen ,Humans ,ARG2 ,affinity maturation ,Multidisciplinary ,Arginase ,antibody engineering ,biology ,ribosome display ,Chemistry ,Biological Sciences ,Complementarity Determining Regions ,Arginase 2 ,Ribosome display ,biology.protein ,Paratope ,Binding Sites, Antibody ,Antibody ,Systematic evolution of ligands by exponential enrichment - Abstract
Significance We describe an antibody optimization strategy that seeks to overcome the restrictive nature of existing affinity-maturation methods, by rapidly exploring a vast sequence space in an unbiased manner through application of PCR techniques and ribosome display. We exemplified the significance of this method by contrasting the crystal structure of the parent and optimized antibodies bound to Arginase 2, which revealed a striking reorientation of the binding paratope, concurrent with distinct improvements in inhibitory potency and binding properties. The nature and magnitude of the epitope expansion was extraordinary and unlikely to have been produced through conventional affinity-maturation methods. This innovative approach demonstrates broad applicability to the optimization of candidate therapeutic antibodies, even those less amenable to CDRH3 targeting., Affinity maturation is a powerful technique in antibody engineering for the in vitro evolution of antigen binding interactions. Key to the success of this process is the expansion of sequence and combinatorial diversity to increase the structural repertoire from which superior binding variants may be selected. However, conventional strategies are often restrictive and only focus on small regions of the antibody at a time. In this study, we used a method that combined antibody chain shuffling and a staggered-extension process to produce unbiased libraries, which recombined beneficial mutations from all six complementarity-determining regions (CDRs) in the affinity maturation of an inhibitory antibody to Arginase 2 (ARG2). We made use of the vast display capacity of ribosome display to accommodate the sequence space required for the diverse library builds. Further diversity was introduced through pool maturation to optimize seven leads of interest simultaneously. This resulted in antibodies with substantial improvements in binding properties and inhibition potency. The extensive sequence changes resulting from this approach were translated into striking structural changes for parent and affinity-matured antibodies bound to ARG2, with a large reorientation of the binding paratope facilitating increases in contact surface and shape complementarity to the antigen. The considerable gains in therapeutic properties seen from extensive sequence and structural evolution of the parent ARG2 inhibitory antibody clearly illustrate the advantages of the unbiased approach developed, which was key to the identification of high-affinity antibodies with the desired inhibitory potency and specificity.
- Published
- 2020
- Full Text
- View/download PDF
40. Abstract 1133: A B7-H4 targeting antibody-drug conjugate shows anti-tumor activity in PARPi and platinum resistant cancers with B7-H4 expression
- Author
-
Sarah B. Gitto, Margaret Whicker, Gareth Davies, Sushil Kumar, Krista Kinneer, Arthur Lewis, Srinivas Mamidi, Sergey Medvedev, Judith Anderton, Jessica Tang, Benjamin Ferman, Steve Coats, Robert W. Wilkinson, Eric J. Brown, Daniel J. Powell, and Fiona Simpkins
