195 results on '"Anita Grigoriadis"'
Search Results
2. Adipose-enriched peri-tumoral stroma, in contrast to myofibroblast-enriched stroma, prognosticates poorer survival in breast cancers
- Author
-
Hannah Si Hui Lau, Veronique Kiak Mien Tan, Benita Kiat Tee Tan, Yirong Sim, Jelmar Quist, Aye Aye Thike, Puay Hoon Tan, Shazib Pervaiz, Anita Grigoriadis, and Kanaga Sabapathy
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Despite our understanding of the genetic basis of intra-tumoral heterogeneity, the role of stromal heterogeneity arising from an altered tumor microenvironment in affecting tumorigenesis is poorly understood. In particular, extensive study on the peri-tumoral stroma in the morphologically normal tissues surrounding the tumor is lacking. Here, we examine the heterogeneity in tumors and peri-tumoral stroma from 8 ER+/PR+/HER2− invasive breast carcinomas, through multi-region transcriptomic profiling by microarray. We describe the regional heterogeneity observed at the intrinsic molecular subtype, pathway enrichment, and cell type composition levels within each tumor and its peri-tumoral region, up to 7 cm from the tumor margins. Moreover, we identify a pro-inflammatory adipose-enriched peri-tumoral subtype which was significantly associated with poorer overall survival in breast cancer patients, in contrast to an adaptive immune cell- and myofibroblast-enriched subtype. These data together suggest that peri-tumoral heterogeneity may be an important determinant of the evolution and treatment of breast cancers.
- Published
- 2023
- Full Text
- View/download PDF
3. B cell profiles, antibody repertoire and reactivity reveal dysregulated responses with autoimmune features in melanoma
- Author
-
Silvia Crescioli, Isabel Correa, Joseph Ng, Zena N. Willsmore, Roman Laddach, Alicia Chenoweth, Jitesh Chauhan, Ashley Di Meo, Alexander Stewart, Eleni Kalliolia, Elena Alberts, Rebecca Adams, Robert J. Harris, Silvia Mele, Giulia Pellizzari, Anna B. M. Black, Heather J. Bax, Anthony Cheung, Mano Nakamura, Ricarda M. Hoffmann, Manuela Terranova-Barberio, Niwa Ali, Ihor Batruch, Antoninus Soosaipillai, Ioannis Prassas, Antigona Ulndreaj, Miyo K. Chatanaka, Rosamund Nuamah, Shichina Kannambath, Pawan Dhami, Jenny L. C. Geh, Alastair D. MacKenzie Ross, Ciaran Healy, Anita Grigoriadis, David Kipling, Panagiotis Karagiannis, Deborah K. Dunn-Walters, Eleftherios P. Diamandis, Sophia Tsoka, James Spicer, Katie E. Lacy, Franca Fraternali, and Sophia N. Karagiannis
- Subjects
Science - Abstract
Abstract B cells are known to contribute to the anti-tumor immune response, especially in immunogenic tumors such as melanoma, yet humoral immunity has not been characterized in these cancers to detail. Here we show comprehensive phenotyping in samples of circulating and tumor-resident B cells as well as serum antibodies in melanoma patients. Memory B cells are enriched in tumors compared to blood in paired samples and feature distinct antibody repertoires, linked to specific isotypes. Tumor-associated B cells undergo clonal expansion, class switch recombination, somatic hypermutation and receptor revision. Compared with blood, tumor-associated B cells produce antibodies with proportionally higher levels of unproductive sequences and distinct complementarity determining region 3 properties. The observed features are signs of affinity maturation and polyreactivity and suggest an active and aberrant autoimmune-like reaction in the tumor microenvironment. Consistent with this, tumor-derived antibodies are polyreactive and characterized by autoantigen recognition. Serum antibodies show reactivity to antigens attributed to autoimmune diseases and cancer, and their levels are higher in patients with active disease compared to post-resection state. Our findings thus reveal B cell lineage dysregulation with distinct antibody repertoire and specificity, alongside clonally-expanded tumor-infiltrating B cells with autoimmune-like features, shaping the humoral immune response in melanoma.
- Published
- 2023
- Full Text
- View/download PDF
4. Inducible localized delivery of an anti-PD-1 scFv enhances anti-tumor activity of ROR1 CAR-T cells in TNBC
- Author
-
Micaela Harrasser, Satyen Harish Gohil, Hiu Lau, Marco Della Peruta, Vincent Muczynski, Dominic Patel, Elena Miranda, Kristiana Grigoriadis, Anita Grigoriadis, David Granger, Rachel Evans, and Amit Chunilal Nathwani
- Subjects
ROR1 ,CAR-T cell therapy ,PD-1 ,Checkpoint blockade ,F i-CAR-T cell ,Single-chain variable fragment (scFv) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Chimeric antigen receptor (CAR)-T cells can induce powerful immune responses in patients with hematological malignancies but have had limited success against solid tumors. This is in part due to the immunosuppressive tumor microenvironment (TME) which limits the activity of tumor-infiltrating lymphocytes (TILs) including CAR-T cells. We have developed a next-generation armored CAR (F i-CAR) targeting receptor tyrosine kinase-like orphan receptor 1 (ROR1), which is expressed at high levels in a range of aggressive tumors including poorly prognostic triple-negative breast cancer (TNBC). The F i-CAR-T is designed to release an anti-PD-1 checkpoint inhibitor upon CAR-T cell activation within the TME, facilitating activation of CAR-T cells and TILs while limiting toxicity. Methods To bolster potency, we developed a F i-CAR construct capable of IL-2-mediated, NFAT-induced secretion of anti-PD-1 single-chain variable fragments (scFv) within the tumor microenvironment, following ROR1-mediated activation. Cytotoxic responses against TNBC cell lines as well as levels and binding functionality of released payload were analyzed in vitro by ELISA and flow cytometry. In vivo assessment of potency of F i-CAR-T cells was performed in a TNBC NSG mouse model. Results F i-CAR-T cells released measurable levels of anti-PD-1 payload with 5 h of binding to ROR1 on tumor and enhanced the cytotoxic effects at challenging 1:10 E:T ratios. Treatment of established PDL1 + TNBC xenograft model with F i-CAR-T cells resulted in significant abrogation in tumor growth and improved survival of mice (71 days), compared to non-armored CAR cells targeting ROR1 (F CAR-T) alone (49 days) or in combination with systemically administered anti-PD-1 antibody (57 days). Crucially, a threefold increase in tumor-infiltrating T cells was observed with F i-CAR-T cells and was associated with increased expression of genes related to cytotoxicity, migration and proliferation. Conclusions Our next-generation of ROR1-targeting inducible armored CAR platform enables the release of an immune stimulating payload only in the presence of target tumor cells, enhancing the therapeutic activity of the CAR-T cells. This technology provided a significant survival advantage in TNBC xenograft models. This coupled with its potential safety attributes merits further clinical evaluation of this approach in TNBC patients.
- Published
- 2022
- Full Text
- View/download PDF
5. Systemic immune reaction in axillary lymph nodes adds to tumor-infiltrating lymphocytes in triple-negative breast cancer prognostication
- Author
-
Fangfang Liu, Thomas Hardiman, Kailiang Wu, Jelmar Quist, Patrycja Gazinska, Tony Ng, Arnie Purushotham, Roberto Salgado, Xiaojing Guo, Sarah E. Pinder, and Anita Grigoriadis
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract The level of stromal tumor-infiltrating lymphocytes (sTILs) in triple-negative (TNBC) and HER2-positive breast cancers convey prognostic information. The importance of systemic immunity to local immunity is unknown in breast cancer. We previously demonstrated that histological alterations in axillary lymph nodes (LNs) carry clinical relevance. Here, we capture local immune responses by scoring TILs at the primary tumor and systemic immune responses by recording the formation of secondary follicles, also known as germinal centers, in 2,857 cancer-free and involved axillary LNs on haematoxylin and eosin (H&E) stained sections from a retrospective cohort of 161 LN-positive triple-negative and HER2-positive breast cancer patients. Our data demonstrate that the number of germinal center formations across all cancer-free LNs, similar to high levels of TILs, is associated with a good prognosis in low TILs TNBC. This highlights the importance of assessing both primary and LN immune responses for prognostication and for future breast cancer research.
- Published
- 2021
- Full Text
- View/download PDF
6. Invasive breast cancer over four decades reveals persisting poor metastatic outcomes in treatment resistant subgroup – the 'ATRESS' phenomenon
- Author
-
Patriek Jurrius, Thomas Green, Hans Garmo, Matthew Young, Massimiliano Cariati, Cheryl Gillett, Anca Mera, Mark Harries, Anita Grigoriadis, Sarah Pinder, Lars Holmberg, and Arnie Purushotham
- Subjects
Breast cancer ,Post-metastatic survival ,ATRESS ,Treatment resistance ,Prognosis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Major advances in breast cancer treatment have led to a reducuction in mortality. However, there are still women who are not cured. We hypothesize there is a sub-group of women with treatment-resistant cancers causing early death. Methods: Between 1975 and 2006, 5392 women with invasive breast cancer underwent surgery at Guy’s Hospital, London. Data on patient demographics, tumour characteristics, treatment regimens, local recurrence, secondary metastasis, and death were prospectively recorded. We considered four time periods (1975–1982, 1983–1990, 1991–1998, 1999–2006). Risks and time to event analysis were performed with Cox proportional hazards model and Kaplan-Meier estimation. Results: Unadjusted hazard ratios for developing metastasis and overall mortality relative to the 1975–1982 cohort decreased steadily to 0.23 and 0.63, respectively in 1999–2006. However, metastasis-free interval shortened, with the proportion of women developing metastasis ≤5 years increasing from 73.9% to 83.0%. Furthermore, median post-metastatic survival decreased from 1.49 years to 0.94 years. Applying our risk criteria identified the presence of ±200 patients in each cohort who developed metastasis early and died within a much shorter time frame. Conclusions: Advances in treatment have decreased the risk of metastasis and improved survival in women with invasive breast cancer over the last 40 years. Despite this, a subpopulation with shorter metastasis-free and post-metastatic survival who are unresponsive to available treatment remains. This may be due to the ATRESS phenomenon (adjuvant therapy-related shortening of survival) secondary to preselection inherent in adjuvant therapy, successful treatment of less malignant tumour cells and treatment-induced resistance in the remaining tumour clones.
- Published
- 2020
- Full Text
- View/download PDF
7. Metabolic profiles to predict long-term cancer and mortality: the use of latent class analysis
- Author
-
Aida Santaolalla, Hans Garmo, Anita Grigoriadis, Sundeep Ghuman, Niklas Hammar, Ingmar Jungner, Göran Walldius, Mats Lambe, Lars Holmberg, and Mieke Van Hemelrijck
- Subjects
Risk stratification ,Biomarkers ,Metabolic profiles ,Latent class analysis ,Disease susceptibility ,Cancer epidemiology ,Cytology ,QH573-671 - Abstract
Abstract Background Metabolites are genetically and environmentally determined. Consequently, they can be used to characterize environmental exposures and reveal biochemical mechanisms that link exposure to disease. To explore disease susceptibility and improve population risk stratification, we aimed to identify metabolic profiles linked to carcinogenesis and mortality and their intrinsic associations by characterizing subgroups of individuals based on serum biomarker measurements. We included 13,615 participants from the Swedish Apolipoprotein MOrtality RISk Study who had measurements for 19 biomarkers representative of central metabolic pathways. Latent Class Analysis (LCA) was applied to characterise individuals based on their biomarker values (according to medical cut-offs), which were then examined as predictors of cancer and death using multivariable Cox proportional hazards models. Results LCA identified four metabolic profiles within the population: (1) normal values for all markers (63% of population); (2) abnormal values for lipids (22%); (3) abnormal values for liver functioning (9%); (4) abnormal values for iron and inflammation metabolism (6%). All metabolic profiles (classes 2–4) increased risk of cancer and mortality, compared to class 1 (e.g. HR for overall death was 1.26 (95% CI: 1.16–1.37), 1.67 (95% CI: 1.47–1.90), and 1.21 (95% CI: 1.05–1.41) for class 2, 3, and 4, respectively). Conclusion We present an innovative approach to risk stratify a well-defined population based on LCA metabolic-defined subgroups for cancer and mortality. Our results indicate that standard of care baseline serum markers, when assembled into meaningful metabolic profiles, could help assess long term risk of disease and provide insight in disease susceptibility and etiology.
- Published
- 2019
- Full Text
- View/download PDF
8. Immune Crosstalk Between Lymph Nodes and Breast Carcinomas, With a Focus on B Cells
- Author
-
Elena Alberts, Isobelle Wall, Dinis Pedro Calado, and Anita Grigoriadis
- Subjects
lymph node ,TILs ,tertiary lymphoid structures ,germinal centers ,breast cancer ,Biology (General) ,QH301-705.5 - Abstract
Lymph nodes (LNs) are highly organized secondary lymphoid organs, and reflective of immune responses to infection, injuries, or the presence of cancer. Extensive molecular and morphological analyses of immune and stromal features in tumors and LNs of breast cancer patients have revealed novel patterns indicative of disease progression. Within LNs, there are dynamic structures called germinal centers (GCs), that act as the immunological hubs for B cell development and generation of affinity matured memory B and antibody-producing plasma cells. Acting as a bridge between systemic and local immunity, associations are observed between the frequency of GCs within cancer-free LNs, the levels of stromal tumor infiltrating lymphocytes, and cancer progression. Scattered throughout the tumor microenvironment (TME) or aggregated in clusters forming tertiary lymphoid structures (TLS), the occurrence of tumor infiltrating B cells (TIL-Bs) has been linked mostly to superior disease trajectories in solid cancers. Recent TIL-Bs profiling studies have revealed a plethora of different TIL-B populations, their functional roles, and whether they are derived from GC reactions in the LN, and/or locally from GC-like structures within the TME remains to be investigated. However, parallels between the immunogenic nature of LNs as a pre-metastatic niche, TIL-B populations within the TME, and the presence of TLS will help to decipher local and widespread TIL-Bs responses and their influence on cancer progression to the lymphatics. Therapies that enhance TIL-Bs responses in the LN GC and/or in GC-like structures in the TME are thus emerging management strategies for breast and other cancer patients.
- Published
- 2021
- Full Text
- View/download PDF
9. Molecular patterns of cancer colonisation in lymph nodes of breast cancer patients
- Author
-
Gaurav Chatterjee, Trupti Pai, Thomas Hardiman, Kelly Avery-Kiejda, Rodney J. Scott, Jo Spencer, Sarah E. Pinder, and Anita Grigoriadis
- Subjects
Expression ,Lymph node ,Premetastatic niche ,Breast cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Lymph node (LN) metastasis is an important prognostic parameter in breast carcinoma, a crucial site for tumour–immune cell interaction and a gateway for further dissemination of tumour cells to other metastatic sites. To gain insight into the underlying molecular changes from the pre-metastatic, via initial colonisation to the fully involved LN, we reviewed transcriptional research along the evolving microenvironment of LNs in human breast cancers patients. Gene expression studies were compiled and subjected to pathway-based analyses, with an emphasis on immune cell-related genes. Of 366 studies, 14 performed genome-wide gene expression comparisons and were divided into six clinical-biological scenarios capturing different stages of the metastatic pathway in the LN, as follows: metastatically involved LNs are compared to their patient-matched primary breast carcinomas (scenario 1) or the normal breast tissue (scenario 2). In scenario 3, uninvolved LNs were compared between LN-positive patients and LN-negative patients. Scenario 4 homed in on the residual uninvolved portion of involved LNs and compared it to the patient-matched uninvolved LNs. Scenario 5 contrasted uninvolved and involved LNs, whilst in scenario 6 involved (sentinel) LNs were assessed between patients with other either positive or negative LNs (non-sentinel). Gene lists from these chronological steps of LN metastasis indicated that gene patterns reflecting deficiencies in dendritic cells and hyper-proliferation of B cells parallel to tumour promoting pathways, including cell adhesion, extracellular matrix remodelling, cell motility and DNA repair, play key roles in the changing microenvironment of a pro-metastatic to a metastatically involved LN. Similarities between uninvolved LNs and the residual uninvolved portion of involved LNs hinted that LN alterations expose systemic tumour-related immune responses in breast cancer patients. Despite the diverse settings, gene expression patterns at different stages of metastatic colonisation in LNs were recognised and may provide potential avenues for clinical interventions to counteract disease progression for breast cancer patients.
- Published
- 2018
- Full Text
- View/download PDF
10. Integrated genomics and functional validation identifies malignant cell specific dependencies in triple negative breast cancer
- Author
-
Nirmesh Patel, Daniel Weekes, Konstantinos Drosopoulos, Patrycja Gazinska, Elodie Noel, Mamun Rashid, Hasan Mirza, Jelmar Quist, Fara Brasó-Maristany, Sumi Mathew, Riccardo Ferro, Ana Mendes Pereira, Cynthia Prince, Farzana Noor, Erika Francesch-Domenech, Rebecca Marlow, Emanuele de Rinaldis, Anita Grigoriadis, Spiros Linardopoulos, Pierfrancesco Marra, and Andrew N. J. Tutt
- Subjects
Science - Abstract
Triple negative breast cancers harbor multiple copy number aberrations driving gene expression changes thought to underpin their malignant phenotypes. Here the authors integrate these features, finding and functionally validating 37 gene addictions among which they identify the mechanism of addiction to KIFC1, a potential selective drug target.
- Published
- 2018
- Full Text
- View/download PDF
11. An RNAi screen of Rho signalling networks identifies RhoH as a regulator of Rac1 in prostate cancer cell migration
- Author
-
Virginia Tajadura-Ortega, Ritu Garg, Richard Allen, Claudia Owczarek, Michael D. Bright, Samuel Kean, Aisyah Mohd-Noor, Anita Grigoriadis, Timothy C. Elston, Klaus M. Hahn, and Anne J. Ridley
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Abstract Background Cell migration is essential for development and tissue repair, but it also contributes to disease. Rho GTPases regulate cell migration, but a comprehensive analysis of how each Rho signalling component affects migration has not been carried out. Results Through an RNA interference screen, and using a prostate cancer cell line, we find that approximately 25% of Rho network components alter migration. Some genes enhance migration while others decrease basal and/or hepatocyte growth factor-stimulated migration. Surprisingly, we identify RhoH as a screen hit. RhoH expression is normally restricted to haematopoietic cells, but we find it is expressed in multiple epithelial cancer cell lines. High RhoH expression in samples from prostate cancer patients correlates with earlier relapse. RhoH depletion reduces cell speed and persistence and decreases migratory polarity. Rac1 activity normally localizes to the front of migrating cells at areas of dynamic membrane movement, but in RhoH-depleted cells active Rac1 is localised around the whole cell periphery and associated with membrane regions that are not extending or retracting. RhoH interacts with Rac1 and with several p21-activated kinases (PAKs), which are Rac effectors. Similar to RhoH depletion, PAK2 depletion increases cell spread area and reduces cell migration. In addition, RhoH depletion reduces lamellipodium extension induced by PAK2 overexpression. Conclusions We describe a novel role for RhoH in prostate cancer cell migration. We propose that RhoH promotes cell migration by coupling Rac1 activity and PAK2 to membrane protrusion. Our results also suggest that RhoH expression levels correlate with prostate cancer progression.
- Published
- 2018
- Full Text
- View/download PDF
12. Gene expression modules in primary breast cancers as risk factors for organotropic patterns of first metastatic spread: a case control study
- Author
-
Katherine Lawler, Efterpi Papouli, Cristina Naceur-Lombardelli, Anca Mera, Kayleigh Ougham, Andrew Tutt, Siker Kimbung, Ingrid Hedenfalk, Jun Zhan, Hongquan Zhang, Richard Buus, Mitch Dowsett, Tony Ng, Sarah E. Pinder, Peter Parker, Lars Holmberg, Cheryl E. Gillett, Anita Grigoriadis, and Arnie Purushotham
- Subjects
Breast cancer ,Metasynchronous metastases ,Gene expression pattern ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Metastases from primary breast cancers can involve single or multiple organs at metastatic disease diagnosis. Molecular risk factors for particular patterns of metastastic spread in a clinical population are limited. Methods A case-control design including 1357 primary breast cancers was used to study three distinct clinical patterns of metastasis, which occur within the first six months of metastatic disease: bone and visceral metasynchronous spread, bone-only, and visceral-only metastasis. Whole-genome expression profiles were obtained using whole genome (WG)-DASL assays from formalin-fixed paraffin-embedded (FFPE) samples. A systematic protocol was developed for handling FFPE samples together with stringent data quality controls to identify robust expression profiling data. A panel of published and novel gene sets were tested for association with these specific patterns of metastatic spread and odds ratios (ORs) were calculated. Results Metasynchronous metastasis to bone and viscera was found in all intrinsic breast cancer subtypes, while immunohistochemically (IHC)-defined receptor status and specific IntClust subgroups were risk factors for visceral-only or bone-only first metastases. Among gene modules, those related to proliferation increased the risk of metasynchronous metastasis (OR (95% CI) = 2.3 (1.1–4.8)) and visceral-only first metastasis (OR (95% CI) = 2.5 (1.2–5.1)) but not bone-only metastasis (OR (95% CI) = 0.97 (0.56–1.7)). A 21-gene module (BV) was identified in estrogen-receptor-positive breast cancers with metasynchronous metastasis to bone and viscera (area under the curve = 0.77), and its expression increased the risk of bone and visceral metasynchronous spread in this population. BV was further orthogonally validated with NanoString nCounter in primary breast cancers, and was reproducible in their matched lymph nodes metastases and an external cohort. Conclusion This case-control study of WG-DASL global expression profiles from FFPE tumour samples, after careful quality control and RNA selection, revealed that gene modules in the primary tumour have differing risks for clinical patterns of metasynchronous first metastases. Moreover, a novel gene module was identified as a putative risk factor for metasynchronous bone and visceral first metastatic spread, with potential implications for disease monitoring and treatment planning.
- Published
- 2017
- Full Text
- View/download PDF
13. Integrin-Mediated Macrophage Adhesion Promotes Lymphovascular Dissemination in Breast Cancer
- Author
-
Rachel Evans, Fabian Flores-Borja, Sina Nassiri, Elena Miranda, Katherine Lawler, Anita Grigoriadis, James Monypenny, Cheryl Gillet, Julie Owen, Peter Gordon, Victoria Male, Anthony Cheung, Farzana Noor, Paul Barber, Rebecca Marlow, Erika Francesch-Domenech, Gilbert Fruhwirth, Mario Squadrito, Borivoj Vojnovic, Andrew Tutt, Frederic Festy, Michele De Palma, and Tony Ng
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Lymphatic vasculature is crucial for metastasis in triple-negative breast cancer (TNBC); however, cellular and molecular drivers controlling lymphovascular metastasis are poorly understood. We define a macrophage-dependent signaling cascade that facilitates metastasis through lymphovascular remodeling. TNBC cells instigate mRNA changes in macrophages, resulting in β4 integrin-dependent adhesion to the lymphovasculature. β4 integrin retains macrophages proximal to lymphatic endothelial cells (LECs), where release of TGF-β1 drives LEC contraction via RhoA activation. Macrophages promote gross architectural changes to lymphovasculature by increasing dilation, hyperpermeability, and disorganization. TGF-β1 drives β4 integrin clustering at the macrophage plasma membrane, further promoting macrophage adhesion and demonstrating the dual functionality of TGF-β1 signaling in this context. β4 integrin-expressing macrophages were identified in human breast tumors, and a combination of vascular-remodeling macrophage gene signature and TGF-β signaling scores correlates with metastasis. We postulate that future clinical strategies for patients with TNBC should target crosstalk between β4 integrin and TGF-β1. : Breast cancer metastasis through lymphatic vessels is associated with poor prognosis. Evans et al. describe β4 integrin-expressing macrophages that regulate lymphatic vessel structure in breast cancer. Macrophage-released TGF-β1 drives lymphatic cell contraction via RhoA activation, culminating in lymphatic hyperpermeability. This study defines a signaling cascade that could be targeted therapeutically. Keywords: lymphovasculature, macrophages, cancer, remodeling, adhesion, contraction, β4 integrin, TGF-β1, RhoA
- Published
- 2019
- Full Text
- View/download PDF
14. Dual Mechanisms of LYN Kinase Dysregulation Drive Aggressive Behavior in Breast Cancer Cells
- Author
-
Giusy Tornillo, Catherine Knowlson, Howard Kendrick, Joe Cooke, Hasan Mirza, Iskander Aurrekoetxea-Rodríguez, Maria d.M. Vivanco, Niamh E. Buckley, Anita Grigoriadis, and Matthew J. Smalley
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The SRC-family kinase LYN is highly expressed in triple-negative/basal-like breast cancer (TNBC) and in the cell of origin of these tumors, c-KIT-positive luminal progenitors. Here, we demonstrate LYN is a downstream effector of c-KIT in normal mammary cells and protective of apoptosis upon genotoxic stress. LYN activity is modulated by PIN1, a prolyl isomerase, and in BRCA1 mutant TNBC PIN1 upregulation activates LYN independently of c-KIT. Furthermore, the full-length LYN splice isoform (as opposed to the Δaa25–45 variant) drives migration and invasion of aggressive TNBC cells, while the ratio of splice variants is informative for breast cancer-specific survival across all breast cancers. Thus, dual mechanisms—uncoupling from upstream signals and splice isoform ratios—drive the activity of LYN in aggressive breast cancers. : Tornillo et al. show that in aggressive breast cancers, LYN activity is deregulated by a change in patterns of splice isoform expression. In BRCA1-dysfunctional breast cancers, LYN activity is upregulated by a prolyl isomerase (PIN1) that is normally repressed by BRCA1. Keywords: BRCA1, LYN kinase, triple-negative/basal-like breast cancer, PIN1, ESRP1, c-KIT
- Published
- 2018
- Full Text
- View/download PDF
15. Robust BRCA1‐like classification of copy number profiles of samples repeated across different datasets and platforms
- Author
-
Philip C. Schouten, Anita Grigoriadis, Thomas Kuilman, Hasan Mirza, Johnathan A. Watkins, Saskia A. Cooke, Ewald van Dyk, Tesa M. Severson, Oscar M. Rueda, Marlous Hoogstraat, Caroline V.M. Verhagen, Rachael Natrajan, Suet-Feung Chin, Esther H. Lips, Janneke Kruizinga, Arno Velds, Marja Nieuwland, Ron M. Kerkhoven, Oscar Krijgsman, Conchita Vens, Daniel Peeper, Petra M. Nederlof, Carlos Caldas, Andrew N. Tutt, Lodewyk F. Wessels, and Sabine C. Linn
- Subjects
BRCA1 ,Breast cancer ,Classification ,Copy number aberration profiles ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Breast cancers with BRCA1 germline mutation have a characteristic DNA copy number (CN) pattern. We developed a test that assigns CN profiles to be ‘BRCA1‐like’ or ‘non‐BRCA1‐like’, which refers to resembling a BRCA1‐mutated tumor or resembling a tumor without a BRCA1 mutation, respectively. Approximately one third of the BRCA1‐like breast cancers have a BRCA1 mutation, one third has hypermethylation of the BRCA1 promoter and one third has an unknown reason for being BRCA1‐like. This classification is indicative of patients' response to high dose alkylating and platinum containing chemotherapy regimens, which targets the inability of BRCA1 deficient cells to repair DNA double strand breaks. We investigated whether this classification can be reliably obtained with next generation sequencing and copy number platforms other than the bacterial artificial chromosome (BAC) array Comparative Genomic Hybridization (aCGH) on which it was originally developed. We investigated samples from 230 breast cancer patients for which a CN profile had been generated on two to five platforms, comprising low coverage CN sequencing, CN extraction from targeted sequencing panels (CopywriteR), Affymetrix SNP6.0, 135K/720K oligonucleotide aCGH, Affymetrix Oncoscan FFPE (MIP) technology, 3K BAC and 32K BAC aCGH. Pairwise comparison of genomic position‐mapped profiles from the original aCGH platform and other platforms revealed concordance. For most cases, biological differences between samples exceeded the differences between platforms within one sample. We observed the same classification across different platforms in over 80% of the patients and kappa values of at least 0.36. Differential classification could be attributed to CN profiles that were not strongly associated to one class. In conclusion, we have shown that the genomic regions that define our BRCA1‐like classifier are robustly measured by different CN profiling technologies, providing the possibility to retro‐ and prospectively investigate BRCA1‐like classification across a wide range of CN platforms.
- Published
- 2015
- Full Text
- View/download PDF
16. Mutation Processes in 293-Based Clones Overexpressing the DNA Cytosine Deaminase APOBEC3B.
- Author
-
Monica K Akre, Gabriel J Starrett, Jelmar S Quist, Nuri A Temiz, Michael A Carpenter, Andrew N J Tutt, Anita Grigoriadis, and Reuben S Harris
- Subjects
Medicine ,Science - Abstract
Molecular, cellular, and clinical studies have combined to demonstrate a contribution from the DNA cytosine deaminase APOBEC3B (A3B) to the overall mutation load in breast, head/neck, lung, bladder, cervical, ovarian, and other cancer types. However, the complete landscape of mutations attributable to this enzyme has yet to be determined in a controlled human cell system. We report a conditional and isogenic system for A3B induction, genomic DNA deamination, and mutagenesis. Human 293-derived cells were engineered to express doxycycline-inducible A3B-eGFP or eGFP constructs. Cells were subjected to 10 rounds of A3B-eGFP exposure that each caused 80-90% cell death. Control pools were subjected to parallel rounds of non-toxic eGFP exposure, and dilutions were done each round to mimic A3B-eGFP induced population fluctuations. Targeted sequencing of portions of TP53 and MYC demonstrated greater mutation accumulation in the A3B-eGFP exposed pools. Clones were generated and microarray analyses were used to identify those with the greatest number of SNP alterations for whole genome sequencing. A3B-eGFP exposed clones showed global increases in C-to-T transition mutations, enrichments for cytosine mutations within A3B-preferred trinucleotide motifs, and more copy number aberrations. Surprisingly, both control and A3B-eGFP clones also elicited strong mutator phenotypes characteristic of defective mismatch repair. Despite this additional mutational process, the 293-based system characterized here still yielded a genome-wide view of A3B-catalyzed mutagenesis in human cells and a system for additional studies on the compounded effects of simultaneous mutation mechanisms in cancer cells.
- Published
- 2016
- Full Text
- View/download PDF
17. Comparative Membranome expression analysis in primary tumors and derived cell lines.
- Author
-
Paolo Uva, Armin Lahm, Andrea Sbardellati, Anita Grigoriadis, Andrew Tutt, and Emanuele de Rinaldis
- Subjects
Medicine ,Science - Abstract
Despite the wide use of cell lines in cancer research, the extent to which their surface properties correspond to those of primary tumors is poorly characterized. The present study addresses this problem from a transcriptional standpoint, analyzing the expression of membrane protein genes--the Membranome--in primary tumors and immortalized in-vitro cultured tumor cells. 409 human samples, deriving from ten independent studies, were analyzed. These comprise normal tissues, primary tumors and tumor derived cell lines deriving from eight different tissues: brain, breast, colon, kidney, leukemia, lung, melanoma, and ovary. We demonstrated that the Membranome has greater power than the remainder of the transcriptome when used as input for the automatic classification of tumor samples. This feature is maintained in tumor derived cell lines. In most cases primary tumors show maximal similarity in Membranome expression with cell lines of same tissue origin. Differences in Membranome expression between tumors and cell lines were analyzed also at the pathway level and biological themes were identified that were differentially regulated in the two settings. Moreover, by including normal samples in the analysis, we quantified the degree to which cell lines retain the Membranome up- and down-regulations observed in primary tumors with respect to their normal counterparts. We showed that most of the Membranome up-regulations observed in primary tumors are lost in the in-vitro cultured cells. Conversely, the majority of Membranome genes down-regulated upon tumor transformation maintain lower expression levels also in the cell lines. This study points towards a central role of Membranome genes in the definition of the tumor phenotype. The comparative analysis of primary tumors and cell lines identifies the limits of cell lines as a model for the study of cancer-related processes mediated by the cell surface. Results presented allow for a more rational use of the cell lines as a model of cancer.
- Published
- 2010
- Full Text
- View/download PDF
18. Integrated functional, gene expression and genomic analysis for the identification of cancer targets.
- Author
-
Elizabeth Iorns, Christopher J Lord, Anita Grigoriadis, Sarah McDonald, Kerry Fenwick, Alan Mackay, Charles A Mein, Rachael Natrajan, Kay Savage, Narinder Tamber, Jorge S Reis-Filho, Nicholas C Turner, and Alan Ashworth
- Subjects
Medicine ,Science - Abstract
The majority of new drug approvals for cancer are based on existing therapeutic targets. One approach to the identification of novel targets is to perform high-throughput RNA interference (RNAi) cellular viability screens. We describe a novel approach combining RNAi screening in multiple cell lines with gene expression and genomic profiling to identify novel cancer targets. We performed parallel RNAi screens in multiple cancer cell lines to identify genes that are essential for viability in some cell lines but not others, suggesting that these genes constitute key drivers of cellular survival in specific cancer cells. This approach was verified by the identification of PIK3CA, silencing of which was selectively lethal to the MCF7 cell line, which harbours an activating oncogenic PIK3CA mutation. We combined our functional RNAi approach with gene expression and genomic analysis, allowing the identification of several novel kinases, including WEE1, that are essential for viability only in cell lines that have an elevated level of expression of this kinase. Furthermore, we identified a subset of breast tumours that highly express WEE1 suggesting that WEE1 could be a novel therapeutic target in breast cancer. In conclusion, this strategy represents a novel and effective strategy for the identification of functionally important therapeutic targets in cancer.
- Published
- 2009
- Full Text
- View/download PDF
19. Molecular and phenotypic characterisation of paediatric glioma cell lines as models for preclinical drug development.
- Author
-
Dorine A Bax, Suzanne E Little, Nathalie Gaspar, Lara Perryman, Lynley Marshall, Marta Viana-Pereira, Tania A Jones, Richard D Williams, Anita Grigoriadis, Gilles Vassal, Paul Workman, Denise Sheer, Rui M Reis, Andrew D J Pearson, Darren Hargrave, and Chris Jones
- Subjects
Medicine ,Science - Abstract
Although paediatric high grade gliomas resemble their adult counterparts in many ways, there appear to be distinct clinical and biological differences. One important factor hampering the development of new targeted therapies is the relative lack of cell lines derived from childhood glioma patients, as it is unclear whether the well-established adult lines commonly used are representative of the underlying molecular genetics of childhood tumours. We have carried out a detailed molecular and phenotypic characterisation of a series of paediatric high grade glioma cell lines in comparison to routinely used adult lines.All lines proliferate as adherent monolayers and express glial markers. Copy number profiling revealed complex genomes including amplification and deletions of genes known to be pivotal in core glioblastoma signalling pathways. Expression profiling identified 93 differentially expressed genes which were able to distinguish between the adult and paediatric high grade cell lines, including a number of kinases and co-ordinated sets of genes associated with DNA integrity and the immune response.These data demonstrate that glioma cell lines derived from paediatric patients show key molecular differences to those from adults, some of which are well known, whilst others may provide novel targets for evaluation in primary tumours. We thus provide the rationale and demonstrate the practicability of using paediatric glioma cell lines for preclinical and mechanistic studies.
- Published
- 2009
- Full Text
- View/download PDF
20. Deep Multi-Scale U-Net Architecture and Label-Noise Robust Training Strategies for Histopathological Image Segmentation.
- Author
-
Nikhil Cherian Kurian, Amit Lehan, Gregory Verghese, Nimish Dharamshi, Swati Meena, Mengyuan Li, Fangfang Liu, Cheryl Gillet, Swapnil Rane, Anita Grigoriadis, and Amit Sethi
- Published
- 2022
- Full Text
- View/download PDF
21. Deep Multi-Scale U-Net Architecture and Noise-Robust Training Strategies for Histopathological Image Segmentation.
- Author
-
Nikhil Cherian Kurian, Amit Lohan, Gregory Verghese, Nimish Dharamshi, Swati Meena, Mengyuan Li, Fangfang Liu, Cheryl Gillet, Swapnil Rane, Anita Grigoriadis, and Amit Sethi
- Published
- 2022
- Full Text
- View/download PDF
22. Multiscale deep learning framework captures systemic immune features in lymph nodes predictive of triple negative breast cancer outcome in large‐scale studies
- Author
-
Gregory Verghese, Mengyuan Li, Fangfang Liu, Amit Lohan, Nikhil Cherian Kurian, Swati Meena, Patrycja Gazinska, Aekta Shah, Aasiyah Oozeer, Terry Chan, Mark Opdam, Sabine Linn, Cheryl Gillett, Elena Alberts, Thomas Hardiman, Samantha Jones, Selvam Thavaraj, J Louise Jones, Roberto Salgado, Sarah E Pinder, Swapnil Rane, Amit Sethi, and Anita Grigoriadis
- Subjects
Pathology and Forensic Medicine - Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure S3 from A Four-gene Decision Tree Signature Classification of Triple-negative Breast Cancer: Implications for Targeted Therapeutics
- Author
-
Anita Grigoriadis, Andrew N.J. Tutt, Christopher J. Lord, Joyce A. O'Shaughnessy, Melinda L. Telli, Maggie C.U. Cheang, Hasan Mirza, and Jelmar Quist
- Abstract
Supplementary Figure S3: Molecular characterisation of the MC6 subtype in six TNBC cohorts.
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Tables S16-S20 from A Four-gene Decision Tree Signature Classification of Triple-negative Breast Cancer: Implications for Targeted Therapeutics
- Author
-
Anita Grigoriadis, Andrew N.J. Tutt, Christopher J. Lord, Joyce A. O'Shaughnessy, Melinda L. Telli, Maggie C.U. Cheang, Hasan Mirza, and Jelmar Quist
- Abstract
Supplementary Table S16: Results from the gene set enrichment analysis of 28 different immune signatures across the METABRIC-set 15 (MC subtypes) classification. Supplementary Table S17: Table representing the copy number status of the five genes part of the MAPK inactivation score across the METABRIC-set 15 (MC subtypes) classification. Supplementary Table S18: Output of the multivariable logistic regression analysis for the MC6 and the BL1 subtype in Sanofi Phase II. Supplementary Table S19: Output of the multivariable logistic regression analysis for the MC6 and the BL1 subtype in Sanofi Phase III. Supplementary Table S20: Results from the drug screen performed between MC6 and non-MC6 TNBC cell lines.
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Information from A Four-gene Decision Tree Signature Classification of Triple-negative Breast Cancer: Implications for Targeted Therapeutics
- Author
-
Anita Grigoriadis, Andrew N.J. Tutt, Christopher J. Lord, Joyce A. O'Shaughnessy, Melinda L. Telli, Maggie C.U. Cheang, Hasan Mirza, and Jelmar Quist
- Abstract
Supplementary Information: Extended methods.
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figures S1 - S16 from Genomic Complexity Profiling Reveals That HORMAD1 Overexpression Contributes to Homologous Recombination Deficiency in Triple-Negative Breast Cancers
- Author
-
Andrew N.J. Tutt, Anita Grigoriadis, Melinda L. Telli, James M. Ford, Shaveta Vinayak, Alan Ashworth, Christopher J. Lord, Jessica Frankum, Hasan Mirza, Maggie C.U. Cheang, Anders Isaksson, Markus Mayrhofer, Maria Jasin, Fabio Vanoli, Sarah Pinder, Cheryl Gillett, Bhavna Sidhu, Shalaka Joshi, Patrycja Gazinska, Vandna Shah, Daniel Weekes, and Johnathan Watkins
- Abstract
Supplementary Figure S1. Frequency of genomic aberrations in the KCL TNBC cohort. Supplementary Figure S2. Conceptual diagrams of Scores of Chromosomal Instability Scarring (SCINS). Supplementary Figure S3. Extent and location of SCINS in TNBC. Supplementary Figure S4. Chromosome-wise distribution of SCINS across four TNBC cohorts. Supplementary Figure S5. SCINS-defined tumour classes identify HORMAD1 to be among the top genes linked with Hi-AiCNA/Hi-CnLOH in PrECOG TNBCs. Supplementary Figure S6. Gene expression signatures and HORMAD1 expression. Supplementary Figure S7. Immunofluorescent localisation of HORMAD1 in cell lines. Supplementary Figure S8. Expression of HORMAD1 in breast cancer cell lines and tumour samples. Supplementary Figure S9. HORMAD1 inhibits activity of the DR-GFP HR reporter. Supplementary Figure S10. Effect of HORMAD1 overexpression and knockdown on cell growth and proliferation. Supplementary Figure S11. HORMAD1 promotes activity of the EJ5 NHEJ reporter. Supplementary Figure S12. Effect of HORMAD1 knockdown on 53BP1 and RAD51 foci in HCC1143. Supplementary Figure S13. BRCA1/2 mutation, platinum sensitivity and genomic scarring or HORMAD1 expression. Supplementary Figure S14. Effect of HORMAD1 knockdown on drug sensitivity in HCC1143. Supplementary Figure S15. ROC analysis of PrECOG TNBCs. Supplementary Figure S16. HORMAD1 expression in unselected and BRCA1/2 wildtype TNBCs.
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Tables S1-S10 from Evaluation of CDK12 Protein Expression as a Potential Novel Biomarker for DNA Damage Response–Targeted Therapies in Breast Cancer
- Author
-
Rachael Natrajan, Andrew R. Green, Yinyin Yuan, Christopher J. Lord, Ian O. Ellis, Emad A. Rhaka, Srinivasan Madhusudan, Stephen Y.T. Chan, Tarek M.A. Abdel-Fatah, Paul M. Moseley, Andrew Tutt, Anita Grigoriadis, Hasan Mirza, James Campbell, Divya Kriplani, Syed Haider, Frances Daley, Sarah L. Maguire, Patty T. Wai, and Kalnisha Naidoo
- Abstract
Supplementary Table S1: CDK12 expression in relation to clinicopathological parameters for the unselected TMA series; Supplementary Table S2: CDK12 expression in relation to clinicopathological parameters for the HER2-positive Herceptin treated series; Supplementary Table S3: CDK12 expression in relation to clinicopathological parameters for the METABRIC TMA series; Supplementary Table S4: Univariate and multivariate analysis of CDK12 in the TMA cohorts; Supplementary Table S5: CDK12 mutations in breast cancer. Taken from cBioportal (42,43); Supplementary Table S6: Correlations of CDK12 mutations, methylation, gene expression and ERBB2 copy number in primary breast cancers from TCGA; Supplementary Table S7: Correlations of CDK12 mutations and gene expression of DNA repair genes in primary tumors from METABRIC. P values from heteroscedastic 2-tailed, t-test; Supplementary Table S8: Correlations of CDK12 protein expression, and miRNA expression in primary tumors from METABRIC. Wilcoxon rank P values are corrected for multiple testing; Supplementary Table S9: Correlations of CDK12 protein expression and gene expression of DNA repair genes in primary tumors from METABRIC. Limma analysis corrected for multiple testing; Supplementary Table S10: Association of CDK12 absent and intermediate (0, 2-6) versus high (7-8) expression with DNA repair proteins in unselected and TNBC. P values from Fishers exact test.
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Methods, Figure Legends, Table Legends from Genomic Complexity Profiling Reveals That HORMAD1 Overexpression Contributes to Homologous Recombination Deficiency in Triple-Negative Breast Cancers
- Author
-
Andrew N.J. Tutt, Anita Grigoriadis, Melinda L. Telli, James M. Ford, Shaveta Vinayak, Alan Ashworth, Christopher J. Lord, Jessica Frankum, Hasan Mirza, Maggie C.U. Cheang, Anders Isaksson, Markus Mayrhofer, Maria Jasin, Fabio Vanoli, Sarah Pinder, Cheryl Gillett, Bhavna Sidhu, Shalaka Joshi, Patrycja Gazinska, Vandna Shah, Daniel Weekes, and Johnathan Watkins
- Abstract
Supplementary Methods, Figure Legends, Table Legends
- Published
- 2023
- Full Text
- View/download PDF
29. Data from A Four-gene Decision Tree Signature Classification of Triple-negative Breast Cancer: Implications for Targeted Therapeutics
- Author
-
Anita Grigoriadis, Andrew N.J. Tutt, Christopher J. Lord, Joyce A. O'Shaughnessy, Melinda L. Telli, Maggie C.U. Cheang, Hasan Mirza, and Jelmar Quist
- Abstract
The molecular complexity of triple-negative breast cancers (TNBCs) provides a challenge for patient management. We set out to characterize this heterogeneous disease by combining transcriptomics and genomics data, with the aim of revealing convergent pathway dependencies with the potential for treatment intervention. A Bayesian algorithm was used to integrate molecular profiles in two TNBC cohorts, followed by validation using five independent cohorts (n = 1,168), including three clinical trials. A four-gene decision tree signature was identified, which robustly classified TNBCs into six subtypes. All four genes in the signature (EXO1, TP53BP2, FOXM1, and RSU1) are associated with either genomic instability, malignant growth, or treatment response. One of the six subtypes, MC6, encompassed the largest proportion of tumors (∼50%) in early diagnosed TNBCs. In TNBC patients with metastatic disease, the MC6 proportion was reduced to 25%, and was independently associated with a higher response rate to platinum-based chemotherapy. In TNBC cell line data, platinum sensitivity was recapitulated, and a sensitivity to the inhibition of the phosphatase PPM1D was revealed. Molecularly, MC6-TNBCs displayed high levels of telomeric allelic imbalances, enrichment of CD4+ and CD8+ immune signatures, and reduced expression of genes negatively regulating the MAPK signaling pathway. These observations suggest that our integrative classification approach may identify TNBC patients with discernible and theoretically pharmacologically tractable features that merit further studies in prospective trials.
- Published
- 2023
- Full Text
- View/download PDF
30. Supplementary Tables S1 - S4 from Genomic Complexity Profiling Reveals That HORMAD1 Overexpression Contributes to Homologous Recombination Deficiency in Triple-Negative Breast Cancers
- Author
-
Andrew N.J. Tutt, Anita Grigoriadis, Melinda L. Telli, James M. Ford, Shaveta Vinayak, Alan Ashworth, Christopher J. Lord, Jessica Frankum, Hasan Mirza, Maggie C.U. Cheang, Anders Isaksson, Markus Mayrhofer, Maria Jasin, Fabio Vanoli, Sarah Pinder, Cheryl Gillett, Bhavna Sidhu, Shalaka Joshi, Patrycja Gazinska, Vandna Shah, Daniel Weekes, and Johnathan Watkins
- Abstract
Supplementary Table S1. KCL TNBC and breast cell line cohort characteristics. Supplementary Table S2. Categories of genomic segment used as the basis for measuring SCINS. Supplementary Table S3. Genes expressed in a SAM of different SCINS clusters. Supplementary Table S4. Gene expression signatures of genomic instability.
- Published
- 2023
- Full Text
- View/download PDF
31. Data from Evaluation of CDK12 Protein Expression as a Potential Novel Biomarker for DNA Damage Response–Targeted Therapies in Breast Cancer
- Author
-
Rachael Natrajan, Andrew R. Green, Yinyin Yuan, Christopher J. Lord, Ian O. Ellis, Emad A. Rhaka, Srinivasan Madhusudan, Stephen Y.T. Chan, Tarek M.A. Abdel-Fatah, Paul M. Moseley, Andrew Tutt, Anita Grigoriadis, Hasan Mirza, James Campbell, Divya Kriplani, Syed Haider, Frances Daley, Sarah L. Maguire, Patty T. Wai, and Kalnisha Naidoo
- Abstract
Disruption of Cyclin-Dependent Kinase 12 (CDK12) is known to lead to defects in DNA repair and sensitivity to platinum salts and PARP1/2 inhibitors. However, CDK12 has also been proposed as an oncogene in breast cancer. We therefore aimed to assess the frequency and distribution of CDK12 protein expression by IHC in independent cohorts of breast cancer and correlate this with outcome and genomic status. We found that 21% of primary unselected breast cancers were CDK12 high, and 10.5% were absent, by IHC. CDK12 positivity correlated with HER2 positivity but was not an independent predictor of breast cancer–specific survival taking HER2 status into account; however, absent CDK12 protein expression significantly correlated with a triple-negative phenotype. Interestingly, CDK12 protein absence was associated with reduced expression of a number of DDR proteins including ATR, Ku70/Ku80, PARP1, DNA-PK, and γH2AX, suggesting a novel mechanism of CDK12-associated DDR dysregulation in breast cancer. Our data suggest that diagnostic IHC quantification of CDK12 in breast cancer is feasible, with CDK12 absence possibly signifying defective DDR function. This may have important therapeutic implications, particularly for triple-negative breast cancers. Mol Cancer Ther; 17(1); 306–15. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Table Descriptions from Evaluation of CDK12 Protein Expression as a Potential Novel Biomarker for DNA Damage Response–Targeted Therapies in Breast Cancer
- Author
-
Rachael Natrajan, Andrew R. Green, Yinyin Yuan, Christopher J. Lord, Ian O. Ellis, Emad A. Rhaka, Srinivasan Madhusudan, Stephen Y.T. Chan, Tarek M.A. Abdel-Fatah, Paul M. Moseley, Andrew Tutt, Anita Grigoriadis, Hasan Mirza, James Campbell, Divya Kriplani, Syed Haider, Frances Daley, Sarah L. Maguire, Patty T. Wai, and Kalnisha Naidoo
- Abstract
Supplementary table descriptions
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary File from Genomic Complexity Profiling Reveals That HORMAD1 Overexpression Contributes to Homologous Recombination Deficiency in Triple-Negative Breast Cancers
- Author
-
Andrew N.J. Tutt, Anita Grigoriadis, Melinda L. Telli, James M. Ford, Shaveta Vinayak, Alan Ashworth, Christopher J. Lord, Jessica Frankum, Hasan Mirza, Maggie C.U. Cheang, Anders Isaksson, Markus Mayrhofer, Maria Jasin, Fabio Vanoli, Sarah Pinder, Cheryl Gillett, Bhavna Sidhu, Shalaka Joshi, Patrycja Gazinska, Vandna Shah, Daniel Weekes, and Johnathan Watkins
- Abstract
SWEAVE document. R code for SCINS analysis.
- Published
- 2023
- Full Text
- View/download PDF
34. Functional screening reveals HORMAD1-driven gene dependencies associated with translesion synthesis and replication stress tolerance
- Author
-
Dalia Tarantino, Callum Walker, Daniel Weekes, Helen Pemberton, Kathryn Davidson, Gonzalo Torga, Jessica Frankum, Ana M. Mendes-Pereira, Cynthia Prince, Riccardo Ferro, Rachel Brough, Stephen J. Pettitt, Christopher J. Lord, Anita Grigoriadis, and Andrew NJ Tutt
- Subjects
DNA Replication ,Cancer Research ,DNA Repair ,Phosphoric Diester Hydrolases ,Cell Cycle Proteins ,Triple Negative Breast Neoplasms ,DNA-Directed DNA Polymerase ,Nucleotidyltransferases ,Genomic Instability ,DNA-Binding Proteins ,X-ray Repair Cross Complementing Protein 1 ,Genetics ,Humans ,Molecular Biology ,DNA Damage - Abstract
HORMAD1 expression is usually restricted to germline cells, but it becomes mis-expressed in epithelial cells in ~60% of triple-negative breast cancers (TNBCs), where it is associated with elevated genomic instability (1). HORMAD1 expression in TNBC is bimodal with HORMAD1-positive TNBC representing a biologically distinct disease group. Identification of HORMAD1-driven genetic dependencies may uncover novel therapies for this disease group. To study HORMAD1-driven genetic dependencies, we generated a SUM159 cell line model with doxycycline-inducible HORMAD1 that replicated genomic instability phenotypes seen in HORMAD1-positive TNBC (1). Using small interfering RNA screens, we identified candidate genes whose depletion selectively inhibited the cellular growth of HORMAD1-expressing cells. We validated five genes (ATR, BRIP1, POLH, TDP1 and XRCC1), depletion of which led to reduced cellular growth or clonogenic survival in cells expressing HORMAD1. In addition to the translesion synthesis (TLS) polymerase POLH, we identified a HORMAD1-driven dependency upon additional TLS polymerases, namely POLK, REV1, REV3L and REV7. Our data confirms that out-of-context somatic expression of HORMAD1 can lead to genomic instability and reveals that HORMAD1 expression induces dependencies upon replication stress tolerance pathways, such as translesion synthesis. Our data also suggest that HORMAD1 expression could be a patient selection biomarker for agents targeting replication stress.
- Published
- 2022
- Full Text
- View/download PDF
35. Data from RORγt+ Innate Lymphoid Cells Promote Lymph Node Metastasis of Breast Cancers
- Author
-
Tony Ng, Andrew N.J. Tutt, Paul Ellis, Sergio A. Quezada, Borivoj Vojnovic, Peter Lane, David R. Withers, Uzma Hasan, Leo M. Carlin, Frank McCaughan, Simon Ameer-Beg, Simon P. Poland, Cheryl E. Gillett, Sarah E. Pinder, Stewart G. Martin, Julie Owen, Manuel Rodriguez-Justo, Dominic Patel, Natalie Woodman, Paul R. Barber, Gilbert O. Fruhwirth, Anita Grigoriadis, Felix Wong, Farzana Noor, Patrycja Gazinska, Anthony Cheung, Peter Gordon, Victoria Male, Rachel Evans, James Monypenny, Katherine Lawler, Fabian Flores-Borja, and Sheeba Irshad
- Abstract
Cancer cells tend to metastasize first to tumor-draining lymph nodes, but the mechanisms mediating cancer cell invasion into the lymphatic vasculature remain little understood. Here, we show that in the human breast tumor microenvironment (TME), the presence of increased numbers of RORγt+ group 3 innate lymphoid cells (ILC3) correlates with an increased likelihood of lymph node metastasis. In a preclinical mouse model of breast cancer, CCL21-mediated recruitment of ILC3 to tumors stimulated the production of the CXCL13 by TME stromal cells, which in turn promoted ILC3–stromal interactions and production of the cancer cell motile factor RANKL. Depleting ILC3 or neutralizing CCL21, CXCL13, or RANKL was sufficient to decrease lymph node metastasis. Our findings establish a role for RORγt+ILC3 in promoting lymphatic metastasis by modulating the local chemokine milieu of cancer cells in the TME. Cancer Res; 77(5); 1083–96. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
36. Supplementary Tables 1-4 from FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas
- Author
-
Alan Ashworth, Sunil R. Lakhani, Jose Palacios, Fernando Carlos Schmitt, David Hardisson, Barbara Weber, Kerry Fenwick, Marjan Iravani, Tim Dexter, Kay Savage, David Sarrio, Anita Grigoriadis, Alan Mackay, Chris Jones, Maryou Ballo Lambros, Nicholas C. Turner, Pete T. Simpson, and Jorge Sergio Reis-Filho
- Abstract
Supplementary Tables 1-4 from FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas
- Published
- 2023
- Full Text
- View/download PDF
37. Supplementary Methods from RORγt+ Innate Lymphoid Cells Promote Lymph Node Metastasis of Breast Cancers
- Author
-
Tony Ng, Andrew N.J. Tutt, Paul Ellis, Sergio A. Quezada, Borivoj Vojnovic, Peter Lane, David R. Withers, Uzma Hasan, Leo M. Carlin, Frank McCaughan, Simon Ameer-Beg, Simon P. Poland, Cheryl E. Gillett, Sarah E. Pinder, Stewart G. Martin, Julie Owen, Manuel Rodriguez-Justo, Dominic Patel, Natalie Woodman, Paul R. Barber, Gilbert O. Fruhwirth, Anita Grigoriadis, Felix Wong, Farzana Noor, Patrycja Gazinska, Anthony Cheung, Peter Gordon, Victoria Male, Rachel Evans, James Monypenny, Katherine Lawler, Fabian Flores-Borja, and Sheeba Irshad
- Abstract
Supplementary Methods described
- Published
- 2023
- Full Text
- View/download PDF
38. Supplementary Data from Tiling Path Genomic Profiling of Grade 3 Invasive Ductal Breast Cancers
- Author
-
Jorge S. Reis-Filho, Alan Ashworth, Horst Buerger, Christopher J. Lord, Jose Palacios, Andrew Tutt, David Hardisson, Narinder Tamber, Kerry Fenwick, Anita Grigoriadis, Alan Mackay, Daniela Hungermann, Laura G. Fulford, Betania Mahler-Araujo, Sydonia Rayter, Radost Vatcheva, Caterina Marchió, David S.P. Tan, Gema Moreno-Bueno, Socorro María Rodríguez-Pinilla, Maryou B. Lambros, and Rachael Natrajan
- Abstract
Supplementary Data from Tiling Path Genomic Profiling of Grade 3 Invasive Ductal Breast Cancers
- Published
- 2023
- Full Text
- View/download PDF
39. Supplementary Table from Repurposing Tin Mesoporphyrin as an Immune Checkpoint Inhibitor Shows Therapeutic Efficacy in Preclinical Models of Cancer
- Author
-
James N. Arnold, James F. Spicer, Sandra S. Diebold, Shahram Kordasti, Joy M. Burchell, Stephen F. Madden, Cheryl E. Gillett, Anita Grigoriadis, Andrew Tutt, Francesco Dazzi, Mieke Van Hemelrijck, Sharanpreet Lall, Paris Kosti, Mary Okesola, Jonathan Caron, James W. Opzoomer, and Tamara Muliaditan
- Abstract
Patient demographic of low and high quartiles of HMOX1 expressing tumors from the METABRIC dataset
- Published
- 2023
- Full Text
- View/download PDF
40. Video S1, Related to Figure 2 from RORγt+ Innate Lymphoid Cells Promote Lymph Node Metastasis of Breast Cancers
- Author
-
Tony Ng, Andrew N.J. Tutt, Paul Ellis, Sergio A. Quezada, Borivoj Vojnovic, Peter Lane, David R. Withers, Uzma Hasan, Leo M. Carlin, Frank McCaughan, Simon Ameer-Beg, Simon P. Poland, Cheryl E. Gillett, Sarah E. Pinder, Stewart G. Martin, Julie Owen, Manuel Rodriguez-Justo, Dominic Patel, Natalie Woodman, Paul R. Barber, Gilbert O. Fruhwirth, Anita Grigoriadis, Felix Wong, Farzana Noor, Patrycja Gazinska, Anthony Cheung, Peter Gordon, Victoria Male, Rachel Evans, James Monypenny, Katherine Lawler, Fabian Flores-Borja, and Sheeba Irshad
- Abstract
Time lapse microscopy experiment of sorted NKp46-ILC3 cells (CD3-, CD11c-, B220, NKp46-, CD127+, CD90.2+) co-cultured with MSC cells as described in Figure 2B. Low magnification (Ã-10) video shows the general clustering pattern of NKp46-ILC3 cells around the MSC cells over a duration of 10 hours.
- Published
- 2023
- Full Text
- View/download PDF
41. Data from Anti-Folate Receptor Alpha–Directed Antibody Therapies Restrict the Growth of Triple-negative Breast Cancer
- Author
-
Sophia N. Karagiannis, Andrew N.J. Tutt, Anita Grigoriadis, James F. Spicer, Cheryl Gillett, Frank O. Nestle, Sarah Pinder, Mariangela Figini, Silvana Canevari, Debra H. Josephs, Daniel Larcombe-Young, Gyula Petranyi, Silvia Crescioli, Heather J. Bax, Giulia Pellizzari, Silvia Mele, Nirmesh Patel, Luned Badder, Angela Clifford, Diana Dominguez Rodriguez, Matthew Fittall, Erika Francesch-Domenech, Rebecca Marlow, Hasan Mirza, Ricarda M. Hoffmann, Patrycja Gazinska, Kristina M. Ilieva, James Opzoomer, and Anthony Cheung
- Abstract
Purpose: Highly aggressive triple-negative breast cancers (TNBCs) lack validated therapeutic targets and have high risk of metastatic disease. Folate receptor alpha (FRα) is a central mediator of cell growth regulation that could serve as an important target for cancer therapy.Experimental Design: We evaluated FRα expression in breast cancers by genomic (n = 3,414) and IHC (n = 323) analyses and its association with clinical parameters and outcomes. We measured the functional contributions of FRα in TNBC biology by RNA interference and the antitumor functions of an antibody recognizing FRα (MOv18-IgG1), in vitro, and in human TNBC xenograft models.Results: FRα is overexpressed in significant proportions of aggressive basal like/TNBC tumors, and in postneoadjuvant chemotherapy–residual disease associated with a high risk of relapse. Expression is associated with worse overall survival. TNBCs show dysregulated expression of thymidylate synthase, folate hydrolase 1, and methylenetetrahydrofolate reductase, involved in folate metabolism. RNA interference to deplete FRα decreased Src and ERK signaling and resulted in reduction of cell growth. An anti-FRα antibody (MOv18-IgG1) conjugated with a Src inhibitor significantly restricted TNBC xenograft growth. Moreover, MOv18-IgG1 triggered immune-dependent cancer cell death in vitro by human volunteer and breast cancer patient immune cells, and significantly restricted orthotopic and patient-derived xenograft growth.Conclusions: FRα is overexpressed in high-grade TNBC and postchemotherapy residual tumors. It participates in cancer cell signaling and presents a promising target for therapeutic strategies such as ADCs, or passive immunotherapy priming Fc-mediated antitumor immune cell responses. Clin Cancer Res; 24(20); 5098–111. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
42. Supplementary Figures & Tables from RORγt+ Innate Lymphoid Cells Promote Lymph Node Metastasis of Breast Cancers
- Author
-
Tony Ng, Andrew N.J. Tutt, Paul Ellis, Sergio A. Quezada, Borivoj Vojnovic, Peter Lane, David R. Withers, Uzma Hasan, Leo M. Carlin, Frank McCaughan, Simon Ameer-Beg, Simon P. Poland, Cheryl E. Gillett, Sarah E. Pinder, Stewart G. Martin, Julie Owen, Manuel Rodriguez-Justo, Dominic Patel, Natalie Woodman, Paul R. Barber, Gilbert O. Fruhwirth, Anita Grigoriadis, Felix Wong, Farzana Noor, Patrycja Gazinska, Anthony Cheung, Peter Gordon, Victoria Male, Rachel Evans, James Monypenny, Katherine Lawler, Fabian Flores-Borja, and Sheeba Irshad
- Abstract
Figure S1, Related to Figure 1 FACS and immunofluorescence validation of Lin-CD127+CD90.2+NKP46- gated ILC3 cells Figure S2, Related to Figure 2 ELISA quantification of CXCL13 and CCL21 chemokines levels in conditioned media (CM) obtained from the bone marrow derived MSC cell line (HS-5) and from the breast cancer cell line 4T1.2 cell line is shown. Figure S3, Related to Figure 2 Proliferation assay of ILC3 cells and ELISA quantification of CXCL13 and CCL21 from siRNA transfected MSC cells. Figure S4, Related to Figure 4 CXCR5 expression on MSC cells. Figure S5, Related to Figure 5 Identification of a RORγT+CD127+CD3- ILC3 cells within the tertiary lymphoid structures within breast cancers. Figure S6, Related to Figure 5 Expression of lymphoid chemokine and chemokine receptor genes in breast cancer datasets. Table S1, Related to Figure 5 Table S1: Clinico-pathological characteristics for the METABRIC sample.
- Published
- 2023
- Full Text
- View/download PDF
43. Supplementary Figure 1 from FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas
- Author
-
Alan Ashworth, Sunil R. Lakhani, Jose Palacios, Fernando Carlos Schmitt, David Hardisson, Barbara Weber, Kerry Fenwick, Marjan Iravani, Tim Dexter, Kay Savage, David Sarrio, Anita Grigoriadis, Alan Mackay, Chris Jones, Maryou Ballo Lambros, Nicholas C. Turner, Pete T. Simpson, and Jorge Sergio Reis-Filho
- Abstract
Supplementary Figure 1 from FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas
- Published
- 2023
- Full Text
- View/download PDF
44. Data from FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas
- Author
-
Alan Ashworth, Sunil R. Lakhani, Jose Palacios, Fernando Carlos Schmitt, David Hardisson, Barbara Weber, Kerry Fenwick, Marjan Iravani, Tim Dexter, Kay Savage, David Sarrio, Anita Grigoriadis, Alan Mackay, Chris Jones, Maryou Ballo Lambros, Nicholas C. Turner, Pete T. Simpson, and Jorge Sergio Reis-Filho
- Abstract
Purpose: Classic lobular carcinomas (CLC) account for 10% to 15% of all breast cancers. At the genetic level, CLCs show recurrent physical loss of chromosome16q coupled with the lack of E-cadherin (CDH1 gene) expression. However, little is known about the putative therapeutic targets for these tumors. The aim of this study was to characterize CLCs at the molecular genetic level and identify putative therapeutic targets.Experimental Design: We subjected 13 cases of CLC to a comprehensive molecular analysis including immunohistochemistry for E-cadherin, estrogen and progesterone receptors, HER2/neu and p53; high-resolution comparative genomic hybridization (HR-CGH); microarray-based CGH (aCGH); and fluorescent and chromogenic in situ hybridization for CCND1 and FGFR1.Results: All cases lacked the expression of E-cadherin, p53, and HER2, and all but one case was positive for estrogen receptors. HR-CGH revealed recurrent gains on 1q and losses on 16q (both, 85%). aCGH showed a good agreement with but higher resolution and sensitivity than HR-CGH. Recurrent, high level gains at 11q13 (CCND1) and 8p12-p11.2 were identified in seven and six cases, respectively, and were validated with in situ hybridization. Examination of aCGH and the gene expression profile data of the cell lines, MDA-MB-134 and ZR-75-1, which harbor distinct gains of 8p12-p11.2, identified FGFR1 as a putative amplicon driver of 8p12-p11.2 amplification in MDA-MB-134. Inhibition of FGFR1 expression using small interfering RNA or a small-molecule chemical inhibitor showed that FGFR1 signaling contributes to the survival of MDA-MB-134 cells.Conclusions: Our findings suggest that receptor FGFR1 inhibitors may be useful as therapeutics in a subset of CLCs.
- Published
- 2023
- Full Text
- View/download PDF
45. Data from Repurposing Tin Mesoporphyrin as an Immune Checkpoint Inhibitor Shows Therapeutic Efficacy in Preclinical Models of Cancer
- Author
-
James N. Arnold, James F. Spicer, Sandra S. Diebold, Shahram Kordasti, Joy M. Burchell, Stephen F. Madden, Cheryl E. Gillett, Anita Grigoriadis, Andrew Tutt, Francesco Dazzi, Mieke Van Hemelrijck, Sharanpreet Lall, Paris Kosti, Mary Okesola, Jonathan Caron, James W. Opzoomer, and Tamara Muliaditan
- Abstract
Purpose: Unprecedented clinical outcomes have been achieved in a variety of cancers by targeting immune checkpoint molecules. This preclinical study investigates heme oxygenase-1 (HO-1), an immunosuppressive enzyme that is expressed in a wide variety of cancers, as a potential immune checkpoint target in the context of a chemotherapy-elicited antitumor immune response. We evaluate repurposing tin mesoporphyrin (SnMP), which has demonstrated safety and efficacy targeting hepatic HO in the clinic for the treatment of hyperbilirubinemia, as an immune checkpoint blockade therapy for the treatment of cancer.Experimental Design: SnMP and genetic inactivation of myeloid HO-1 were evaluated alongside 5-fluorouracil in an aggressive spontaneous murine model of breast cancer (MMTV-PyMT). Single-cell RNA sequencing analysis, tumor microarray, and clinical survival data from breast cancer patients were used to support the clinical relevance of our observations.Results: We demonstrate that SnMP inhibits immune suppression of chemotherapy-elicited CD8+ T cells by targeting myeloid HO-1 activity in the tumor microenvironment. Microarray and survival data from breast cancer patients reveal that HO-1 is a poor prognostic factor in patients receiving chemotherapy. Single-cell RNA-sequencing analysis suggests that the myeloid lineage is a significant source of HO-1 expression, and is co-expressed with the immune checkpoints PD-L1/2 in human breast tumors. In vivo, we therapeutically compare the efficacy of targeting these two pathways alongside immune-stimulating chemotherapy, and demonstrate that the efficacy of SnMP compares favorably with PD-1 blockade in preclinical models.Conclusions: SnMP could represent a novel immune checkpoint therapy, which may improve the immunological response to chemotherapy. Clin Cancer Res; 24(7); 1617–28. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
46. Supplementary data from Anti-Folate Receptor Alpha–Directed Antibody Therapies Restrict the Growth of Triple-negative Breast Cancer
- Author
-
Sophia N. Karagiannis, Andrew N.J. Tutt, Anita Grigoriadis, James F. Spicer, Cheryl Gillett, Frank O. Nestle, Sarah Pinder, Mariangela Figini, Silvana Canevari, Debra H. Josephs, Daniel Larcombe-Young, Gyula Petranyi, Silvia Crescioli, Heather J. Bax, Giulia Pellizzari, Silvia Mele, Nirmesh Patel, Luned Badder, Angela Clifford, Diana Dominguez Rodriguez, Matthew Fittall, Erika Francesch-Domenech, Rebecca Marlow, Hasan Mirza, Ricarda M. Hoffmann, Patrycja Gazinska, Kristina M. Ilieva, James Opzoomer, and Anthony Cheung
- Abstract
Supplementary Figure Legends, Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
47. Data from Tiling Path Genomic Profiling of Grade 3 Invasive Ductal Breast Cancers
- Author
-
Jorge S. Reis-Filho, Alan Ashworth, Horst Buerger, Christopher J. Lord, Jose Palacios, Andrew Tutt, David Hardisson, Narinder Tamber, Kerry Fenwick, Anita Grigoriadis, Alan Mackay, Daniela Hungermann, Laura G. Fulford, Betania Mahler-Araujo, Sydonia Rayter, Radost Vatcheva, Caterina Marchió, David S.P. Tan, Gema Moreno-Bueno, Socorro María Rodríguez-Pinilla, Maryou B. Lambros, and Rachael Natrajan
- Abstract
Purpose: To characterize the molecular genetic profiles of grade 3 invasive ductal carcinomas of no special type using high-resolution microarray-based comparative genomic hybridization (aCGH) and to identify recurrent amplicons harboring putative therapeutic targets associated with luminal, HER-2, and basal-like tumor phenotypes.Experimental Design: Ninety-five grade 3 invasive ductal carcinomas of no special type were classified into luminal, HER-2, and basal-like subgroups using a previously validated immunohistochemical panel. Tumor samples were microdissected and subjected to aCGH using a tiling path 32K BAC array platform. Selected regions of recurrent amplification were validated by means of in situ hybridization. Expression of genes pertaining to selected amplicons was investigated using quantitative real-time PCR and gene silencing was done using previously validated short hairpin RNA constructs.Results: We show that basal-like and HER-2 tumors are characterized by “sawtooth” and “firestorm” genetic patterns, respectively, whereas luminal cancers were more heterogeneous. Apart from confirming known amplifications associated with basal-like (1q21, 10p, and 12p), luminal (8p12, 11q13, and 11q14), and HER-2 (17q12) cancers, we identified previously unreported recurrent amplifications associated with each molecular subgroup: 19q12 in basal-like, 1q32.1 in luminal, and 14q12 in HER-2 cancers. PPM1D gene amplification (17q23.2) was found in 20% and 8% of HER-2 and luminal cancers, respectively. Silencing of PPM1D by short hairpin RNA resulted in selective loss of viability in tumor cell lines harboring the 17q23.2 amplification.Conclusions: Our results show the power of aCGH analysis in unraveling the genetic profiles of specific subgroups of cancer and for the identification of novel therapeutic targets.
- Published
- 2023
- Full Text
- View/download PDF
48. Supplementary figures from Anti-Folate Receptor Alpha–Directed Antibody Therapies Restrict the Growth of Triple-negative Breast Cancer
- Author
-
Sophia N. Karagiannis, Andrew N.J. Tutt, Anita Grigoriadis, James F. Spicer, Cheryl Gillett, Frank O. Nestle, Sarah Pinder, Mariangela Figini, Silvana Canevari, Debra H. Josephs, Daniel Larcombe-Young, Gyula Petranyi, Silvia Crescioli, Heather J. Bax, Giulia Pellizzari, Silvia Mele, Nirmesh Patel, Luned Badder, Angela Clifford, Diana Dominguez Rodriguez, Matthew Fittall, Erika Francesch-Domenech, Rebecca Marlow, Hasan Mirza, