166 results on '"Awen Gallimore"'
Search Results
2. Enhanced antitumor immunity through sequential targeting of PI3Kδ and LAG3
- Author
-
Emma Jones, Andrew Godkin, Sarah Nicol Lauder, Kathryn Smart, Veerle Kersemans, Danny Allen, Jake Scott, Ana Pires, Stefan Milutinovic, Michelle Somerville, Sean Smart, Paul Kinchesh, Elena Lopez-Guadamillas, Ellyn Hughes, Martin Scurr, Lori S Friedman, Bart Vanhaesebroeck, and Awen Gallimore
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Despite striking successes, immunotherapies aimed at increasing cancer-specific T cell responses are unsuccessful in most patients with cancer. Inactivating regulatory T cells (Treg) by inhibiting the PI3Kδ signaling enzyme has shown promise in preclinical models of tumor immunity and is currently being tested in early phase clinical trials in solid tumors.Methods Mice bearing 4T1 mammary tumors were orally administered a PI3Kδ inhibitor (PI-3065) daily and tumor growth, survival and T cell infiltrate were analyzed in the tumor microenvironment. A second treatment schedule comprised PI3Kδ inhibitor with anti-LAG3 antibodies administered sequentially 10 days later.Results As observed in human immunotherapy trials with other agents, immunomodulation by PI3Kδ-blockade led to 4T1 tumor regressor and non-regressor mice. Tumor infiltrating T cells in regressors were metabolically fitter than those in non-regressors, with significant enrichments of antigen-specific CD8+ T cells, T cell factor 1 (TCF1)+ T cells and CD69− T cells, compatible with induction of a sustained tumor-specific T cell response. Treg numbers were significantly reduced in both regressor and non-regressor tumors compared with untreated tumors. The remaining Treg in non-regressor tumors were however significantly enriched with cells expressing the coinhibitory receptor LAG3, compared with Treg in regressor and untreated tumors. This striking difference prompted us to sequentially block PI3Kδ and LAG3. This combination enabled successful therapy of all mice, demonstrating the functional importance of LAG3 in non-regression of tumors on PI3Kδ inhibition therapy. Follow-up studies, performed using additional cancer cell lines, namely MC38 and CT26, indicated that a partial initial response to PI3Kδ inhibition is an essential prerequisite to a sequential therapeutic benefit of anti-LAG3 antibodies.Conclusions These data indicate that LAG3 is a key bottleneck to successful PI3Kδ-targeted immunotherapy and provide a rationale for combining PI3Kδ/LAG3 blockade in future clinical studies.
- Published
- 2020
- Full Text
- View/download PDF
3. L-Selectin Enhanced T Cells Improve the Efficacy of Cancer Immunotherapy
- Author
-
H. Angharad Watson, Ruban R. P. Durairaj, Julia Ohme, Markella Alatsatianos, Hanan Almutairi, Rebar N. Mohammed, Miriam Vigar, Sophie G. Reed, Stephen J. Paisey, Christopher Marshall, Awen Gallimore, and Ann Ager
- Subjects
L-selectin/CD62L ,T cells ,melanoma ,adoptive T cell therapy ,cancer immunotherapy ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The homing molecule, L-selectin (CD62L), is commonly used as a T cell activation marker, since expression is downregulated following engagement of the T cell receptor. Studies in mice have shown that CD62L+ central memory T cells are better at controlling tumor growth than CD62L− effector memory T cells, while L-selectin knockout T cells are poor at controlling tumor growth. Here, we test the hypothesis that T cells expressing genetically modified forms of L-selectin that are maintained following T cell activation (L-selectin enhanced T cells) are better at controlling tumor growth than wild type T cells. Using mouse models of adoptive cell therapy, we show that L-selectin enhancement improves the efficacy of CD8+ T cells in controlling solid and disseminated tumor growth. L-selectin knockout T cells had no effect. Checkpoint blockade inhibitors synergized with wild type and L-selectin enhanced T cells but had no effect in the absence of T cell transfers. Reduced tumor growth by L-selectin enhanced T cells correlated with increased frequency of CD8+ tumor infiltrating T cells 21 days after commencing therapy. Longitudinal tracking of Zirconium-89 (89Zr) labeled T cells using PET-CT showed that transferred T cells localize to tumors within 1 h and accumulate over the following 7 days. L-selectin did not promote T cell homing to tumors within 18 h of transfer, however the early activation marker CD69 was upregulated on L-selectin positive but not L-selectin knockout T cells. L-selectin positive and L-selectin knockout T cells homed equally well to tumor-draining lymph nodes and spleens. CD69 expression was upregulated on both L-selectin positive and L-selectin knockout T cells but was significantly higher on L-selectin expressing T cells, particularly in the spleen. Clonal expansion of isolated L-selectin enhanced T cells was slower, and L-selectin was linked to expression of proliferation marker Ki67. Together these findings demonstrate that maintaining L-selectin expression on tumor-specific T cells offers an advantage in mouse models of cancer immunotherapy. The beneficial role of L-selectin is unrelated to its' well-known role in T cell homing and, instead, linked to activation of therapeutic T cells inside tumors. These findings suggest that L-selectin may benefit clinical applications in T cell selection for cancer therapy and for modifying CAR-T cells to broaden their clinical scope.
- Published
- 2019
- Full Text
- View/download PDF
4. Tertiary Lymphoid Structures in Cancer: Drivers of Antitumor Immunity, Immunosuppression, or Bystander Sentinels in Disease?
- Author
-
Emily Jayne Colbeck, Ann Ager, Awen Gallimore, and Gareth Wyn Jones
- Subjects
tertiary lymphoid structures ,cancer immunotherapy ,high endothelial venules ,lymphoid neogenesis ,tumor microenvironment ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Secondary lymphoid organs are integral to initiation and execution of adaptive immune responses. These organs provide a setting for interactions between antigen-specific lymphocytes and antigen-presenting cells recruited from local infected or inflamed tissues. Secondary lymphoid organs develop as a part of a genetically preprogrammed process during embryogenesis. However, organogenesis of secondary lymphoid tissues can also be recapitulated in adulthood during de novo lymphoid neogenesis of tertiary lymphoid structures (TLSs). These ectopic lymphoid-like structures form in the inflamed tissues afflicted by various pathological conditions, including cancer, autoimmunity, infection, or allograft rejection. Studies are beginning to shed light on the function of such structures in different disease settings, raising important questions regarding their contribution to progression or resolution of disease. Data show an association between the tumor-associated TLSs and a favorable prognosis in various types of human cancer, attracting the speculation that TLSs support effective local antitumor immune responses. However, definitive evidence for the role for TLSs in fostering immune responses in vivo are lacking, with current data remaining largely correlative by nature. In fact, some more recent studies have even demonstrated an immunosuppressive, tumor-promoting role for cancer-associated TLSs. In this review, we will discuss what is known about the development of cancer-associated TLSs and the current understanding of their potential role in the antitumor immune response.
- Published
- 2017
- Full Text
- View/download PDF
5. Three-dimensional Imaging Reveals Immune-driven Tumor-associated High Endothelial Venules as a Key Correlate of Tumor Rejection Following Depletion of Regulatory T Cells
- Author
-
Stefan Milutinovic, Jun Abe, Emma Jones, Inken Kelch, Kathryn Smart, Sarah N. Lauder, Michelle Somerville, Carl Ware, Andrew Godkin, Jens V. Stein, Gib Bogle, and Awen Gallimore
- Abstract
High endothelial venules (HEV) are specialized post capillary venules that recruit naïve T cells and B cells into secondary lymphoid organs (SLO) such as lymph nodes (LN). Expansion of HEV networks in SLOs occurs following immune activation to support development of an effective immune response. In this study, we used a carcinogen-induced model of fibrosarcoma to examine HEV remodeling after depletion of regulatory T cells (Treg). We used light sheet fluorescence microscopy imaging to visualize entire HEV networks, subsequently applying computational tools to enable topological mapping and extraction of numerical descriptors of the networks. While these analyses revealed profound cancer- and immune-driven alterations to HEV networks within LNs, these changes did not identify successful responses to treatment. The presence of HEV networks within tumors did however clearly distinguish responders from nonresponders. Finally, we show that a successful treatment response is dependent on coupling tumor-associated HEV (TA-HEV) development to T-cell activation implying that T-cell activation acts as the trigger for development of TA-HEVs which subsequently serve to amplify the immune response by facilitating extravasation of T cells into the tumor mass. Significance: We used three-dimensional imaging methods with computational tools to analyze networks of specialized blood vessels called HEVs in LNs and tumors. By applying these techniques in a mouse model of carcinogen-induced tumors, we could identify network changes after depletion of Tregs.
- Published
- 2022
- Full Text
- View/download PDF
6. The Link between T cell Activation and Development of Functionally Useful Tumour Associated High Endothelial Venules
- Author
-
Stefan Milutinovic and Awen Gallimore
- Abstract
High endothelial venules (HEVs) are specialised postcapillary venules that specifically serve to recruit circulating lymphocytes to secondary lymphoid organs (SLOs) where cognate antigens can be encountered, and immune responses can be initiated. The presence of HEV-like vessels in primary human solid tumours and their association with lymphocyte infiltration and favourable clinical outcomes and response to immunotherapy have provided a rationale for therapeutically inducing these vessels in tumours for immunotherapeutic benefit. Here we specifically discuss evidence for a link between T cell activation and development of useful tumour associated HEV (TA-HEV). We discuss the molecular and functional features of TA-HEV, highlighting the benefits for promoting tumour immunity and the important unanswered questions that need to be addressed before TA-HEV induction can be optimised for immunotherapeutic benefit.
