18 results on '"Angela L. Ferguson"'
Search Results
2. Late-stage MC38 tumours recapitulate features of human colorectal cancer – implications for appropriate timepoint selection in preclinical studies
- Author
-
Nicholas J. Shields, Estelle M. Peyroux, Angela L. Ferguson, Megan Steain, Silke Neumann, and Sarah L. Young
- Subjects
colorectal cancer ,immunotherapy ,syngeneic preclinical models ,MC38 ,immune exclusion ,t cell exhaustion ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Anti-tumour T cell responses play a crucial role in controlling the progression of colorectal cancer (CRC), making this disease a promising candidate for immunotherapy. However, responses to immune-targeted therapies are currently limited to subpopulations of patients and specific types of cancer. Clinical studies have therefore focussed on identifying biomarkers that predict immunotherapy responses and elucidating the immunological landscapes of different cancers. Meanwhile, our understanding of how preclinical tumour models resemble human disease has fallen behind, despite their crucial role in immune-targeted drug development. A deeper understanding of these models is therefore needed to improve the development of immunotherapies and the translation of findings made in these systems. MC38 colon adenocarcinoma is a widely used preclinical model, yet how it recapitulates human colorectal cancer remains poorly defined. This study investigated the tumour-T cell immune landscape of MC38 tumours using histology, immunohistochemistry, and flow cytometry. We demonstrate that early-stage tumours exhibit a nascent TME, lacking important immune-resistance mechanisms of clinical interest, while late-stage tumours exhibit a mature TME resembling human tumours, with desmoplasia, T cell exhaustion, and T cell exclusion. Consequently, these findings clarify appropriate timepoint selection in the MC38 model when investigating both immunotherapies and mechanisms that contribute to immunotherapy resistance. Overall, this study provides a valuable resource that will enable appropriate application of the MC38 model and expedite the development and clinical translation of new immunotherapies.
- Published
- 2023
- Full Text
- View/download PDF
3. Exposure to solar ultraviolet radiation establishes a novel immune suppressive lipidome in skin-draining lymph nodes
- Author
-
Benita C. Y. Tse, Angela L. Ferguson, Yen Chin Koay, Georges E. Grau, Anthony S. Don, and Scott N. Byrne
- Subjects
immune regulation ,immune suppression ,lipids ,ultraviolet radiation ,mass spectrometry ,extracellular vesicles ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The ability of ultraviolet radiation to suppress the immune system is thought to be central to both its beneficial (protection from autoimmunity) and detrimental (carcinogenic) effects. Previous work revealed a key role for lipids particularly platelet-activating factor and sphingosine-1-phosphate in mediating UV-induced immune suppression. We therefore hypothesized that there may be other UV-induced lipids that have immune regulatory roles. To assess this, mice were exposed to an immune suppressive dose of solar-simulated UV (8 J/cm2). Lipidomic analysis identified 6 lipids (2 acylcarnitines, 2 neutral lipids, and 2 phospholipids) with significantly increased levels in the skin-draining lymph nodes of UV-irradiated mice. Imaging mass spectrometry of the lipids in combination with imaging mass cytometry identification of lymph node cell subsets indicated a preferential location of UV-induced lipids to T cell areas. In vitro co-culture of skin-draining lymph node lipids with lymphocytes showed that lipids derived from UV-exposed mice have no effect on T cell activation but significantly inhibited T cell proliferation, indicating that the lipids play an immune regulatory role. These studies are important first steps in identifying novel lipids that contribute to UV-mediated immune suppression.