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: Platinum and PARP inhibitors (PARPi) demonstrate activity in breast and ovarian cancers, but drug resistance ultimately emerges. B7-H4 is over expressed in breast and ovarian cancer compared to normal tissues making it a promising target for therapies, however, to date it has not been evaluated if the expression is maintained in drug resistant cancer. Here we examine B7-H4 expression in high grade serous ovarian carcinoma (HGSOC) tumors at diagnosis and post-platinum or PARPi-resistance. We also evaluate the activity of a novel B7-H4-directed antibody-drug conjugate (ADC) bearing the pyrrolobenzodiazepine-dimer payload tesirine, in preclinical models of breast and ovarian cancer, including those resistant to standard of care therapies. Experimental Design: B7-H4 expression was measured by quantitative flow cytometry and immunohistochemistry. ADC efficacy was tested against multiple cell lines in vitro and patient-derived xenografts (PDX) in vivo. The effect of ADC treatment on cell cycle, DNA damage, and apoptosis was measured using flow cytometry. Results: B7-H4 was over-expressed in 92% of HGSOC tumors at diagnosis (n = 12), persisted in recurrent matched samples after platinum treatment, and was expressed at similar levels at multiple metastatic sites after acquired multi-drug resistance (n = 4 donors). Treatment with the ADC resulted in target-specific growth inhibition in a panel of ten B7-H4 expressing ovarian and breast cancer cell lines (IC50 6.18-273.9 pM, median 48.3 pM). Co-cultures of B7-H4 +/- lines support bystander killing effects leading to further tumor cell death in B7-H4- clones. In platinum- or PARPi-resistant ovarian cancer cells, ADC treatment significantly decreased viability and colony formation (P < 0.001) while increasing S or G2/M phase cell cycle arrest (P < 0.001) and DNA damage (P < 0.0001), ultimately leading to apoptosis (P < 0.01). A single 0.3 mg/kg dose of the ADC resulted in tumor regression in 61% of breast and ovarian PDX models tested (n = 23), where lower activity was identified in B7-H4 low or negative expressing models (P = 0.048). In PARPi- and platinum-resistant HGSOC models (n = 3), continuous B7-H4 ADC treatment (dosed once every 28 days) resulted in sustained anti-tumor activity, leading to complete (7 of 16) or partial responses (3 of 16) and increased survival (P < 0.004). Conclusions: These data support B7-H4 as an attractive ADC target for treatment of drug-resistant HGSOC. Citation Format: Sarah B. Gitto, Margaret Whicker, Gareth Davies, Sushil Kumar, Krista Kinneer, Arthur Lewis, Srinivas Mamidi, Sergey Medvedev, Judith Anderton, Jessica Tang, Benjamin Ferman, Steve Coats, Robert W. Wilkinson, Eric J. Brown, Daniel J. Powell, Fiona Simpkins. A B7-H4 targeting antibody-drug conjugate shows anti-tumor activity in PARPi and platinum resistant cancers with B7-H4 expression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1133.
- Published
- 2023
- Full Text
- View/download PDF
41. Deep phenotyping of surface stimulatory and inhibitory co-receptors on cancer-resident T and NK cells reveals cell subsets within the tumor-reactive CTL population that are uniquely defined by NKG2A expression
- Author
-
James Hair, Matthew J Robinson, Robert W Wilkinson, and Simon J Dovedi
- Subjects
Killer Cells, Natural ,Neoplasms ,Molecular Medicine ,Humans ,Flow Cytometry ,Biochemistry ,Analytical Chemistry ,Biotechnology ,T-Lymphocytes, Cytotoxic - Abstract
The field of Immuno-Oncology (IO) is evolving to utilise novel antibody backbones that can co-target multiple cell-surface stimulatory and inhibitory co-receptors (SICR). This approach necessitates a better understanding of SICR co-expression at the single-cell level on IO-relevant tumor-infiltrating leukocyte (TIL) cell types such as T and natural killer (NK) cells. Using high-dimensional flow cytometry we established a comprehensive SICR profile for tumor-resident T and NK cells across a range of human solid tumors where there is a clear need for improved immunotherapeutic intervention. Leveraging the power of our large flow panel, we performed deep-phenotyping of the critical CD8
- Published
- 2022
42. Activity of murine surrogate antibodies for durvalumab and tremelimumab lacking effector function and the ability to deplete regulatory T cells in mouse models of cancer
- Author
-
Kelly McGlinchey, Scott A. Hammond, John Hood, Ross Stewart, Grace Adjei, Weimin Chen, John Andrews, Darren J. Schofield, Robert W. Wilkinson, Maria A T Groves, Leslie Wetzel, Jennifer L. Percival-Alwyn, Michael Oberst, Mateusz Rytelewski, LeeAnn Machiesky, Andrew Leinster, Claire Dobson, Nadia Luheshi, Raymond Rothstein, Michelle Morrow, and Amanda Watkins
- Subjects
PD-L1 ,Durvalumab ,medicine.drug_class ,durvalumab ,medicine.medical_treatment ,Immunology ,Kaplan-Meier Estimate ,Biology ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,T-Lymphocytes, Regulatory ,B7-H1 Antigen ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antineoplastic Agents, Immunological ,tremelimumab ,Cell Line, Tumor ,Report ,medicine ,Immunology and Allergy ,Animals ,Humans ,CTLA-4 Antigen ,murine surrogates ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Mice, Inbred BALB C ,Effector ,Antibodies, Monoclonal ,Immunotherapy ,Neoplasms, Experimental ,Tumor Burden ,Mice, Inbred C57BL ,CTLA-4 ,030220 oncology & carcinogenesis ,Cancer research ,Female ,immunotherapy ,Tremelimumab ,medicine.drug - Abstract
Preclinical studies of PD-L1 and CTLA-4 blockade have relied heavily on mouse syngeneic tumor models with intact immune systems, which facilitate dissection of immunosuppressive mechanisms in the tumor microenvironment. Commercially developed monoclonal antibodies (mAbs) targeting human PD-L1, PD-1, and CTLA-4 may not demonstrate cross-reactive binding to their mouse orthologs, and surrogate anti-mouse antibodies are often used in their place to inhibit these immune checkpoints. In each case, multiple choices exist for surrogate antibodies, which differ with respect to species of origin, affinity, and effector function. To develop relevant murine surrogate antibodies for the anti-human PD-L1 mAb durvalumab and the anti-human CTLA-4 mAb tremelimumab, rat/mouse chimeric or fully murine mAbs engineered for reduced effector function were developed and compared with durvalumab and tremelimumab. Characterization included determination of target affinity, in vivo effector function, pharmacokinetic profile, and anti-tumor efficacy in mouse syngeneic tumor models. Results showed that anti–PD-L1 and anti–CTLA-4 murine surrogates with pharmacologic properties similar to those of durvalumab and tremelimumab demonstrated anti-tumor activity in a subset of commonly used mouse syngeneic tumor models. This activity was not entirely dependent on antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis effector function, or regulatory T-cell depletion, as antibodies engineered to lack these features showed activity in models historically sensitive to checkpoint inhibition, albeit at a significantly lower level than antibodies with intact effector function.
- Published
- 2021
43. Design and Efficacy of a Monovalent Bispecific PD-1/CTLA4 Antibody That Enhances CTLA4 Blockade on PD-1
- Author
-
Simon J, Dovedi, Matthew J, Elder, Chunning, Yang, Suzanne I, Sitnikova, Lorraine, Irving, Anna, Hansen, James, Hair, Des C, Jones, Sumati, Hasani, Bo, Wang, Seock-Ah, Im, Ben, Tran, Deepa S, Subramaniam, Shelby D, Gainer, Kapil, Vashisht, Arthur, Lewis, Xiaofang, Jin, Stacy, Kentner, Kathy, Mulgrew, Yaya, Wang, Michael G, Overstreet, James, Dodgson, Yanli, Wu, Asis, Palazon, Michelle, Morrow, Godfrey J, Rainey, Gareth J, Browne, Frances, Neal, Thomas V, Murray, Aleksandra D, Toloczko, William, Dall'Acqua, Ikbel, Achour, Daniel J, Freeman, Robert W, Wilkinson, and Yariv, Mazor
- Subjects
Male ,Stomach Neoplasms ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,Humans ,CTLA-4 Antigen ,Immunotherapy ,Adenocarcinoma ,Middle Aged ,Antibodies, Monoclonal, Humanized ,Kidney Neoplasms ,Adenocarcinoma, Clear Cell - Abstract
The clinical benefit of PD-1 blockade can be improved by combination with CTLA4 inhibition but is commensurate with significant immune-related adverse events suboptimally limiting the doses of anti-CTLA4 mAb that can be used. MEDI5752 is a monovalent bispecific antibody designed to suppress the PD-1 pathway and provide modulated CTLA4 inhibition favoring enhanced blockade on PD-1
- Published
- 2020
44. P03.13 Age-induced changes in anti-tumor immunity alter the tumor immune infiltrate and reduce response to immune-oncology treatments
- Author
-
Robert W. Wilkinson, Simon J. Dovedi, Matthew J Robinson, Viia Valge-Archer, Michelle Morrow, and Suzanne I. Sitnikova
- Subjects
Oncology ,medicine.medical_specialty ,education.field_of_study ,Naive T cell ,business.industry ,Receptor expression ,Population ,Spleen ,Immune checkpoint ,Blockade ,medicine.anatomical_structure ,Immune system ,Internal medicine ,medicine ,business ,education ,CD8 - Abstract
Background Immuno-Oncology research relies heavily on murine syngeneic tumor models. However, whilst the median age for a cancer diagnosis is 65 years or older, for practical purposes the majority of preclinical studies are conducted in young mice, despite the fact that ageing has been shown to have a significant impact on the immune response. Materials and Methods Using aged mice bearing CT26 tumors, we analysed how aging impacts the immune composition of the tumor, spleen and tumor-draining lymph nodes by flow cytometry. Results We found many age-related changes between aged (60–72 weeks old) and young (6–8 weeks old) mice, such as a reduction in the naive T cell population and a decreased CD8/Treg ratio in aged animals. Profiling of co-inhibitory and co-stimulatory receptor expression levels on immune cells in aged versus young mice also identified altered expression profiles in both the periphery and tumor. We hypothesised that these differences may contribute to impaired anti-cancer immune responses in aged mice. To investigate this, we compared the anti-tumor efficacy of immune checkpoint blockade (PD-L1 and CTLA-4) and T-cell costimulation (OX-40) in aged versus young mice. Our data demonstrate that aged mice retained their capacity to generate effective anti-tumor immune responses, albeit often attenuated when compared to the responses observed in young mice. Conclusions These differences highlight the potential importance of age-related immunological changes in assessing and refining the translational insights gained from preclinical mouse models. Disclosure Information S. Sitnikova: A. Employment (full or part-time); Significant; AstraZeneca. M. Morrow: A. Employment (full or part-time); Significant; AstraZeneca. V. Valge-Archer: A. Employment (full or part-time); Significant; AstraZeneca. R.W. Wilkinson: A. Employment (full or part-time); Significant; AstraZeneca. M.J. Robinson: A. Employment (full or part-time); Significant; AstraZeneca. S.J. Dovedi: A. Employment (full or part-time); Significant; AstraZeneca.
- Published
- 2020
- Full Text
- View/download PDF
45. Fractionated Radiation Therapy Stimulates Antitumor Immunity Mediated by Both Resident and Infiltrating Polyclonal T-cell Populations when Combined with PD-1 Blockade
- Author
-
Simon J. Dovedi, Edmund Poon, Ryan O. Emerson, Marissa Vignali, Erik Yusko, Harlan Robins, Jamie Honeychurch, Robert W. Wilkinson, Ross Stewart, Catherine Sanders, Eleanor J. Cheadle, Amy Popple, Michelle Morrow, and Timothy M Illidge
- Subjects
0301 basic medicine ,Cancer Research ,Antineoplastic Agents, Hormonal ,Combination therapy ,Receptors, Antigen, T-Cell, alpha-beta ,medicine.medical_treatment ,T cell ,Programmed Cell Death 1 Receptor ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Antigen ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Tumor microenvironment ,Radiotherapy ,Manchester Cancer Research Centre ,business.industry ,ResearchInstitutes_Networks_Beacons/mcrc ,Immunogenicity ,T-cell receptor ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Survival Rate ,Radiation therapy ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Cytokines ,business - Abstract
Purpose: Radiotherapy is a highly effective anticancer treatment forming part of the standard of care for the majority of patients, but local and distal disease recurrence remains a major cause of mortality. Radiotherapy is known to enhance tumor immunogenicity; however, the contribution and mechanisms of radiotherapy-induced immune responses are unknown. Experimental Design: The impact of low-dose fractionated radiotherapy (5 × 2 Gy) alone and in combination with αPD-1 mAb on the tumor microenvironment was evaluated by flow cytometry and next-generation sequencing of the T-cell receptor (TCR) repertoire. A dual-tumor model was used, with fractionated radiotherapy delivered to a single tumor site to enable evaluation of the local and systemic response to treatment and ability to induce abscopal responses outside the radiation field. Results: We show that fractionated radiotherapy leads to T-cell infiltration at the irradiated site; however, the TCR landscape remains dominated by polyclonal expansion of preexisting T-cell clones. Adaptive resistance via the PD-1/PD-L1 pathway restricts the generation of systemic anticancer immunity following radiotherapy, which can be overcome through combination with αPD-1 mAb leading to improved local and distal tumor control. Moreover, we show that effective clearance of tumor following combination therapy is dependent on both T cells resident in the tumor at the time of radiotherapy and infiltrating T cells. Conclusions: These data provide evidence that radiotherapy can enhance T-cell trafficking to locally treated tumor sites and augment preexisting anticancer T-cell responses with the capacity to mediate regression of out-of-field tumor lesions when delivered in combination with αPD-1 mAb therapy. Clin Cancer Res; 23(18); 5514–26. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
46. A pharmacokinetic-pharmacodynamic model predicting tumour growth inhibition after intermittent administration with the mTOR kinase inhibitor AZD8055
- Author
-
James W.T. Yates, Karen Woods, Robert W. Wilkinson, Sarah V. Holt, Kirsty Payne, Barry R. Davies, Armelle Logie, and Sylvie Guichard
- Subjects
0301 basic medicine ,Pharmacology ,business.industry ,In vitro ,03 medical and health sciences ,Route of administration ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,MTOR Kinase Inhibitor AZD8055 ,chemistry ,Pharmacokinetics ,In vivo ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Medicine ,Growth inhibition ,business ,PI3K/AKT/mTOR pathway - Abstract
Background AZD8055 is a potent orally available mTOR kinase inhibitor with in vitro and in vivo antitumour activity against a range of tumour types. Preclinical studies showed that AZD8055 induced a dose-dependent pharmacodynamic effect in xenograft models in vivo but a lack of understanding of the relative contributions of the maximum inhibition of the biomarkers and the duration of inhibition to the antitumour effect limited the rational design of experiments to optimize the dose and schedules. Methods In this study, a mathematical modeling approach was developed to relate pharmacodynamics and antitumour activity using preclinical data generated in mice bearing U87-MG xenografts. Results Refinement and validation of the model was carried out in a panel of additional human tumour xenograft models with different growth rates and different sensitivity to AZD8055 (from partial growth inhibition to regression). Finally, the model was applied to accurately predict the efficacy of high, intermittent dosing schedules of AZD8055. Conclusion Overall, this new model linking pharmacokinetics, pharmacodynamic biomarkers and efficacy across multiple tumour xenografts with different sensitivity to AZD8055 was able to identify the optimal dose and route of administration to maximize the antitumour efficacy in pre-clinical models and its potential for translation into man.
- Published
- 2017
- Full Text
- View/download PDF
47. A Novel Murine GITR Ligand Fusion Protein Induces Antitumor Activity as a Monotherapy That Is Further Enhanced in Combination with an OX40 Agonist
- Author
-
Ching Ching Leow, Scott A. Hammond, Michael Oberst, Li Yan, Stefanie R. Mullins, Lesley Young, Michelle Morrow, Amanda Watkins, Lisa Bamber, Nicholas Holoweckyj, Jane Coates Ulrichsen, Robert W. Wilkinson, Kathy Mulgrew, Rebecca Leyland, John Andrews, Emily Offer, Ross Stewart, Natalie J. Tigue, Athula Herath, David A Leinster, Philip R. Mallinder, and Kelly McGlinchey
- Subjects
0301 basic medicine ,Agonist ,Cancer Research ,Oncogene Proteins, Fusion ,medicine.drug_class ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Melanoma, Experimental ,OX40 Ligand ,CD8-Positive T-Lymphocytes ,Biology ,Pharmacology ,B7-H1 Antigen ,Mice ,03 medical and health sciences ,Glucocorticoid-Induced TNFR-Related Protein ,0302 clinical medicine ,Cancer immunotherapy ,medicine ,Animals ,Humans ,CTLA-4 Antigen ,Receptor ,Membrane Glycoproteins ,Fusion protein ,Disease Models, Animal ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Tumor Necrosis Factors ,biology.protein ,Antibody ,CD8 - Abstract
Purpose: To generate and characterize a murine GITR ligand fusion protein (mGITRL-FP) designed to maximize valency and the potential to agonize the GITR receptor for cancer immunotherapy. Experimental Design: The EC50 value of the mGITRL-FP was compared with an anti-GITR antibody in an in vitro agonistic cell–based reporter assay. We assessed the impact of dose, schedule, and Fc isotype on antitumor activity and T-cell modulation in the CT26 tumor model. The activity of the mGITRL-FP was compared with an agonistic murine OX40L-FP targeting OX40, in CT26 and B16F10-Luc2 tumor models. Combination of the mGITRL-FP with antibodies targeting PD-L1, PD-1, or CTLA-4 was analyzed in mice bearing CT26 tumors. Results: The mGITRL-FP had an almost 50-fold higher EC50 value compared with an anti-murine GITR antibody. Treatment of CT26 tumor-bearing mice with mGITRL-FP–mediated significant antitumor activity that was dependent on isotype, dose, and duration of exposure. The antitumor activity could be correlated with the increased proliferation of peripheral CD8+ and CD4+ T cells and a significant decrease in the frequency of intratumoral Tregs. The combination of mGITRL-FP with mOX40L-FP or checkpoint inhibitor antagonists enhanced antitumor immunity above that of monotherapy treatment. Conclusions: These results suggest that therapeutically targeting GITR represents a unique approach to cancer immunotherapy and suggests that a multimeric fusion protein may provide increased agonistic potential versus an antibody. In addition, these data provide, for the first time, early proof of concept for the potential combination of GITR targeting agents with OX40 agonists and PD-L1 antagonists. Clin Cancer Res; 23(13); 3416–27. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
48. Intratumoral administration of the Toll‐like receptor 7/8 agonist 3M‐052 enhances interferon‐driven tumor immunogenicity and suppresses metastatic spread in preclinical triple‐negative breast cancer
- Author
-
Robert W. Wilkinson, Hendrika M. Duivenvoorden, Damien Zanker, Alex J. Spurling, Stefanie R. Mullins, Belinda S. Parker, Tina Robinson, Natasha K Brockwell, Katie L. Owen, Paul J. Hertzog, and Jasmine M. Zakhour
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,medicine.medical_treatment ,Immunology ,Metastasis ,Metastasis Suppression ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,metastasis ,Immunology and Allergy ,Medicine ,TLR agonist ,General Nursing ,Triple-negative breast cancer ,Tumor microenvironment ,Mammary tumor ,business.industry ,Original Articles ,interferon ,Immunotherapy ,medicine.disease ,Primary tumor ,triple‐negative breast cancer ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Original Article ,immunotherapy ,CD8+ T cell ,lcsh:RC581-607 ,business - Abstract
Objectives Loss of tumor‐inherent type I interferon (IFN) signalling has been closely linked to accelerated metastatic progression via decreased immunogenicity and antitumor immunity. Previous studies in murine models of triple‐negative breast cancer (TNBC) demonstrate that systemic IFN inducers are effective antimetastatic agents, via sustained antitumor CD8+ T‐cell responses. Repeated systemic dosing with recombinant IFNs or IFN inducers is associated with significant toxicities; hence, the use of alternate intratumoral agents is an active area of investigation. It is critical to investigate the impact of intratumoral agents on subsequent metastatic spread to predict clinical impact. Methods In this study, the local and systemic impact of the intratumoral Toll‐like receptor (TLR) 7/8 agonist 3M‐052 alone or in combination with anti‐PD1 was evaluated in metastatic TNBC models. The IFN‐α receptor (IFNAR1) blocking antibody, MAR1‐5A3, along with immune‐deficient mice and ex vivo assays are utilised to examine the key targets of this agent that are critical for an antimetastatic response. Results Single intratumoral administration of 3M‐052 reduced mammary tumor growth, induced a T‐cell‐inflamed tumor microenvironment (TME) and reduced metastatic spread to lung. Metastasis suppression was reliant on IFN signalling and an antitumor immune response, in contrast to primary tumor growth inhibition, which was retained in NSG and CD8+ T‐cell‐depleted mice. 3M‐052 action was demonstrated via dendritic cell activation and production of type I IFN and other pro‐inflammatory cytokines to initiate a T‐cell‐inflamed TME and promote tumor cell antigen presentation. Conclusion This work supports neoadjuvant TLR agonist‐based immunotherapeutics as realistic options for immune activation in the TME and long‐term metastatic protection in TNBC., Here, we present preclinical findings on the efficacy of direct tumor adminstration of an interferon inducer in triple‐negative breast cancer. The use of an intratumoral Toll‐like receptor agonist (3M‐052), formulated for retention at the primary tumor site, was sufficient to induce a T‐cell‐inflamed tumor microenvironment in mouse models of triple‐negative breast cancer. A single injection of 3M‐052 into the primary tumor dramatically reduced subsequent metastatic spread to lung, and this was dependent on the antitumor immune response.
- Published
- 2020
- Full Text
- View/download PDF
49. Abstract 1695: Activation of B cells by CD73 blocking antibodies
- Author
-
Zachary A. Cooper, Michelle Hsueh, James Hair, Jim E. Eyles, Fabien Garcon, Simon J. Dovedi, Robert W. Wilkinson, Elena Bibikova, Laura Dallaway, Kris Sachsenmeier, Maria Letizia Giardino Torchia, Rajesh Kumar, Alwin Schuller, and Gordon Moody
- Subjects
Cancer Research ,Oncology ,Chemistry ,Blocking antibody ,Molecular biology - Abstract
The catabolism of ATP into immunosuppressive adenosine contributes to the dysfunction of tumor infiltrating leukocytes (TIL). CD73 is an ectonucleotidase which catabolizes conversion of AMP to adenosine and is expressed on a range of immune cells including B cells. Oleclumab (MEDI9447) is a monoclonal antibody specific for human CD73 and is currently in clinical development for the treatment of cancer. B cells constitute a significant proportion of human TIL; however, their importance to Immuno-Oncology (IO) treatments remains unclear. Recent publications demonstrate that B cells sustain inflammation and predict response to immune checkpoint blockade in human melanoma, their presence within the tumor microenvironment is correlated with improved prognosis in several human tumor types, and CD73 expression has been shown to vary on different human B cell subtypes. Further investigations into the role of CD73 in control of human B cell function are therefore warranted. We show that blockade of CD73 by oleclumab and other monoclonal antibodies on peripheral blood B cells from healthy human donors upregulates expression of CD69, CD83 and CD86, and induces secretion of IL-6, MIP-1α and MIP-1β. Activation is mediated by antibodies that block and internalize CD73 and is reduced by pharmacological inhibition of BTK, thus highlighting involvement of the canonical B-cell receptor signaling pathway. Furthermore, our analyses highlight a differential effect of CD73 blockade on individual B-cell populations, with the most robust increases in activation marker expression being observed on naïve subsets that retain expression of IgD. Our research has identified that oleclumab activates human peripheral blood B cells. Given the renewed interest in B-cell biology in IO, this is an area we believe warrants further clinical investigation. Citation Format: James Hair, Fabien Garcon, Michelle Hsueh, Laura Dallaway, Elena Bibikova, Maria Letizia Giardino Torchia, Gordon Moody, Alwin Schuller, Simon J. Dovedi, Zachary A. Cooper, Kris Sachsenmeier, Rakesh Kumar, Jim Eyles, Robert W. Wilkinson. Activation of B cells by CD73 blocking antibodies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1695.
- Published
- 2021
- Full Text
- View/download PDF
50. Abstract 1584: Efficacy and pharmacodynamic effect of anti-CD73/PD-L1 monoclonal antibodies in combination with chemotherapy: Observations from mouse tumor models
- Author
-
Amanda Watkins, Alwin Schuller, Fabien Garcon, Nadia Luheshi, Kristina M. Ilieva, Kelli Ryan, Stephanie Ling, Andreas Dannhorn, Gozde Kar, Tim Slidel, Jude Anderton, Brajesh P. Kaistha, Robert W. Wilkinson, Stef Mullins, Jim E. Eyles, Kris Sachsenmeier, Elena Galvani, Rajesh Kumar, and Zachary A. Cooper
- Subjects
Cancer Research ,Chemotherapy ,biology ,business.industry ,medicine.drug_class ,medicine.medical_treatment ,Monoclonal antibody ,Oncology ,PD-L1 ,Pharmacodynamics ,biology.protein ,Cancer research ,Medicine ,Mouse tumor ,business - Abstract
Intratumoral adenosine is a key immunosuppressive factor linked to poor prognosis and reduced efficacy of T cell checkpoint inhibitors. CD73 is an ectoenzyme and key node in the catabolic pathway responsible for sequential hydrolysis of extracellular ATP to adenosine. ATP is released from necrotic and damaged tumor cells; a phenomenon enhanced as consequence of cytotoxic chemotherapy or radiotherapy. A CD73 inhibiting human monoclonal IgG1-TM antibody, Oleclumab, is currently in phase 2 clinical development for treatment of patients with various solid tumors. The combination of CD73 inhibition with chemotherapy and T cell checkpoint inhibition was tested using two murine cancer models, CT26 (colorectal) or MCA205 (fibrosarcoma) implanted subcutaneously in BALB/c mice or C57BL/6 mice, respectively. Tumor bearing mice were treated with combinations of oxaliplatin and 5-fluorouracil and murine surrogate monoclonal antibodies for Oleclumab and Durvalumab (anti-PD-L1). CT26 implanted mice were also treated with the murine surrogate antibodies in the presence and absence of Docetaxel. In an attempt to define contribution of components, comparator groups received monotherapies and other iterations of the combination. Treatment with anti-CD73 and anti-PD-L1 antibodies, concomitantly with chemotherapy, resulted in improved tumor growth inhibition, plus increased proportions of complete tumor regression (P Citation Format: Jim Eyles, Amanda Watkins, Kristina Ilieva, Stef Mullins, Jude Anderton, Elena Galvani, Fabien Garcon, Kelli Ryan, Brajesh P. Kaistha, Andreas Dannhorn, Stephanie Ling, Tim Slidel, Gozde Kar, Alwin Schuller, Zachary A. Cooper, Kris Sachsenmeier, Nadia Luheshi, Rakesh Kumar, Robert W. Wilkinson. Efficacy and pharmacodynamic effect of anti-CD73/PD-L1 monoclonal antibodies in combination with chemotherapy: Observations from mouse tumor models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1584.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.