- Published
- 2023
- Full Text
- View/download PDF
7. Figure S2 from Three-dimensional Imaging Reveals Immune-driven Tumor-associated High Endothelial Venules as a Key Correlate of Tumor Rejection Following Depletion of Regulatory T Cells
- Author
-
Awen Gallimore, Gib Bogle, Jens V. Stein, Andrew Godkin, Carl Ware, Michelle Somerville, Sarah N. Lauder, Kathryn Smart, Inken Kelch, Emma Jones, Jun Abe, and Stefan Milutinovic
- Abstract
Naïve LN HEV Characterisation.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figures from Immune Remodeling of the Extracellular Matrix Drives Loss of Cancer Stem Cells and Tumor Rejection
- Author
-
Awen Gallimore, Andrew Godkin, Robert Andrews, William J. Watkins, Matthew J. Smalley, Rhiannon French, James P. Hindley, Howard Kendrick, Stefan Milutinovic, Michelle Somerville, Sarah N. Lauder, Kathryn Smart, Emma Jones, Alexander Greenshields-Watson, and Ana Pires
- Abstract
Supplementary Figures 1-8
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Three-dimensional Imaging Reveals Immune-driven Tumor-associated High Endothelial Venules as a Key Correlate of Tumor Rejection Following Depletion of Regulatory T Cells
- Author
-
Awen Gallimore, Gib Bogle, Jens V. Stein, Andrew Godkin, Carl Ware, Michelle Somerville, Sarah N. Lauder, Kathryn Smart, Inken Kelch, Emma Jones, Jun Abe, and Stefan Milutinovic
- Abstract
High endothelial venules (HEV) are specialized post capillary venules that recruit naïve T cells and B cells into secondary lymphoid organs (SLO) such as lymph nodes (LN). Expansion of HEV networks in SLOs occurs following immune activation to support development of an effective immune response. In this study, we used a carcinogen-induced model of fibrosarcoma to examine HEV remodeling after depletion of regulatory T cells (Treg). We used light sheet fluorescence microscopy imaging to visualize entire HEV networks, subsequently applying computational tools to enable topological mapping and extraction of numerical descriptors of the networks. While these analyses revealed profound cancer- and immune-driven alterations to HEV networks within LNs, these changes did not identify successful responses to treatment. The presence of HEV networks within tumors did however clearly distinguish responders from nonresponders. Finally, we show that a successful treatment response is dependent on coupling tumor-associated HEV (TA-HEV) development to T-cell activation implying that T-cell activation acts as the trigger for development of TA-HEVs which subsequently serve to amplify the immune response by facilitating extravasation of T cells into the tumor mass.Significance:We used three-dimensional imaging methods with computational tools to analyze networks of specialized blood vessels called HEVs in LNs and tumors. By applying these techniques in a mouse model of carcinogen-induced tumors, we could identify network changes after depletion of Tregs.
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Immune Remodeling of the Extracellular Matrix Drives Loss of Cancer Stem Cells and Tumor Rejection
- Author
-
Awen Gallimore, Andrew Godkin, Robert Andrews, William J. Watkins, Matthew J. Smalley, Rhiannon French, James P. Hindley, Howard Kendrick, Stefan Milutinovic, Michelle Somerville, Sarah N. Lauder, Kathryn Smart, Emma Jones, Alexander Greenshields-Watson, and Ana Pires
- Abstract
The nature of the tumor microenvironment (TME) influences the ability of tumor-specific T cells to control tumor growth. In this study, we performed an unbiased comparison of the TME of regulatory T-cell (Treg)–replete and Treg-depleted carcinogen-induced tumors, including Treg-depleted responding (regressing) and non-responding (growing) tumors. This analysis revealed an inverse relationship between extracellular matrix (ECM) and T-cell infiltrates where responding tumors were T-cell rich and ECM poor, whereas the converse was observed in non-responder tumors. For this reason, we hypothesized that the ECM acted as a barrier to successful T-cell infiltration and tumor rejection. However, further experiments revealed that this was not the case but instead showed that an effective T-cell response dramatically altered the density of ECM in the TME. Along with loss of ECM and high numbers of infiltrating T cells, responder tumors were distinguished by the development of lymphatic and blood vessel networks with specialized immune function. ECM-rich tumors exhibited a stem cell–like gene expression profile and superior tumor-initiating capacity, whereas such features were absent in responder tumors. Overall, these findings define an extended role for an effective immune response, not just in direct killing of tumor cells but in widescale remodeling of the TME to favor loss of ECM, elimination of cancer stem cells, and propagation of adaptive immunity.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Movie 1 from Three-dimensional Imaging Reveals Immune-driven Tumor-associated High Endothelial Venules as a Key Correlate of Tumor Rejection Following Depletion of Regulatory T Cells
- Author
-
Awen Gallimore, Gib Bogle, Jens V. Stein, Andrew Godkin, Carl Ware, Michelle Somerville, Sarah N. Lauder, Kathryn Smart, Inken Kelch, Emma Jones, Jun Abe, and Stefan Milutinovic
- Abstract
Global overview of intra-tumoral HEVs in a 4T1 regressor tumour following PI-3065 treatment.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Movie 2 from Three-dimensional Imaging Reveals Immune-driven Tumor-associated High Endothelial Venules as a Key Correlate of Tumor Rejection Following Depletion of Regulatory T Cells
- Author
-
Awen Gallimore, Gib Bogle, Jens V. Stein, Andrew Godkin, Carl Ware, Michelle Somerville, Sarah N. Lauder, Kathryn Smart, Inken Kelch, Emma Jones, Jun Abe, and Stefan Milutinovic
- Abstract
Global overview of intra-tumoral HEVs in a 4T1 regressor tumour following PI-3065 treatment.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Movie 3 from Three-dimensional Imaging Reveals Immune-driven Tumor-associated High Endothelial Venules as a Key Correlate of Tumor Rejection Following Depletion of Regulatory T Cells
- Author
-
Awen Gallimore, Gib Bogle, Jens V. Stein, Andrew Godkin, Carl Ware, Michelle Somerville, Sarah N. Lauder, Kathryn Smart, Inken Kelch, Emma Jones, Jun Abe, and Stefan Milutinovic
- Abstract
Global overview of intra-tumoral HEVs in a 4T1 non-regressor tumour following PI-3065 treatment.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Data from Treg Depletion Licenses T Cell–Driven HEV Neogenesis and Promotes Tumor Destruction
- Author
-
Awen Gallimore, Ann Ager, Carl F. Ware, Andrew Godkin, Lee Parry, Anwen Williams, Scott Cutting, Molly Browne, Ralph Schulz, Kathryn Smart, James P. Hindley, Emma Jones, and Emily J. Colbeck
- Abstract
S1. Tumor HEV identified by PNAd staining are also positive for MAdCAM-1. S2. Efficacy of monoclonal antibody treatments to deplete immune cell subsets. S3. Splenic Marginal Zone B cells are profoundly decreased after treatment with LTbetaR.Fc. S4. Splenic Follicular Dendritic Cells and MAdCAM-1 staining are lost after treatment with LTbetaR.Fc or TNFRII.Ig. S5. Lymph Node architecture is disrupted following treatment with LTbetaR.Fc or TNFRII.Ig. S6. Tumor HEV identified by PNAd staining following blockade of LTbetaR or TNFR signalling are also positive for MAdCAM-1. S7. Agonism of LTβR induces formation of High Endothelial Venules in Treg replete tumors, but without concomitant increased T cell infiltration and reduced tumor growth. S8. Relative gene expression of TNF and LTalpha by intratumoral CD4+ and CD8+ T cells and dendritic cells (DC). Data are expressed as fold change in gene expression relative to splenic B cells, and represent two independent experiments.
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Treg Depletion Licenses T Cell–Driven HEV Neogenesis and Promotes Tumor Destruction
- Author
-
Awen Gallimore, Ann Ager, Carl F. Ware, Andrew Godkin, Lee Parry, Anwen Williams, Scott Cutting, Molly Browne, Ralph Schulz, Kathryn Smart, James P. Hindley, Emma Jones, and Emily J. Colbeck
- Abstract
T-cell infiltration into tumors represents a critical bottleneck for immune-mediated control of cancer. We previously showed that this bottleneck can be overcome by depleting immunosuppressive Foxp3+ regulatory T cells (Tregs), a process that can increase frequencies of tumor-infiltrating lymphocytes through promoting the development of specialized portals for lymphocyte entry, namely high endothelial venules (HEVs). In this paper, we used a carcinogen-induced tumor model that allows for coevolution of the tumor microenvironment and the immune response to demonstrate that Treg depletion not only results in widespread disruption to HEV networks in lymph nodes (LNs) but also activates CD8+ T cells, which then drive intratumoral HEV development. Formation of these vessels contrasts with ontogenic HEV development in LNs in that the process is dependent on the TNF receptor and independent of lymphotoxin β receptor–mediated signaling. These intratumoral HEVs do not express the chemokine CCL21, revealing a previously undescribed intratumoral blood vessel phenotype. We propose a model where Treg depletion enables a self-amplifying loop of T-cell activation, which promotes HEV development, T-cell infiltration, and ultimately, tumor destruction. The findings point to a need to test for HEV development as part of ongoing clinical studies in patients with cancer. Cancer Immunol Res; 5(11); 1005–15. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figures 1 - 4 from Escalating Regulation of 5T4-Specific IFN-γ+ CD4+ T Cells Distinguishes Colorectal Cancer Patients from Healthy Controls and Provides a Target for In Vivo Therapy
- Author
-
Andrew Godkin, Awen Gallimore, Tom Hockey, Adam Christian, Simon Phillips, Rachel Hargest, Mike Davies, Hayley Bridgeman, Kathryn Smart, Clare Brown, Rohit Srinivasan, Tom Pembroke, Anja Bloom, and Martin Scurr
- Abstract
PDF file - 10925K, S. Fig. 1. Cultured 5T4-specific CD4+ T cells can be cloned from CRC patients (A) and healthy donors (B). S. Fig. 2. A comparison of 5T4 responses and age amongst CRC patients. S. Fig. 3. 5T4 Immunohistochemistry staining of frozen sections. S. Fig. 4. CT scans demonstrating the growth of a single metastatic lung lesion in a patient who did not respond to low-dose cyclophosphamide treatment.
- Published
- 2023
- Full Text
- View/download PDF
17. Data from Escalating Regulation of 5T4-Specific IFN-γ+ CD4+ T Cells Distinguishes Colorectal Cancer Patients from Healthy Controls and Provides a Target for In Vivo Therapy
- Author
-
Andrew Godkin, Awen Gallimore, Tom Hockey, Adam Christian, Simon Phillips, Rachel Hargest, Mike Davies, Hayley Bridgeman, Kathryn Smart, Clare Brown, Rohit Srinivasan, Tom Pembroke, Anja Bloom, and Martin Scurr
- Abstract
The relationship between the adaptive CD4+ T-cell response and human cancer is unclear. The oncofetal antigen 5T4 is expressed in many human carcinomas, including colorectal cancer cells, but has limited expression on normal tissues. We previously identified anti-5T4 CD4+ T cells in a proportion of patients with colorectal cancer, and we extended this study to examine whether the quality or quantity of the T-cell response reflects tumor stage. An overlapping peptide library spanning 5T4 was used as a target to enumerate cognate IFN-γ+ CD4+ T cells [measured as spot-forming cells (SFC)/105 cultured T cells] in peripheral blood–derived lymphocytes following a 14-day in vitro culture period comparing patients preoperatively (n = 27) to healthy controls (n = 17). Robust 5T4-specific T-cell responses were present in 100% of healthy donors. There was a steady loss of T-cell responses with advancing tumors with a significant negative correlation from stage I to III (P = 0.008). The predictability of the decline meant 5 were only found in subjects with stage III colorectal cancer. The mechanism of loss of T-cell response is independent of HLA-DR type or patient age but does correspond to increases in Foxp3+ regulatory T cells (Treg). Using low-dose cyclophosphamide to reduce the proportion of Tregs in vivo resulted in increased anti-5T4 T-cell responses in patients with colorectal cancer. The selective loss of 5T4-specific IFN-γ+ CD4+ T-cell responses implies a link between tumor stage and antitumor Th1 effector function; depleting Tregs can enhance such responses. Cancer Immunol Res; 1(6); 416–25. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure Legend from Escalating Regulation of 5T4-Specific IFN-γ+ CD4+ T Cells Distinguishes Colorectal Cancer Patients from Healthy Controls and Provides a Target for In Vivo Therapy
- Author
-
Andrew Godkin, Awen Gallimore, Tom Hockey, Adam Christian, Simon Phillips, Rachel Hargest, Mike Davies, Hayley Bridgeman, Kathryn Smart, Clare Brown, Rohit Srinivasan, Tom Pembroke, Anja Bloom, and Martin Scurr
- Abstract
PDF file - 65K
- Published
- 2023
- Full Text
- View/download PDF
19. Table S1 from Treg Depletion Licenses T Cell–Driven HEV Neogenesis and Promotes Tumor Destruction
- Author
-
Awen Gallimore, Ann Ager, Carl F. Ware, Andrew Godkin, Lee Parry, Anwen Williams, Scott Cutting, Molly Browne, Ralph Schulz, Kathryn Smart, James P. Hindley, Emma Jones, and Emily J. Colbeck
- Abstract
Table S1
- Published
- 2023
- Full Text
- View/download PDF
20. Data from Cancer Antigen Discovery Is Enabled by RNA Sequencing of Highly Purified Malignant and Nonmalignant Cells
- Author
-
Andrew Godkin, Awen Gallimore, Richard J. Stanton, Alan L. Parker, Robert Andrews, Kevin E. Ashelford, Adam D. Christian, Simon Phillips, Rachel Hargest, Michael M. Davies, James A. Davies, Stephanie E.A. Burnell, Sarah L. Hulin-Curtis, Yuan Chen, Michelle S. Somerville, Emma Campbell, Alex Greenshields-Watson, and Martin J. Scurr
- Abstract
Purpose:Broadly expressed, highly differentiated tumor-associated antigens (TAA) can elicit antitumor immunity. However, vaccines targeting TAAs have demonstrated disappointing clinical results, reflecting poor antigen selection and/or immunosuppressive mechanisms.Experimental Design:Here, a panel of widely expressed, novel colorectal TAAs were identified by performing RNA sequencing of highly purified colorectal tumor cells in comparison with patient-matched colonic epithelial cells; tumor cell purification was essential to reveal these genes. Candidate TAA protein expression was confirmed by IHC, and preexisting T-cell immunogenicity toward these antigens tested.Results:The most promising candidate for further development is DNAJB7 [DnaJ heat shock protein family (Hsp40) member B7], identified here as a novel cancer-testis antigen. It is expressed in many tumors and is strongly immunogenic in patients with cancers originating from a variety of sites. DNAJB7-specific T cells were capable of killing colorectal tumor lines in vitro, and the IFNγ+ response was markedly magnified by control of immunosuppression with cyclophosphamide in patients with cancer.Conclusions:This study highlights how prior methods that sequence whole tumor fractions (i.e., inclusive of alive/dead stromal cells) for antigen identification may have limitations. Through tumor cell purification and sequencing, novel candidate TAAs have been identified for future immunotherapeutic targeting.
- Published
- 2023
- Full Text
- View/download PDF
21. Data from Low-Dose Cyclophosphamide Induces Antitumor T-Cell Responses, which Associate with Survival in Metastatic Colorectal Cancer
- Author
-
Andrew Godkin, Awen Gallimore, Robert Hills, Richard Harrop, Daniel Blount, Sarah Gwynne, Robert Jones, Alison Brewster, Richard Adams, Hayley Bridgeman, Kathryn Smart, Amanda Thomson, David Roberts, Anja Bloom, Tom Pembroke, and Martin Scurr
- Abstract
Purpose: Anticancer T-cell responses can control tumors, but immunosuppressive mechanisms in vivo prevent their function. The role of regulatory T cells (Tregs) in metastatic colorectal cancer is unclear. We have previously shown depletion of Tregs enhances colorectal cancer–specific effector T-cell responses. Low-dose cyclophosphamide targets Tregs in animal models and some human studies; however, the effect of cyclophosphamide in metastatic colorectal cancer is unknown.Experimental Design: Fifty-five patients with metastatic colorectal cancer were enrolled in a phase I/II trial and randomly assigned to receive 2-week-long courses of low-dose (50 mg twice a day) cyclophosphamide or not. The absolute number, phenotype, and antitumor function of peripheral blood–derived lymphocyte subsets were monitored throughout treatment, as well as during 18-month follow-up.Results: Initially, cyclophosphamide reduced proliferation in all lymphocyte subsets; however, a rapid mobilization of effector T cells overcame this decrease, leading to increased absolute T-cell numbers. In contrast, a reduction in proportional and absolute Treg, B-cell, and NK-cell numbers occurred. The expansion and subsequent activation of effector T cells was focused on tumor-specific T cells, producing both granzyme B and IFNγ. Cyclophosphamide-treated patients demonstrating the most enhanced IFNγ+ tumor-specific T-cell responses exhibited a significant delay in tumor progression [HR = 0.29; 95% confidence interval (CI), 0.12–0.69; P = 0.0047), compared with nonresponders and no-treatment controls.Conclusions: Cyclophosphamide-induced Treg depletion is mirrored by a striking boost in antitumor immunity. This study provides the first direct evidence of the benefit of naturally primed T cells in patients with metastatic colorectal cancer. Our results also support the concept that nonmutated self-antigens may act as useful targets for immunotherapies. Clin Cancer Res; 23(22); 6771–80. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Data from Low-Dose Cyclophosphamide Induces Antitumor T-Cell Responses, which Associate with Survival in Metastatic Colorectal Cancer
- Author
-
Andrew Godkin, Awen Gallimore, Robert Hills, Richard Harrop, Daniel Blount, Sarah Gwynne, Robert Jones, Alison Brewster, Richard Adams, Hayley Bridgeman, Kathryn Smart, Amanda Thomson, David Roberts, Anja Bloom, Tom Pembroke, and Martin Scurr
- Abstract
Supplementary Figure 1. CONSORT diagram. Supplementary Figure 2. 5T4 peptide pools and matrix system. Supplementary Figure 3. B-cell number in response to cyclophosphamide. Supplementary Figure 4. Expression of activation markers in T-cell subsets during CPM treatment. Supplementary Figure 5. Immunological responses following the completion of CPM treatment. Supplementary Figure 6. Analysis of immunological responses over 8 courses of CPM in patient 102.
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figures from Cancer Antigen Discovery Is Enabled by RNA Sequencing of Highly Purified Malignant and Nonmalignant Cells
- Author
-
Andrew Godkin, Awen Gallimore, Richard J. Stanton, Alan L. Parker, Robert Andrews, Kevin E. Ashelford, Adam D. Christian, Simon Phillips, Rachel Hargest, Michael M. Davies, James A. Davies, Stephanie E.A. Burnell, Sarah L. Hulin-Curtis, Yuan Chen, Michelle S. Somerville, Emma Campbell, Alex Greenshields-Watson, and Martin J. Scurr
- Abstract
Supplementary Figures 1-7
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Figure 5 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Author
-
Awen Gallimore, Julian Dyson, Andrew J. Godkin, David A. Price, Yogesh Singh, Gareth J. Betts, Katherine K. Wynn, Kristin Ladell, Sarah N. Lauder, Emma Jones, Cristina Ferreira, and James P. Hindley
- Abstract
Supplementary Figure 5 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Figure 6 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Author
-
Awen Gallimore, Julian Dyson, Andrew J. Godkin, David A. Price, Yogesh Singh, Gareth J. Betts, Katherine K. Wynn, Kristin Ladell, Sarah N. Lauder, Emma Jones, Cristina Ferreira, and James P. Hindley
- Abstract
Supplementary Figure 6 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figures 1-3 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Author
-
Awen Gallimore, Julian Dyson, Andrew J. Godkin, David A. Price, Yogesh Singh, Gareth J. Betts, Katherine K. Wynn, Kristin Ladell, Sarah N. Lauder, Emma Jones, Cristina Ferreira, and James P. Hindley
- Abstract
Supplementary Figures 1-3 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Figure 7 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Author
-
Awen Gallimore, Julian Dyson, Andrew J. Godkin, David A. Price, Yogesh Singh, Gareth J. Betts, Katherine K. Wynn, Kristin Ladell, Sarah N. Lauder, Emma Jones, Cristina Ferreira, and James P. Hindley
- Abstract
Supplementary Figure 7 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Figure 4 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Author
-
Awen Gallimore, Julian Dyson, Andrew J. Godkin, David A. Price, Yogesh Singh, Gareth J. Betts, Katherine K. Wynn, Kristin Ladell, Sarah N. Lauder, Emma Jones, Cristina Ferreira, and James P. Hindley
- Abstract
Supplementary Figure 4 from Analysis of the T-Cell Receptor Repertoires of Tumor-Infiltrating Conventional and Regulatory T Cells Reveals No Evidence for Conversion in Carcinogen-Induced Tumors
- Published
- 2023
- Full Text
- View/download PDF
29. PI3K Isoform Immunotherapy for Solid Tumours
- Author
-
Jake, Scott, Lauren, Rees, Awen, Gallimore, and Sarah N, Lauder
- Subjects
Phosphatidylinositol 3-Kinases ,Neoplasms ,Humans ,Protein Isoforms ,Immunotherapy ,CD8-Positive T-Lymphocytes ,Phosphatidylinositol 3-Kinase ,Phosphatidylinositols ,Immune Checkpoint Inhibitors ,Phosphoinositide-3 Kinase Inhibitors - Abstract
Improving the anti-tumour T cell response as a consequence of immunotherapy can result in eradication of tumour burden, however, the majority of patients fail with current treatment regimens and so novel immunotherapies with greater efficacy and improved tolerability are needed. The phosphoinositide-3-kinase (PI3K) family members that are directly involved in cell signalling comprise PI3Kα, PI3Kβ, PI3Kδ and PI3Kγ, with the latter two isoforms expressed primarily by leukocytes. The survival and optimal function of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs) is dependent on PI3Kδ, whereas tumour-associated macrophages (TAMs), use PI3Kγ. Blocking these signalling isoforms can boost development of effective anti-cancer immune responses and result in control of tumour burden. The dependence on different PI3K isoforms in immune cells makes targeting this pathway an attractive approach for tumour immunotherapy. Herein, we discuss how inhibiting specific PI3K isoforms in pro-tumoural Tregs, MDSCS and TAMs can unleash a powerful anti-tumour immune response, driven by CD8
- Published
- 2022
30. The Dual Role of High Endothelial Venules in Cancer Progression versus Immunity
- Author
-
Stefan Milutinovic, Awen Gallimore, Jens V. Stein, Jun Abe, and Andrew James Godkin
- Subjects
0301 basic medicine ,Cancer Research ,Cell Plasticity ,High endothelial venules ,Cell ,Spleen ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Cell Movement ,Immunity ,Neoplasms ,Parenchyma ,medicine ,Humans ,Cancer ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Endothelium, Lymphatic ,Sentinel Lymph Node ,Function (biology) - Abstract
Secondary lymphoid organs (SLOs) are important initiators and regulators of immunity. To carry out this function, the blood vasculature must deliver oxygen and nutrients and recruit circulating lymphocytes into the SLO parenchyma, where they encounter cognate antigen. High endothelial venules (HEVs) are specialised postcapillary venules that specifically serve this function and are found in all SLOs except spleen. It is becoming clear that alterations to HEV network density and/or morphology can result in immune activation or, as recently implicated, in providing an exit route for tumour cell dissemination and metastases. In this review, the structural plasticity of HEVs, the regulatory pathways underpinning this plasticity, and the relevance of these pathways to cancer progression will be discussed.
- Published
- 2021
- Full Text
- View/download PDF
31. Molecular characterization of HLA class II binding to the LAG‐3 T cell co‐inhibitory receptor
- Author
-
Georgina H. Mason, David K. Cole, Frédéric Triebel, Awen Gallimore, Andrew James Godkin, Bruce J. MacLachlan, and Alexander Greenshields-Watson
- Subjects
0301 basic medicine ,Molecular immunology and signaling ,LAG3 ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Immunology ,Cell ,T cells ,Receptors, Antigen, T-Cell ,Biology ,immune checkpoint inhibitors ,Jurkat Cells ,03 medical and health sciences ,0302 clinical medicine ,LAG‐3 ,Costimulatory and Inhibitory T-Cell Receptors ,Downregulation and upregulation ,Cancer immunotherapy ,Antigens, CD ,HLA Antigens ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Immunology and Allergy ,Basic ,Receptor ,Research Articles ,cancer immunotherapy ,Lymphocyte Activation Gene 3 Protein ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,CD4 Antigens ,pHLA‐II ,biology.protein ,Research Article|Basic ,Antibody ,Function (biology) ,030215 immunology - Abstract
Immune checkpoint inhibitors (antibodies that block the T cell co‐inhibitory receptors PD‐1/PD‐L1 or CTLA‐4) have revolutionized the treatment of some forms of cancer. Importantly, combination approaches using drugs that target both pathways have been shown to boost the efficacy of such treatments. Subsequently, several other T cell inhibitory receptors have been identified for the development of novel immune checkpoint inhibitors. Included in this list is the co‐inhibitory receptor lymphocyte activation gene‐3 (LAG‐3), which is upregulated on T cells extracted from tumor sites that have suppressive or exhausted phenotypes. However, the molecular rules that govern the function of LAG‐3 are still not understood. Using surface plasmon resonance combined with a novel bead‐based assay (AlphaScreenTM), we demonstrate that LAG‐3 can directly and specifically interact with intact human leukocyte antigen class II (HLA‐II) heterodimers. Unlike the homologue CD4, which has an immeasurably weak affinity using these biophysical approaches, LAG‐3 binds with low micromolar affinity. We further validated the interaction at the cell surface by staining LAG‐3+ cells with pHLA‐II‐multimers. These data provide new insights into the mechanism by which LAG‐3 initiates T cell inhibition., We demonstrate that human LAG‐3 binds directly to peptide‐human leukocyte antigen class II complex (pHLA‐II) with low micromolar affinity. These findings provide new insights into the mechanism by which LAG‐3 initiates T cell inhibition and has implication for the development of novel immune checkpoint inhibitors for cancer immunotherapy.
- Published
- 2020
- Full Text
- View/download PDF
32. Primary breast tumours but not lung metastases induce protective anti-tumour immune responses after Treg-depletion
- Author
-
Andrew James Godkin, Andrew Blainey, Anja Bloom, Sarah Nicol Lauder, Jake Scott, Awen Gallimore, Emma Jones, Molly Browne, E. Hughes, Michelle Somerville, Ann Ager, and Kathryn Smart
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Mouse ,medicine.medical_treatment ,Immunology ,T cells ,Triple Negative Breast Neoplasms ,Tregs ,chemical and pharmacologic phenomena ,Disease ,T-Lymphocytes, Regulatory ,Lymphocyte Depletion ,Metastasis ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Breast cancer ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Immunology and Allergy ,Lung ,business.industry ,FOXP3 ,Cancer ,Forkhead Transcription Factors ,Immunotherapy ,Resection ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Original Article ,business - Abstract
Although metastatic disease is responsible for the majority of cancer deaths, tests of novel immunotherapies in mouse tumour models often focus on primary tumours without determining whether these therapies also target metastatic disease. This study examined the impact of depleting Foxp3+ regulatory T cells (Treg), on lung metastases, using a mouse model of breast cancer. After Treg-depletion, generation of an immune response to the primary tumour was a critical determinant for limiting development of metastasis. Indeed, resection of the primary tumour abrogated any effect of Treg-depletion on metastases. In addition, whilst the immune response, generated by the primary tumour, prevented metastases development, it had little impact on controlling established disease. Collectively, the data indicate that metastatic cells in the lung are not controlled by immune responses induced by the primary tumour. These findings indicate that targeting Tregs alone will not suffice for treating lung metastases. Electronic supplementary material The online version of this article (10.1007/s00262-020-02603-x) contains supplementary material, which is available to authorised users.
- Published
- 2020
- Full Text
- View/download PDF
33. Tumor-Associated High Endothelial Venules: Inroads Enabling Immune Control of Cancer Progression
- Author
-
Awen Gallimore
- Subjects
Cancer Research ,Tertiary Lymphoid Structures ,Venules ,Neoplasms ,Immunology ,Humans ,Lymph Nodes ,Lymphocytes ,Article - Abstract
High endothelial venules (HEVs) are specialized post-capillary venules that recruit naïve lymphocytes to lymph nodes. HEVs are essential for the development of adaptive immunity. HEVs can also develop in tumors where they are thought to be important for recruiting naïve T cells and B cells into the tumors and locally enhancing antitumor immunity by supporting the formation of tertiary lymphoid structures. Herein, we used comparative transcriptome analysis of human breast cancer to investigate genes differentially expressed between tumor-associated HEVs and the rest of the tumor vasculature. Tumor vessels highly expressing HEV-upregulated genes, such as the homeobox gene MEOX2 and the tetraspanin gene TSPAN7, were associated with extensive infiltration of T and B cells and the occurrence of tertiary lymphoid structures, which is known to predict therapeutic responses to immune checkpoint inhibitors. Moreover, high transcript counts of these genes in clinical tumor specimens were associated with a significant survival benefit in advanced breast cancer. The molecular signature of HEVs identified herein may be useful for guiding immunotherapies and provides a new direction for investigating tumor-associated HEVs and their clinical significance.
- Published
- 2022
34. Exploiting ECM remodelling to promote immune-mediated tumour destruction
- Author
-
Awen Gallimore, Ana Pires, and Stephanie E A Burnell
- Subjects
Antigenicity ,T cell ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Biology ,Cancer treatment ,Extracellular Matrix ,Extracellular matrix ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,Tumour destruction ,Neoplasms ,Cancer research ,medicine ,Tumor Microenvironment ,Immunology and Allergy ,Humans ,Immunotherapy - Abstract
Cancer immunotherapy represents a significant breakthrough in cancer treatment mainly due to the ability to harness the activities of cancer-specific T cells. Despite this, most cancers remain resistant to T cell attack. Many reasons have been proposed to explain this, ranging from a lack of antigenicity through to the immunosuppressive effects of the tumour microenvironment. In this review, we examine the relationship between the immune system and a key component of the tumour microenvironment, namely the extracellular matrix (ECM). Specifically, we explore the reciprocal effects of immune cells and the tumour ECM and how the processes underpinning this relationship act to either promote or restrain tumour progression.
- Published
- 2022
35. PI3K Isoform Immunotherapy for Solid Tumours
- Author
-
Jake Scott, Lauren Rees, Awen Gallimore, and Sarah N. Lauder
- Published
- 2022
- Full Text
- View/download PDF
36. Whole blood‐based measurement of SARS‐CoV‐2‐specific T cells reveals asymptomatic infection and vaccine immunogenicity in healthy subjects and patients with solid‐organ cancers
- Author
-
Charlotte Young, Lucy C. Fairclough, Andrew D. Westwell, Kerry Roberts, Kate Davies, Thomas Tozer, Stephanie E A Burnell, Paddy Tighe, Tara Rees, Amanda Jackson, B. Paul Morgan, Awen Gallimore, Martin J. Scurr, Michelle Somerville, Mererid Evans, Andrew James Godkin, George Lippiatt, Wioleta M. Zelek, Mark R. Wills, Helen Lawton, and Lorenzo Capitani
- Subjects
Adult ,Male ,COVID-19 Vaccines ,Adolescent ,T cell ,Immunology ,T cells ,Asymptomatic ,SARS‐CoV‐2 ,Interferon-gamma ,Immunogenicity, Vaccine ,Immunity ,COVID‐19 ,vaccine ,Blood plasma ,medicine ,Immunology and Allergy ,antibodies ,Humans ,Aged ,Aged, 80 and over ,Immunity, Cellular ,biology ,business.industry ,SARS-CoV-2 ,Vaccination ,Cancer ,COVID-19 ,Original Articles ,Middle Aged ,Th1 Cells ,medicine.disease ,Vaccine efficacy ,medicine.anatomical_structure ,Carrier State ,biology.protein ,Original Article ,Female ,Antibody ,medicine.symptom ,business - Abstract
Accurate assessment of SARS‐CoV‐2 immunity is critical in evaluating vaccine efficacy and devising public health policies. Whilst the exact nature of effective immunity remains incompletely defined, SARS‐CoV‐2‐specific T‐cell responses are a critical feature that will likely form a key correlate of protection against COVID‐19. Here, we developed and optimized a high‐throughput whole blood‐based assay to determine the T‐cell response associated with prior SARS‐CoV‐2 infection and/or vaccination amongst 231 healthy donors and 68 cancer patients. Following overnight in vitro stimulation with SARS‐CoV‐2‐specific peptides, blood plasma samples were analysed for TH1‐type cytokines. Highly significant differential IFN‐γ+/IL‐2+ SARS‐CoV‐2‐specific T‐cell responses were seen amongst previously infected COVID‐19‐positive healthy donors in comparison with unknown / naïve individuals (p, SARS‐CoV‐2 pandemic continues to cause morbidity and mortality especially in vulnerable patients. Monitoring individuals T and B cell responses to the virus is desirable in high risk populations after infection and/or vaccination. This paper describes a straightforward approach on a small whole blood sample to measure these responses.
- Published
- 2021
37. Sequential targeting of PI3Kδ and LAG3 as an effective anti-cancer approach
- Author
-
Sarah Nicol Lauder, Awen Gallimore, and Bart Vanhaesebroeck
- Subjects
Cancer Research ,LAG3 ,Class I Phosphatidylinositol 3-Kinases ,medicine.medical_treatment ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Antigens, CD ,Neoplasms ,Tumor Microenvironment ,Animals ,Humans ,Medicine ,Immune Checkpoint Inhibitors ,030304 developmental biology ,0303 health sciences ,business.industry ,Comment ,Cancer ,Drug Synergism ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Lymphocyte Activation Gene 3 Protein ,Immune checkpoint ,3. Good health ,Blockade ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Tumour immunology ,business - Abstract
SummaryEmerging studies have demonstrated the potential of PI3Kδ blockade as an immunotherapy for solid tumours. In pre-clinical models, we recently demonstrated that anti-LAG3 immune checkpoint blockade vastly potentiated PI3Kδ-based immunotherapy, enabling successful tumour control in all treated mice.
- Published
- 2021
- Full Text
- View/download PDF
38. Treg-driven tumour control by PI3Kδ inhibition limits myeloid-derived suppressor cell expansion
- Author
-
Sarah N. Lauder, Kathryn Smart, Valentina M. T. Bart, Ana Pires, Jake Scott, Stefan Milutinovic, Andrew Godkin, Bart Vanhaesebroeck, and Awen Gallimore
- Subjects
Cancer Research ,Mice ,Oncology ,Myeloid-Derived Suppressor Cells ,Neoplasms ,Tumor Microenvironment ,Animals ,T-Lymphocytes, Regulatory ,Cell Proliferation - Abstract
Background Recent studies have demonstrated that blocking the PI3Kδ signalling enzyme (by administering a small molecule inhibitor, PI-3065) can potently improve the anti-tumour T-cell response through direct inhibition of Tregs. This treatment also has a negative impact on MDSC numbers but the primary mechanism driving this effect has remained unclear. Methods The 4T1 breast cancer mouse model was used in combination with PI-3065 to gain insights into the effect of PI3Kδ inhibition on MDSCs. Results PI-3065 treatment resulted in a concomitant reduction in MDSC expansion and tumour size. However, targeting Tregs independent of PI-3065 was also associated with reduced tumour volume and MDSC numbers. Surgical removal of tumours resulted in a rapid and significant decline in MDSC numbers, whilst ex vivo studies using cells from PI-3065-treated mice demonstrated no direct effect of the inhibitor on MDSC activity. Conclusions Our data suggest that MDSCs are not inhibited directly by PI-3065 treatment but that their reduced recruitment and immunosuppression within the tumour microenvironment is an indirect consequence of PI3Kδ-inhibition-driven tumour control. This indicates that PI3Kδ inhibition drives tumour immunity by breaking down multiple immunosuppressive pathways through both direct mechanisms (on Treg) and indirect mechanisms, secondary to tumour control (on MDSCs).
- Published
- 2021
39. Pouring petrol on the flames: using oncolytic virotherapies to enhance tumour immunogenicity
- Author
-
Andrew James Godkin, Alan L. Parker, Elise Moses, Rebecca J. Bayliss, Georgina H. Mason, Alicia Teijeira Crespo, Lorenzo Capitani, Awen Gallimore, James A. Davies, and Stephanie E A Burnell
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Reviews ,Review ,CD8-Positive T-Lymphocytes ,Biology ,Immunotherapy, Adoptive ,model systems ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,In vivo ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,antibodies ,Immunology and Allergy ,Virotherapy ,Oncolytic Virotherapy ,CAR T cells ,Immunogenicity ,Immunotherapy ,Combined Modality Therapy ,Rats ,Oncolytic virus ,Disease Models, Animal ,Oncolytic Viruses ,030104 developmental biology ,Cancer research ,Immunogenic cell death ,Immunization ,immunotherapy ,virotherapy ,Ex vivo ,030215 immunology - Abstract
Oncolytic viruses possess the ability to infect, replicate and lyse malignantly transformed tumour cells. This oncolytic activity amplifies the therapeutic advantage and induces a form of immunogenic cell death, characterized by increased CD8 + T‐cell infiltration into the tumour microenvironment. This important feature of oncolytic viruses can result in the warming up of immunologically ‘cold’ tumour types, presenting the enticing possibility that oncolytic virus treatment combined with immunotherapies may enhance efficacy. In this review, we assess some of the most promising candidates that might be used for oncolytic virotherapy: immunotherapy combinations. We assess their potential as separate agents or as agents combined into a single therapy, where the immunotherapy is encoded within the genome of the oncolytic virus. The development of such advanced agents will require increasingly sophisticated model systems for their preclinical assessment and evaluation. In vivo rodent model systems are fraught with limitations in this regard. Oncolytic viruses replicate selectively within human cells and therefore require human xenografts in immune‐deficient mice for their evaluation. However, the use of immune‐deficient rodent models hinders the ability to study immune responses against any immunomodulatory transgenes engineered within the viral genome and expressed within the tumour microenvironment. There has therefore been a shift towards the use of more sophisticated ex vivo patient‐derived model systems based on organoids and explant co‐cultures with immune cells, which may be more predictive of efficacy than contrived and artificial animal models. We review the best of those model systems here., Oncolytic virotherapies have immense potential to ‘heat up’ otherwise immunologically ‘cold’ tumour types. In this review, the capacity of virotherapies to synergize with or express a range of types of immunotherapies is explored. Optimizing such combinations in preclinical models is complex, and we therefore review the most clinically relevant rodent and ex vivo human models available for such assessment.
- Published
- 2021
40. Whole blood-based measurement of SARS-CoV-2-specific T cell responses reveals asymptomatic infection and vaccine efficacy in healthy subjects and patients with solid organ cancers
- Author
-
Thomas Tozer, Kate Davies, Lucy C. Fairclough, Martin J. Scurr, Michelle Somerville, Kerry Roberts, Andrew James Godkin, Mererid Evans, George Lippiatt, Andrew D. Westwell, Tara Rees, Mark R. Wills, Helen Lawton, Stephanie E A Burnell, Awen Gallimore, Amanda Jackson, Wioleta M. Zelek, Lorenzo Capitani, Charlotte Aimee Young, and B. Paul Morgan
- Subjects
education.field_of_study ,biology ,business.industry ,T cell ,Population ,Vaccine efficacy ,Vaccination ,Immune system ,medicine.anatomical_structure ,Antigen ,Immunity ,Immunology ,biology.protein ,medicine ,Antibody ,education ,business - Abstract
Accurate assessment of SARS-CoV-2 immunity in the population is critical to evaluating vaccine efficacy and devising public health policies. Whilst the exact nature of effective immunity remains incompletely defined, SARS-CoV-2-specific T cell responses are a critical feature of the immune response that will likely form a key correlate of protection against COVID-19. Here, we developed and optimised a high-throughput whole blood-based assay to determine the T cell response associated with prior SARS-CoV-2 infection and/or vaccination amongst 156 healthy donors and 67 cancer patients. Following overnight in vitro stimulation with SARS-CoV-2-specific peptides, blood plasma samples were harvested and analysed for TH1-type effector cytokines (IFN-γ and IL-2). Amongst healthy donors, highly significant differential IFN-γ+/IL-2+ SARS-CoV-2-specific T cell responses were seen amongst vaccinated or previously infected COVID-19-positive individuals in comparison to unknown/naïve individuals (P < 0.0001). IL-2 production from T cells in response to SARS-CoV-2 derived antigens was a highly predictive diagnostic assay (P < 0.0001; 96.0% sensitivity, 93.9% specificity); measurement of IFN-γ+ SARS-CoV-2 specific T cell responses was equally effective at identifying asymptomatic (antibody and T cell positive) participants. A single dose of COVID-19 vaccine induced IFN-γ and/or IL-2 SARS-CoV-2-specific T cell responses in 28/29 (96.6%) of healthy donors, reducing significantly to 27/56 (48.2%) when measured in cancer patients (P = 0.0003). Overall, this cost-effective standardisable test ensures accurate and comparable assessments of SARS-CoV-2-specific T cell responses amenable to widespread population immunity testing.
- Published
- 2021
- Full Text
- View/download PDF
41. Using methylcholanthrene-induced fibrosarcomas to study tumor immunology
- Author
-
Kathryn Smart, Andrew James Godkin, Stefan Milutinovic, Sarah Nicol Lauder, Awen Gallimore, and Ana Pires
- Subjects
0303 health sciences ,Tumor microenvironment ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,Immune system ,chemistry ,Stroma ,Methylcholanthrene ,medicine ,Cancer research ,Carcinogenesis ,Tumor immunology ,Carcinogen ,030304 developmental biology - Abstract
Mouse models of cancer are essential in furthering our understanding both of the mechanisms that drive tumor development and the immune response that develops in parallel, and also in providing a platform for testing novel anti-cancer therapies. The majority of solid tumor models available rely on the injection of existing cancer cell lines into naive hosts which, while providing quick and reproducible model systems, typically lack the development of a tumor microenvironment that recapitulates those seen in human cancers. Administration of the carcinogen 3-methylcholanthrene (MCA), allows tumors to develop in situ, forming a tumor microenvironment with an established stroma and vasculature. This article provides a detailed set of protocols for the administration of MCA into mice and the subsequent monitoring of tumors. Protocols are also provided for some of the routinely used downstream applications that can be used for MCA tumors.
- Published
- 2021
- Full Text
- View/download PDF
42. Neutrophilia, lymphopenia and myeloid dysfunction: a living review of the quantitative changes to innate and adaptive immune cells which define COVID-19 pathology
- Author
-
Tehmina Bharuchq, Bruce MacLachlan, Ceri Fielding, Adrian L Smith, Fadi Issa, Emily Thornton, Raphael Sanches Peres, Calliope A Dendrou, Dorothée L Berthold, Liliana Cifuentes, Alicia Teijeira Crespo, Elizabeth H Mann, Arvind Sami, Anne Chauveau, Dominic S Alonzi, Anís Gammage, Fabian Fischer, Valentina M T Bart, Fangfang Lu, Miriam O'Hanlon, Lynn B Dustin, Pragati Sabberwal, Ewoud B. Compeer, Mariana Borsa, Rowie Borst, Hannah Almuttaqi, David J Ahern, Eleanor J Pring, Emma Jones, Ester Gea-Mallorquí, Kate Liddiard, Dingxi Zhou, Amy Cross, Sara Danielli, Max Quastel, Shayda Maleki-Toyserkani, Owen R Moon, Ruban Rex Peter Durairaj, Vicky Batchelor, Barbora Schonfeldova, Angus K T Wann, Lorenzo Capitani, Petros Ligoxygakis, Sandra Dimonte, Clara Eléonore Pavillet, Lion F K Uhl, Luke C Davies, Kristin Ladell, Stephanie Jean Hanna, Felix Richter, Freya R Shepherd, Sarah Hulin-Curtis, Stephanie Burnell, Amy Susan Codd, Anita Milicic, Quentin Sattentau, Juliane Brun, David Oliver Scourfield, Sophie Reed, Jan Rehwinkel, Ghada Alsaleh, D Oliver Scourfield, Michael Tellier, Reginald James Matthews, Eleanor Pring, Emma Mitchell, Anna Katharina Simon, Joseph D Wilson, Niamh Richmond, Van Dien Nguyen, Sarah N Lauder, Zihan Zhu, Tharini A Selvakumar, Ana Pires, Cariad Shorten, Richard Williams, Erinke van Grinsven, Sarah Galloway, Aljawharah Alrubayyi, Stephanie J. Hanna, Felix Clemens Richter, Julie M Mazet, Dimitra Peppa, Clarissa Coveney, Awen Gallimore, Cornelia Heuberger, Owen Moon, Ruth Jones, Helene Borrmann, Arthur Dyer, Lucy Chapman, Jelena S Bezbradica, Andrew Godkin, Rebecca Bayliss, Amy S. Codd, Anna M Marzeda, Athena Cavounidis, Patrícia R S Rodrigues, Alice J B Robinson, and Ewoud Bernardus Compeer
- Subjects
Myeloid ,Coronavirus disease 2019 (COVID-19) ,severity ,Review Article ,clinical ,AcademicSubjects/MED00160 ,cell counts ,recovery ,Immune system ,neutrophils ,Lymphopenia ,neutrophilia ,Medicine ,Lymphocytes ,B cells ,SARS-CoV-2 ,business.industry ,General Medicine ,Neutrophilia ,medicine.anatomical_structure ,Immunology ,AcademicSubjects/SCI00960 ,prognosis ,AcademicSubjects/MED00770 ,medicine.symptom ,monocytes ,business ,AcademicSubjects/MED00690 - Abstract
Destabilization of balanced immune cell numbers and frequencies is a common feature of viral infections. This occurs due to, and further enhances, viral immune evasion and survival. Since the discovery of the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), which manifests in coronavirus disease 2019 (COVID-19), a great number of studies have described the association between this virus and pathologically increased or decreased immune cell counts. In this review, we consider the absolute and relative changes to innate and adaptive immune cell numbers, in COVID-19. In severe disease particularly, neutrophils are increased, which can lead to inflammation and tissue damage. Dysregulation of other granulocytes, basophils and eosinophils represents an unusual COVID-19 phenomenon. Contrastingly, the impact on the different types of monocytes leans more strongly to an altered phenotype, e.g. HLA-DR expression, rather than numerical changes. However, it is the adaptive immune response that bears the most profound impact of SARS-CoV-2 infection. T cell lymphopenia correlates with increased risk of intensive care unit admission and death; therefore, this parameter is particularly important for clinical decision-making. Mild and severe diseases differ in the rate of immune cell counts returning to normal levels post disease. Tracking the recovery trajectories of various immune cell counts may also have implications for long-term COVID-19 monitoring. This review represents a snapshot of our current knowledge, showing that much has been achieved in a short period of time. Alterations in counts of distinct immune cells represent an accessible metric to inform patient care decisions or predict disease outcomes.
- Published
- 2021
- Full Text
- View/download PDF
43. Immune Remodelling of the Extracellular Matrix Drives Loss of Cancer Stem Cells and Tumor Rejection
- Author
-
Emma Jones, Rhiannon French, William J. Watkins, Matthew J. Smalley, Alexander Greenshields-Watson, James P. Hindley, Awen Gallimore, Sarah N. Lauder, Michelle Somerville, Robert Andrews, Andrew James Godkin, Kathryn Smart, Stefan Milutinovic, Ana Pires, and Howard Kendrick
- Subjects
0301 basic medicine ,Cancer Research ,Immunology ,Biology ,T-Lymphocytes, Regulatory ,Article ,Extracellular matrix ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Cancer stem cell ,Cell Line, Tumor ,Neoplasms ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Tumor microenvironment ,Mice, Inbred BALB C ,medicine.disease ,Acquired immune system ,Extracellular Matrix ,030104 developmental biology ,medicine.anatomical_structure ,Lymphatic system ,030220 oncology & carcinogenesis ,Cancer research ,Neoplastic Stem Cells ,Female ,Infiltration (medical) ,Blood vessel - Abstract
The nature of the tumor microenvironment (TME) influences the ability of tumor-specific T cells to control tumor growth. In this study, we performed an unbiased comparison of the TME of regulatory T-cell (Treg)–replete and Treg-depleted carcinogen-induced tumors, including Treg-depleted responding (regressing) and non-responding (growing) tumors. This analysis revealed an inverse relationship between extracellular matrix (ECM) and T-cell infiltrates where responding tumors were T-cell rich and ECM poor, whereas the converse was observed in non-responder tumors. For this reason, we hypothesized that the ECM acted as a barrier to successful T-cell infiltration and tumor rejection. However, further experiments revealed that this was not the case but instead showed that an effective T-cell response dramatically altered the density of ECM in the TME. Along with loss of ECM and high numbers of infiltrating T cells, responder tumors were distinguished by the development of lymphatic and blood vessel networks with specialized immune function. ECM-rich tumors exhibited a stem cell–like gene expression profile and superior tumor-initiating capacity, whereas such features were absent in responder tumors. Overall, these findings define an extended role for an effective immune response, not just in direct killing of tumor cells but in widescale remodeling of the TME to favor loss of ECM, elimination of cancer stem cells, and propagation of adaptive immunity.
- Published
- 2020
44. Enhanced antitumor immunity through sequential targeting of PI3Kδ and LAG3
- Author
-
Andrew James Godkin, Paul Kinchesh, Jake Scott, Emma Jones, Sarah Nicol Lauder, Elena Lopez-Guadamillas, Stefan Milutinovic, Veerle Kersemans, Martin J. Scurr, LS Friedman, Danny Allen, Kathryn Smart, Bart Vanhaesebroeck, Michelle Somerville, Awen Gallimore, Ana Pires, Sean Smart, and E. Hughes
- Subjects
0301 basic medicine ,lymphocytes ,Cancer Research ,LAG3 ,Class I Phosphatidylinositol 3-Kinases ,medicine.medical_treatment ,T cell ,Immunology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigens, CD ,Neoplasms ,medicine ,Tumor Microenvironment ,Immunology and Allergy ,Animals ,Humans ,Receptor ,RC254-282 ,T-lymphocytes ,Pharmacology ,Tumor microenvironment ,biology ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,Basic Tumor Immunology ,Immunotherapy ,tumor-infiltrating ,medicine.disease ,Lymphocyte Activation Gene 3 Protein ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Molecular Medicine ,Female ,Antibody ,business ,CD8 - Abstract
BackgroundDespite striking successes, immunotherapies aimed at increasing cancer-specific T cell responses are unsuccessful in most patients with cancer. Inactivating regulatory T cells (Treg) by inhibiting the PI3Kδ signaling enzyme has shown promise in preclinical models of tumor immunity and is currently being tested in early phase clinical trials in solid tumors.MethodsMice bearing 4T1 mammary tumors were orally administered a PI3Kδ inhibitor (PI-3065) daily and tumor growth, survival and T cell infiltrate were analyzed in the tumor microenvironment. A second treatment schedule comprised PI3Kδ inhibitor with anti-LAG3 antibodies administered sequentially 10 days later.ResultsAs observed in human immunotherapy trials with other agents, immunomodulation by PI3Kδ-blockade led to 4T1 tumor regressor and non-regressor mice. Tumor infiltrating T cells in regressors were metabolically fitter than those in non-regressors, with significant enrichments of antigen-specific CD8+T cells, T cell factor 1 (TCF1)+T cells and CD69−T cells, compatible with induction of a sustained tumor-specific T cell response. Treg numbers were significantly reduced in both regressor and non-regressor tumors compared with untreated tumors. The remaining Treg in non-regressor tumors were however significantly enriched with cells expressing the coinhibitory receptor LAG3, compared with Treg in regressor and untreated tumors. This striking difference prompted us to sequentially block PI3Kδ and LAG3. This combination enabled successful therapy of all mice, demonstrating the functional importance of LAG3 in non-regression of tumors on PI3Kδ inhibition therapy. Follow-up studies, performed using additional cancer cell lines, namely MC38 and CT26, indicated that a partial initial response to PI3Kδ inhibition is an essential prerequisite to a sequential therapeutic benefit of anti-LAG3 antibodies.ConclusionsThese data indicate that LAG3 is a key bottleneck to successful PI3Kδ-targeted immunotherapy and provide a rationale for combining PI3Kδ/LAG3 blockade in future clinical studies.
- Published
- 2020
45. CD4 + T cells recognize conserved influenza A epitopes through shared patterns of V-Gene usage and complementary biochemical features
- Author
-
Robert Andrews, Sarah Nicol Lauder, Aaron Wall, Kathryn Smart, Bruce J. MacLachlan, Meriem Attaf, Pierre J. Rizkallah, Dhanasekaran Vijaykrishna, Yuan Chen, Georgina H. Mason, Andrew James Godkin, Garry Dolton, Mikhail Shugay, David K. Cole, Angharad Lloyd, Cristina Rius, Andrew K. Sewell, Miguel L. Grau, Andrea J. A. Schauenburg, Alexander Greenshields-Watson, Hywel Hughes, Kathryn E. Strange, Awen Gallimore, James Geary, Sarah Hulin-Curtis, and Thomas Whalley
- Subjects
0301 basic medicine ,T cell ,CD4 T cells ,chemical and pharmacologic phenomena ,Human leukocyte antigen ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Epitope ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Influenza A virus ,Gene ,lcsh:QH301-705.5 ,Genetics ,T-cell receptor ,peptide epitopes ,030104 developmental biology ,medicine.anatomical_structure ,HLA class II ,lcsh:Biology (General) ,pHLA mutlimer ,T cell receptor ,influenza ,030217 neurology & neurosurgery ,CD8 - Abstract
Summary: T cell recognition of peptides presented by human leukocyte antigens (HLAs) is mediated by the highly variable T cell receptor (TCR). Despite this built-in TCR variability, individuals can mount immune responses against viral epitopes by using identical or highly related TCRs expressed on CD8+ T cells. Characterization of these TCRs has extended our understanding of the molecular mechanisms that govern the recognition of peptide-HLA. However, few examples exist for CD4+ T cells. Here, we investigate CD4+ T cell responses to the internal proteins of the influenza A virus that correlate with protective immunity. We identify five internal epitopes that are commonly recognized by CD4+ T cells in five HLA-DR1+ subjects and show conservation across viral strains and zoonotic reservoirs. TCR repertoire analysis demonstrates several shared gene usage biases underpinned by complementary biochemical features evident in a structural comparison. These epitopes are attractive targets for vaccination and other T cell therapies.
- Published
- 2020
46. Treg Depletion Licenses T Cell–Driven HEV Neogenesis and Promotes Tumor Destruction
- Author
-
James P. Hindley, Ralph Schulz, Lee Parry, Emily J. Colbeck, Emma Jones, Scott Cutting, Awen Gallimore, Andrew James Godkin, Anwen Sian Williams, Ann Ager, Kathryn Smart, Carl F. Ware, and Molly Browne
- Subjects
0301 basic medicine ,Cancer Research ,T cell ,Immunology ,High endothelial venules ,Biology ,Lymphotoxin beta ,T-Lymphocytes, Regulatory ,Lymphocyte Depletion ,Receptors, Tumor Necrosis Factor ,Article ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Lymphotoxin beta Receptor ,Neoplasms ,medicine ,Animals ,Tumor microenvironment ,FOXP3 ,Dendritic Cells ,R1 ,030104 developmental biology ,medicine.anatomical_structure ,Endothelium, Vascular ,CD8 ,Methylcholanthrene ,030215 immunology ,CCL21 - Abstract
T-cell infiltration into tumors represents a critical bottleneck for immune-mediated control of cancer. We previously showed that this bottleneck can be overcome by depleting immunosuppressive Foxp3+ regulatory T cells (Tregs), a process that can increase frequencies of tumor-infiltrating lymphocytes through promoting the development of specialized portals for lymphocyte entry, namely high endothelial venules (HEVs). In this paper, we used a carcinogen-induced tumor model that allows for coevolution of the tumor microenvironment and the immune response to demonstrate that Treg depletion not only results in widespread disruption to HEV networks in lymph nodes (LNs) but also activates CD8+ T cells, which then drive intratumoral HEV development. Formation of these vessels contrasts with ontogenic HEV development in LNs in that the process is dependent on the TNF receptor and independent of lymphotoxin β receptor–mediated signaling. These intratumoral HEVs do not express the chemokine CCL21, revealing a previously undescribed intratumoral blood vessel phenotype. We propose a model where Treg depletion enables a self-amplifying loop of T-cell activation, which promotes HEV development, T-cell infiltration, and ultimately, tumor destruction. The findings point to a need to test for HEV development as part of ongoing clinical studies in patients with cancer. Cancer Immunol Res; 5(11); 1005–15. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
47. Human leukocyte antigen (HLA) class II peptide flanking residues tune the immunogenicity of a human tumor-derived epitope
- Author
-
Andrea J. A. Schauenburg, Matthieu Besneux, Bruce J. MacLachlan, David K. Cole, Barbara Szomolay, Tim Elliott, Awen Gallimore, Garry Dolton, Andrew K. Sewell, Alexander Greenshields-Watson, Athanasios Papakyriakou, Pierre J. Rizkallah, Andrew James Godkin, and Georgina H. Mason
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Antigen presentation ,Immunology ,Context (language use) ,Human leukocyte antigen ,peptide flanking residues ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,Biochemistry ,T-cell biology ,Epitope ,structure-function ,Protein Structure, Secondary ,03 medical and health sciences ,Epitopes ,Antigen ,Humans ,tumor immunology ,Amino Acid Sequence ,crystallography ,Molecular Biology ,Binding Sites ,Membrane Glycoproteins ,030102 biochemistry & molecular biology ,Chemistry ,Immunogenicity ,T-cell receptor ,HLA-DR1 Antigen ,Cell Biology ,Molecular biology ,Tumor antigen ,molecular dynamics ,antigen recognition ,Protein Structure, Tertiary ,antigen presentation ,030104 developmental biology ,Colorectal Neoplasms ,Peptides ,Protein Binding - Abstract
CD4+ T-cells recognize peptide antigens, in the context of human leukocyte antigen (HLA) class II molecules (HLA-II), which through peptide-flanking residues (PFRs) can extend beyond the limits of the HLA binding. The role of the PFRs during antigen recognition is not fully understood; however, recent studies have indicated that these regions can influence T-cell receptor (TCR) affinity and pHLA-II stability. Here, using various biochemical approaches including peptide sensitivity ELISA and ELISpot assays, peptide-binding assays and HLA-II tetramer staining, we focused on CD4+ T-cell responses against a tumor antigen, 5T4 oncofetal trophoblast glycoprotein (5T4), which have been associated with improved control of colorectal cancer. Despite their weak TCR-binding affinity, we found that anti-5T4 CD4+ T-cells are polyfunctional and that their PFRs are essential for TCR recognition of the core bound nonamer. The high-resolution (1.95 Å) crystal structure of HLA-DR1 presenting the immunodominant 20-mer peptide 5T4111-130, combined with molecular dynamic simulations, revealed how PFRs explore the HLA-proximal space to contribute to antigen reactivity. These findings advance our understanding of what constitutes an HLA-II epitope and indicate that PFRs can tune weak affinity TCR-pHLA-II interactions.
- Published
- 2019
48. Cancer Antigen Discovery Is Enabled by RNA Sequencing of Highly Purified Malignant and Nonmalignant Cells
- Author
-
Yuan Chen, Stephanie E A Burnell, Michael M. Davies, Rachel Hargest, Adam Christian, Andrew James Godkin, Emma Campbell, Robert Andrews, Simon Phillips, Awen Gallimore, Sarah Hulin-Curtis, Alan L. Parker, Michelle Somerville, Alexander Greenshields-Watson, James A. Davies, Martin J. Scurr, Kevin E. Ashelford, and Richard J. Stanton
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,Cancer Research ,Stromal cell ,T cell ,T-Lymphocytes ,Antineoplastic Agents ,Biology ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,Neoplasms ,medicine ,Tumor Cells, Cultured ,Humans ,Sequence Analysis, RNA ,Immunogenicity ,Gene Expression Profiling ,Cancer ,High-Throughput Nucleotide Sequencing ,medicine.disease ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Leukocytes, Mononuclear ,Immunohistochemistry - Abstract
Purpose: Broadly expressed, highly differentiated tumor-associated antigens (TAA) can elicit antitumor immunity. However, vaccines targeting TAAs have demonstrated disappointing clinical results, reflecting poor antigen selection and/or immunosuppressive mechanisms. Experimental Design: Here, a panel of widely expressed, novel colorectal TAAs were identified by performing RNA sequencing of highly purified colorectal tumor cells in comparison with patient-matched colonic epithelial cells; tumor cell purification was essential to reveal these genes. Candidate TAA protein expression was confirmed by IHC, and preexisting T-cell immunogenicity toward these antigens tested. Results: The most promising candidate for further development is DNAJB7 [DnaJ heat shock protein family (Hsp40) member B7], identified here as a novel cancer-testis antigen. It is expressed in many tumors and is strongly immunogenic in patients with cancers originating from a variety of sites. DNAJB7-specific T cells were capable of killing colorectal tumor lines in vitro, and the IFNγ+ response was markedly magnified by control of immunosuppression with cyclophosphamide in patients with cancer. Conclusions: This study highlights how prior methods that sequence whole tumor fractions (i.e., inclusive of alive/dead stromal cells) for antigen identification may have limitations. Through tumor cell purification and sequencing, novel candidate TAAs have been identified for future immunotherapeutic targeting.
- Published
- 2019
49. Regulatory T cells in cancer: where are we now?
- Author
-
Sergio A. Quezada, Rahul Roychoudhuri, and Awen Gallimore
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Disease ,Treg cell ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,Review Series: Tregs in Cancer: Where are we now? ,Neoplasms ,Immunology and Allergy ,Medicine ,Animals ,Humans ,business.industry ,Neoplasms therapy ,Cancer ,Immunosuppression ,medicine.disease ,3. Good health ,030104 developmental biology ,Systemic toxicity ,Phenotype ,Tumor Escape ,Immunotherapy ,business ,Neuroscience ,Patient stratification ,030215 immunology ,Signal Transduction - Abstract
There have been substantial strides forward in our understanding of the contribution of regulatory T (Treg) cells to cancer immunosuppression. In this issue, we present a series of papers highlighting emerging themes on this topic relevant not only to our understanding of the fundamental biology of tumour immunosuppression but also to the design of new immunotherapeutic approaches. The substantially shared biology of CD4(+) conventional T (Tconv) and Treg cells necessitates a detailed understanding of the potentially opposing functional consequences that immunotherapies will have on Treg and Tconv cells, a prominent example being the potential for Treg‐mediated hyperprogressive disease following anti‐PD‐1 therapy. Such understanding will aid patient stratification and the rational design of combination therapies. It is also becoming clear, however, that Treg cells within tumours exhibit distinct biological features to both Tconv cells and Treg cells in other tissues. These distinct features provide the opportunity for development of targeted immunotherapies with greater efficacy and reduced potential for inducing systemic toxicity.
- Published
- 2019
50. The nature of the human T cell response to the cancer antigen 5T4 is determined by the balance of regulatory and inflammatory T cells of the same antigen-specificity: implications for vaccine design
- Author
-
Simon Phillips, Martin J. Scurr, Bruce J. MacLachlan, Matthieu Besneux, Rachel Hargest, Awen Gallimore, Adam Christian, Michael M. Davies, Alexander Greenshields-Watson, and Andrew James Godkin
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Cancer Research ,Cyclophosphamide ,Colorectal cancer ,T cell ,T-Lymphocytes ,Immunology ,T cells ,5T4 ,Context (language use) ,Biology ,Antigen specificity ,Cancer Vaccines ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,Epitopes ,Interferon-gamma ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,medicine ,Immunology and Allergy ,Humans ,Cells, Cultured ,Cancer ,Immunogenicity ,HLA-DR Antigens ,Regulatory T cells ,Th1 Cells ,medicine.disease ,Vaccination ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Drug Design ,Original Article ,Colorectal Neoplasms ,Peptides ,medicine.drug - Abstract
The oncofoetal antigen 5T4 is a promising T cell target in the context of colorectal cancer, as demonstrated by a recent clinical study where 5T4-specific T cell responses, induced by vaccination or cyclophosphamide, were associated with a significantly prolonged survival of patients with metastatic disease. Whilst Th1-type (IFN-γ+) responses specific to 5T4, and other oncofoetal antigens, are often readily detectable in early stage CRC patients and healthy donors, their activity is suppressed as the cancer progresses by CD4+CD25hiFoxp3+ regulatory T cells (Treg) which contribute to the immunosuppressive environment conducive to tumour growth. This study mapped the fine specificity of Th1 and Treg cell responses to the 5T4 protein. Surprisingly, both immunogenic peptides and those recognised by Tregs clustered in the same HLA-DR transcending epitope-rich hotspots within the 5T4 protein. Similarly, regions of low Th1-cell immunogenicity also did not contain peptides capable of stimulating Tregs, further supporting the notion that Treg and Th1 cells recognise the same peptides. Understanding the rules which govern the balance of Th1 and Treg cells responding to a given peptide specificity is, therefore, of fundamental importance to designing strategies for manipulating the balance in favour of Th1 cells, and thus the most effective anti-cancer T cell responses. Electronic supplementary material The online version of this article (10.1007/s00262-018-2266-1) contains supplementary material, which is available to authorized users.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.