- Published
- 2023
- Full Text
- View/download PDF
4. A single dose, BCG-adjuvanted COVID-19 vaccine provides sterilising immunity against SARS-CoV-2 infection
- Author
-
Claudio Counoupas, Matt D. Johansen, Alberto O. Stella, Duc H. Nguyen, Angela L. Ferguson, Anupriya Aggarwal, Nayan D. Bhattacharyya, Alice Grey, Owen Hutchings, Karishma Patel, Rezwan Siddiquee, Erica L. Stewart, Carl G. Feng, Nicole G. Hansbro, Umaimainthan Palendira, Megan C. Steain, Bernadette M. Saunders, Jason K. K. Low, Joel P. Mackay, Anthony D. Kelleher, Warwick J. Britton, Stuart G. Turville, Philip M. Hansbro, and James A. Triccas
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Global control of COVID-19 requires broadly accessible vaccines that are effective against SARS-CoV-2 variants. In this report, we exploit the immunostimulatory properties of bacille Calmette-Guérin (BCG), the existing tuberculosis vaccine, to deliver a vaccination regimen with potent SARS-CoV-2-specific protective immunity. Combination of BCG with a stabilised, trimeric form of SARS-CoV-2 spike antigen promoted rapid development of virus-specific IgG antibodies in the blood of vaccinated mice, that was further augmented by the addition of alum. This vaccine formulation, BCG:CoVac, induced high-titre SARS-CoV-2 neutralising antibodies (NAbs) and Th1-biased cytokine release by vaccine-specific T cells, which correlated with the early emergence of T follicular helper cells in local lymph nodes and heightened levels of antigen-specific plasma B cells after vaccination. Vaccination of K18-hACE2 mice with a single dose of BCG:CoVac almost completely abrogated disease after SARS-CoV-2 challenge, with minimal inflammation and no detectable virus in the lungs of infected animals. Boosting BCG:CoVac-primed mice with a heterologous vaccine further increased SARS-CoV-2-specific antibody responses, which effectively neutralised B.1.1.7 and B.1.351 SARS-CoV-2 variants of concern. These findings demonstrate the potential for BCG-based vaccination to protect against major SARS-CoV-2 variants circulating globally.
- Published
- 2021
- Full Text
- View/download PDF
5. Distinct pretreatment innate immune landscape and posttreatment T cell responses underlie immunotherapy-induced colitis
- Author
-
Kazi J. Nahar, Felix Marsh-Wakefield, Robert V. Rawson, Tuba N. Gide, Angela L. Ferguson, Ruth Allen, Camelia Quek, Ines Pires da Silva, Stephen Tattersal, Christopher J. Kiely, Neomal Sandanayake, Matteo S. Carlino, Geoff McCaughan, James S. Wilmott, Richard A. Scolyer, Georgina V. Long, Alexander M. Menzies, and Umaimainthan Palendira
- Subjects
Immunology ,Oncology ,Medicine - Abstract
Immune-related adverse events represent a major hurdle to the success of immunotherapy. The immunological mechanisms underlying their development and relation to antitumor responses are poorly understood. By examining both systemic and tissue-specific immune changes induced by combination anti–CTLA-4 and anti–PD-1 immunotherapy, we found distinct repertoire changes in patients who developed moderate-severe colitis, irrespective of their antitumor response to therapy. The proportion of circulating monocytes were significantly increased at baseline in patients who subsequently developed colitis compared with patients who did not develop colitis, and biopsies from patients with colitis showed monocytic infiltration of both endoscopically and histopathologically normal and inflamed regions of colon. The magnitude of systemic expansion of T cells following commencement of immunotherapy was also greater in patients who developed colitis. Importantly, we show expansion of specific T cell subsets within inflamed regions of the colon, including tissue-resident memory CD8+ T cells and Th1 CD4+ T cells in patients who developed colitis. Our data also suggest that CD8+ T cell expansion was locally induced, while Th1 cell expansion was systemic. Together, our data show that exaggerated innate and T cell responses to combination immunotherapy synergize to propel colitis in susceptible patients.
- Published
- 2022
- Full Text
- View/download PDF
6. Detailed spatial immunophenotyping of primary melanomas reveals immune cell subpopulations associated with patient outcome
- Author
-
Grace H. Attrill, Hansol Lee, Annie T. Tasker, Nurudeen A. Adegoke, Angela L. Ferguson, Ines Pires da Silva, Robyn P. M. Saw, John F. Thompson, Umaimainthan Palendira, Georgina V. Long, Peter M. Ferguson, Richard A. Scolyer, and James S. Wilmott
- Subjects
primary melanoma ,immunophenotyping ,spatial pathology ,T cell phenotypes ,Clinicopathological features ,Immunologic diseases. Allergy ,RC581-607 - Abstract
While the tumor immune microenvironment (TIME) of metastatic melanoma has been well characterized, the primary melanoma TIME is comparatively poorly understood. Additionally, although the association of tumor-infiltrating lymphocytes with primary melanoma patient outcome has been known for decades, it is not considered in the current AJCC melanoma staging system. Detailed immune phenotyping of advanced melanoma has revealed multiple immune biomarkers, including the presence of CD8+ T-cells, for predicting response to immunotherapies. However, in primary melanomas, immune biomarkers are lacking and CD8+ T-cells have yet to be extensively characterized. As recent studies combining immune features and clinicopathologic characteristics have created more accurate predictive models, this study sought to characterize the TIME of primary melanomas and identify predictors of patient outcome. We first phenotyped CD8+ T cells in fresh stage II primary melanomas using flow cytometry (n = 6), identifying a CD39+ tumor-resident CD8+ T-cell subset enriched for PD-1 expression. We then performed Opal multiplex immunohistochemistry and quantitative pathology-based immune profiling of CD8+ T-cell subsets, along with B cells, NK cells, Langerhans cells and Class I MHC expression in stage II primary melanoma specimens from patients with long-term follow-up (n = 66), comparing patients based on their recurrence status at 5 years after primary diagnosis. A CD39+CD103+PD-1- CD8+ T-cell population (P2) comprised a significantly higher proportion of intratumoral and stromal CD8+ T-cells in patients with recurrence-free survival (RFS) ≥5 years vs those with RFS
- Published
- 2022
- Full Text
- View/download PDF
7. Intratumoral CD16+ Macrophages Are Associated with Clinical Outcomes of Patients with Metastatic Melanoma Treated with Combination Anti-PD-1 and Anti-CTLA-4 Therapy
- Author
-
Hansol Lee, Angela L. Ferguson, Camelia Quek, Ismael A. Vergara, Ines Pires daSilva, Ruth Allen, Tuba Nur Gide, Jordan W. Conway, Lambros T. Koufariotis, Nicholas K. Hayward, Nicola Waddell, Matteo S. Carlino, Alexander M. Menzies, Robyn P.M. Saw, Elena Shklovskaya, Helen Rizos, Serigne Lo, Richard A. Scolyer, Georgina V. Long, Umaimainthan Palendira, and James S. Wilmott
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: This study characterizes intratumoral macrophage populations within baseline melanoma biopsies from patients with advanced melanoma who received either anti-PD-1 monotherapy or a combination with anti-CTLA-4. Particularly, FcγRIIIa (CD16)-expressing macrophage densities were investigated for associations with response and progression-free survival. Experimental Design: Patients with advanced melanoma who received either anti-PD-1 monotherapy or combination anti-PD-1 and anti-CTLA-4 were retrospectively identified. Macrophage populations were analyzed within baseline melanoma biopsies via multiplex IHC in relation to treatment outcomes. Results: Patients who responded to combination immune checkpoint inhibitor contained higher CD16+ macrophage densities than those who did not respond (196 vs. 7 cells/mm2; P = 0.0041). There was no diffidence in CD16+ macrophage densities in the PD-1 monotherapy-treated patients based on response (118 vs. 89 cells/mm2; P = 0.29). A significantly longer 3-year progression-free survival was observed in combination-treated patients with high intratumoral densities of CD16+ macrophages compared with those with low densities (87% vs. 42%, P = 0.0056, n = 40). No association was observed in anti-PD-1 monotherapy-treated patients (50% vs. 47%, P = 0.4636, n = 50). Melanoma biopsies with high densities of CD16+ macrophages contained upregulated gene expression of critical T-cell recruiting chemokines (CXCL9, CXCL10, and CXCL11). Conclusions: Our data demonstrate that tumor microenvironments enriched with CD16+ macrophages are favorable for response to combination anti-PD-1 and anti-CTLA-4 therapy but not anti-PD-1 monotherapy. These data provides a potential biomarker of response for combination immunotherapies in patients with metastatic melanoma.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure S1 from Intratumoral CD16+ Macrophages Are Associated with Clinical Outcomes of Patients with Metastatic Melanoma Treated with Combination Anti-PD-1 and Anti-CTLA-4 Therapy
- Author
-
James S. Wilmott, Umaimainthan Palendira, Georgina V. Long, Richard A. Scolyer, Serigne Lo, Helen Rizos, Elena Shklovskaya, Robyn P.M. Saw, Alexander M. Menzies, Matteo S. Carlino, Nicola Waddell, Nicholas K. Hayward, Lambros T. Koufariotis, Jordan W. Conway, Tuba Nur Gide, Ruth Allen, Ines Pires daSilva, Ismael A. Vergara, Camelia Quek, Angela L. Ferguson, and Hansol Lee
- Abstract
Cohort and dataset overview
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table S3 from Intratumoral CD16+ Macrophages Are Associated with Clinical Outcomes of Patients with Metastatic Melanoma Treated with Combination Anti-PD-1 and Anti-CTLA-4 Therapy
- Author
-
James S. Wilmott, Umaimainthan Palendira, Georgina V. Long, Richard A. Scolyer, Serigne Lo, Helen Rizos, Elena Shklovskaya, Robyn P.M. Saw, Alexander M. Menzies, Matteo S. Carlino, Nicola Waddell, Nicholas K. Hayward, Lambros T. Koufariotis, Jordan W. Conway, Tuba Nur Gide, Ruth Allen, Ines Pires daSilva, Ismael A. Vergara, Camelia Quek, Angela L. Ferguson, and Hansol Lee
- Abstract
Differential expression analysis gene list of FACS sorted CD16+ compared to CD16- macrophages from human melanoma dissociates.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure from High-Dimensional and Spatial Analysis Reveals Immune Landscape–Dependent Progression in Cutaneous Squamous Cell Carcinoma
- Author
-
Umaimainthan Palendira, Ruta Gupta, Ellis Patrick, Jonathan R. Clark, Marie Ranson, Bruce Ashford, Carsten E. Palme, Sydney Ch'ng, Tsu-Hui H. Low, Jenny H. Lee, Thomas Ye, Ruth O. Allen, Ashleigh R. Sharman, and Angela L. Ferguson
- Abstract
Supplementary Figure from High-Dimensional and Spatial Analysis Reveals Immune Landscape–Dependent Progression in Cutaneous Squamous Cell Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
11. Data from High-Dimensional and Spatial Analysis Reveals Immune Landscape–Dependent Progression in Cutaneous Squamous Cell Carcinoma
- Author
-
Umaimainthan Palendira, Ruta Gupta, Ellis Patrick, Jonathan R. Clark, Marie Ranson, Bruce Ashford, Carsten E. Palme, Sydney Ch'ng, Tsu-Hui H. Low, Jenny H. Lee, Thomas Ye, Ruth O. Allen, Ashleigh R. Sharman, and Angela L. Ferguson
- Abstract
Purpose:The tumor immune microenvironment impacts the biological behavior of the tumor, but its effect on clinical outcomes in head and neck cutaneous squamous cell carcinomas (HNcSCC) is largely unknown.Experimental Design:We compared the immune milieu of high-risk HNcSCC that never progressed to metastasis with those that metastasized using multiparameter imaging mass cytometry. The cohort included both immunosuppressed patients (IS) and patients with an absence of clinical immune-suppression (ACIS). Spatial analyses were used to identify cellular interactions that were associated with tumor behavior.Results:Nonprogressing primary HNcSCC were characterized by higher CD8+ and CD4+ T-cell responses, including numerically increased regulatory T cells. In contrast, primary lesions from HNcSCC patients who progressed were largely devoid of T cells with lower numbers of innate immune cells and increased expression of checkpoint receptors and in the metastatic lesions were characterized by an accumulation of B cells. Spatial analysis reveals multiple cellular interactions associated with nonprogressing primary tumors that were distinct in primary tumors of disease-progressing patients. Cellular regional analysis of the tumor microenvironment also shows squamous cell–enriched tumor regions associated with primary nonprogressing tumors.Conclusions:Effective responses from both CD8+ and CD4+ T cells in the tumor microenvironment are essential for immune control of primary HNcSCC. Our findings indicate that the early events that shape the immune responses in primary tumors dictate progression and disease outcomes in HNcSCC.
- Published
- 2023
- Full Text
- View/download PDF
12. Data from Intratumoral CD16+ Macrophages Are Associated with Clinical Outcomes of Patients with Metastatic Melanoma Treated with Combination Anti-PD-1 and Anti-CTLA-4 Therapy
- Author
-
James S. Wilmott, Umaimainthan Palendira, Georgina V. Long, Richard A. Scolyer, Serigne Lo, Helen Rizos, Elena Shklovskaya, Robyn P.M. Saw, Alexander M. Menzies, Matteo S. Carlino, Nicola Waddell, Nicholas K. Hayward, Lambros T. Koufariotis, Jordan W. Conway, Tuba Nur Gide, Ruth Allen, Ines Pires daSilva, Ismael A. Vergara, Camelia Quek, Angela L. Ferguson, and Hansol Lee
- Abstract
Purpose:This study characterizes intratumoral macrophage populations within baseline melanoma biopsies from patients with advanced melanoma who received either anti-PD-1 monotherapy or a combination with anti-CTLA-4. Particularly, FcγRIIIa (CD16)-expressing macrophage densities were investigated for associations with response and progression-free survival.Experimental Design:Patients with advanced melanoma who received either anti-PD-1 monotherapy or combination anti-PD-1 and anti-CTLA-4 were retrospectively identified. Macrophage populations were analyzed within baseline melanoma biopsies via multiplex IHC in relation to treatment outcomes.Results:Patients who responded to combination immune checkpoint inhibitor contained higher CD16+ macrophage densities than those who did not respond (196 vs. 7 cells/mm2; P = 0.0041). There was no diffidence in CD16+ macrophage densities in the PD-1 monotherapy-treated patients based on response (118 vs. 89 cells/mm2; P = 0.29). A significantly longer 3-year progression-free survival was observed in combination-treated patients with high intratumoral densities of CD16+ macrophages compared with those with low densities (87% vs. 42%, P = 0.0056, n = 40). No association was observed in anti-PD-1 monotherapy-treated patients (50% vs. 47%, P = 0.4636, n = 50). Melanoma biopsies with high densities of CD16+ macrophages contained upregulated gene expression of critical T-cell recruiting chemokines (CXCL9, CXCL10, and CXCL11).Conclusions:Our data demonstrate that tumor microenvironments enriched with CD16+ macrophages are favorable for response to combination anti-PD-1 and anti-CTLA-4 therapy but not anti-PD-1 monotherapy. These data provides a potential biomarker of response for combination immunotherapies in patients with metastatic melanoma.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table from High-Dimensional and Spatial Analysis Reveals Immune Landscape–Dependent Progression in Cutaneous Squamous Cell Carcinoma
- Author
-
Umaimainthan Palendira, Ruta Gupta, Ellis Patrick, Jonathan R. Clark, Marie Ranson, Bruce Ashford, Carsten E. Palme, Sydney Ch'ng, Tsu-Hui H. Low, Jenny H. Lee, Thomas Ye, Ruth O. Allen, Ashleigh R. Sharman, and Angela L. Ferguson
- Abstract
Supplementary Table from High-Dimensional and Spatial Analysis Reveals Immune Landscape–Dependent Progression in Cutaneous Squamous Cell Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
14. A unique cytotoxic CD4+T cells signature defines critical COVID-19
- Author
-
Sarah Baird, Caroline L. Ashley, Felix Marsh-Wakefield, Sibel Alca, Thomas M. Ashhurst, Angela L. Ferguson, Hannah Lukeman, Claudio Counoupas, Jeffrey J. Post, Pamela Konecny, Adam Bartlett, Marianne Martinello, Rowena A. Bull, Andrew Lloyd, Alice Grey, Owen Hutchings, Umaimainthan Palendira, Warwick J. Britton, Megan Steain, and James A. Triccas
- Abstract
Background and objectivesSARS-CoV-2 infection causes a spectrum of clinical disease presentation, ranging from asymptomatic to fatal. While neutralising antibody (NAb) responses correlate with protection against symptomatic and severe infection, the contribution of the T cell response to the resolution or progression of disease is still unclear. Optimal protective immunity may require activation of distinct immune pathways. As such, defining the contribution of individual T cell subsets to disease outcome is imperative to inform the development of next-generation COVID-19 vaccines. To address this, we performed immunophenotyping of T cell responses in unvaccinated individuals, representing the full spectrum of COVID-19 clinical presentation.MethodsSpectral cytometry was performed on peripheral blood mononuclear cell samples from patients with PCR-confirmed SARS-CoV-2 infection. Computational and manual analyses were used to identify T cell populations associated with distinct disease states through unbiased clustering, principal component analysis and discriminant analysis.ResultsCritical SARS-CoV-2 infection was characterised by an increase in activated and cytotoxic CD4+(CTL) cells of a T follicular helper (TFH) or effector memory re-expressing CD45RA (TEMRA) phenotype. These CD4+CTLs were largely absent in those with less severe disease. In contrast, those with asymptomatic or mild disease were associated with high proportions of naïve T cells and reduced expression of activation markers.ConclusionHighly activated and cytotoxic CD4+T cell responses may contribute to cell-mediated host tissue damage and progression of COVID-19. Potential for induction of these detrimental T cell responses should be considered when developing and implementing effective COVID-19 control strategies.
- Published
- 2023
- Full Text
- View/download PDF
15. High-Dimensional and Spatial Analysis Reveals Immune Landscape-Dependent Progression in Cutaneous Squamous Cell Carcinoma
- Author
-
Angela L. Ferguson, Ashleigh R. Sharman, Ruth O. Allen, Thomas Ye, Jenny H. Lee, Tsu-Hui H. Low, Sydney Ch'ng, Carsten E. Palme, Bruce Ashford, Marie Ranson, Jonathan R. Clark, Ellis Patrick, Ruta Gupta, and Umaimainthan Palendira
- Subjects
Cancer Research ,Spatial Analysis ,Skin Neoplasms ,Oncology ,Head and Neck Neoplasms ,Squamous Cell Carcinoma of Head and Neck ,Carcinoma, Squamous Cell ,Tumor Microenvironment ,Humans - Abstract
Purpose:The tumor immune microenvironment impacts the biological behavior of the tumor, but its effect on clinical outcomes in head and neck cutaneous squamous cell carcinomas (HNcSCC) is largely unknown.Experimental Design:We compared the immune milieu of high-risk HNcSCC that never progressed to metastasis with those that metastasized using multiparameter imaging mass cytometry. The cohort included both immunosuppressed patients (IS) and patients with an absence of clinical immune-suppression (ACIS). Spatial analyses were used to identify cellular interactions that were associated with tumor behavior.Results:Nonprogressing primary HNcSCC were characterized by higher CD8+ and CD4+ T-cell responses, including numerically increased regulatory T cells. In contrast, primary lesions from HNcSCC patients who progressed were largely devoid of T cells with lower numbers of innate immune cells and increased expression of checkpoint receptors and in the metastatic lesions were characterized by an accumulation of B cells. Spatial analysis reveals multiple cellular interactions associated with nonprogressing primary tumors that were distinct in primary tumors of disease-progressing patients. Cellular regional analysis of the tumor microenvironment also shows squamous cell–enriched tumor regions associated with primary nonprogressing tumors.Conclusions:Effective responses from both CD8+ and CD4+ T cells in the tumor microenvironment are essential for immune control of primary HNcSCC. Our findings indicate that the early events that shape the immune responses in primary tumors dictate progression and disease outcomes in HNcSCC.
- Published
- 2022
16. Distinct pretreatment innate immune landscape and posttreatment T cell responses underlie immunotherapy-induced colitis
- Author
-
Kazi J. Nahar, Felix Marsh-Wakefield, Robert V. Rawson, Tuba N. Gide, Angela L. Ferguson, Ruth Allen, Camelia Quek, Ines Pires da Silva, Stephen Tattersal, Christopher J. Kiely, Neomal Sandanayake, Matteo S. Carlino, Geoff McCaughan, James S. Wilmott, Richard A. Scolyer, Georgina V. Long, Alexander M. Menzies, and Umaimainthan Palendira
- Subjects
Programmed Cell Death 1 Receptor ,Humans ,Immunologic Factors ,General Medicine ,Immunotherapy ,CD8-Positive T-Lymphocytes ,Colitis ,Immunity, Innate - Abstract
Immune-related adverse events represent a major hurdle to the success of immunotherapy. The immunological mechanisms underlying their development and relation to antitumor responses are poorly understood. By examining both systemic and tissue-specific immune changes induced by combination anti-CTLA-4 and anti-PD-1 immunotherapy, we found distinct repertoire changes in patients who developed moderate-severe colitis, irrespective of their antitumor response to therapy. The proportion of circulating monocytes were significantly increased at baseline in patients who subsequently developed colitis compared with patients who did not develop colitis, and biopsies from patients with colitis showed monocytic infiltration of both endoscopically and histopathologically normal and inflamed regions of colon. The magnitude of systemic expansion of T cells following commencement of immunotherapy was also greater in patients who developed colitis. Importantly, we show expansion of specific T cell subsets within inflamed regions of the colon, including tissue-resident memory CD8+ T cells and Th1 CD4+ T cells in patients who developed colitis. Our data also suggest that CD8+ T cell expansion was locally induced, while Th1 cell expansion was systemic. Together, our data show that exaggerated innate and T cell responses to combination immunotherapy synergize to propel colitis in susceptible patients.
- Published
- 2021
17. Approaches to spatially resolving the tumour immune microenvironment of hepatocellular carcinoma
- Author
-
Felix Marsh-Wakefield, Angela L Ferguson, Ken Liu, Cositha Santhakumar, Geoffrey McCaughan, and Umaimainthan Palendira
- Subjects
Oncology - Abstract
Hepatocellular carcinoma (HCC) is a common and deadly cancer worldwide. Many factors contribute to mortality and place an individual at high risk of developing HCC, including viral infection, alcohol intake, metabolic-associated disease, autoimmunity and genetic liver disorders. Although there are many therapeutics available, much about this disease remains to be understood. This is most evident when investigating the tumour microenvironment (TME). Both innate and adaptive immune cells have been associated with carcinogenesis within the TME of HCC patients. The ability to interrogate the TME more thoroughly with spatial technologies continues to improve, both at the experimental and analytical stages. This review provides insight into technologies available to investigate the TME, and how such technologies are beneficial for improving our understanding of HCC carcinogenesis.
- Published
- 2022
- Full Text
- View/download PDF
18. A unique cytotoxic CD4+ T cell‐signature defines critical COVID‐19
- Author
-
Sarah Baird, Caroline L Ashley, Felix Marsh‐Wakefield, Sibel Alca, Thomas M Ashhurst, Angela L Ferguson, Hannah Lukeman, Claudio Counoupas, Jeffrey J Post, Pamela Konecny, Adam Bartlett, Marianne Martinello, Rowena A Bull, Andrew Lloyd, Alice Grey, Owen Hutchings, Umaimainthan Palendira, Warwick J Britton, Megan Steain, and James A Triccas
- Subjects
CD4‐CTLs ,COVID‐19 ,SARS‐CoV‐2 ,spectral cytometry ,T cells ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Abstract Objectives SARS‐CoV‐2 infection causes a spectrum of clinical disease presentation, ranging from asymptomatic to fatal. While neutralising antibody (NAb) responses correlate with protection against symptomatic and severe infection, the contribution of the T‐cell response to disease resolution or progression is still unclear. As newly emerging variants of concern have the capacity to partially escape NAb responses, defining the contribution of individual T‐cell subsets to disease outcome is imperative to inform the development of next‐generation COVID‐19 vaccines. Methods Immunophenotyping of T‐cell responses in unvaccinated individuals was performed, representing the full spectrum of COVID‐19 clinical presentation. Computational and manual analyses were used to identify T‐cell populations associated with distinct disease states. Results Critical SARS‐CoV‐2 infection was characterised by an increase in activated and cytotoxic CD4+ lymphocytes (CTL). These CD4+ CTLs were largely absent in asymptomatic to severe disease states. In contrast, non‐critical COVID‐19 was associated with high frequencies of naïve T cells and lack of activation marker expression. Conclusion Highly activated and cytotoxic CD4+ T‐cell responses may contribute to cell‐mediated host tissue damage and progression of COVID‐19. Induction of these potentially detrimental T‐cell responses should be considered when developing and implementing effective COVID‐19 control strategies.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.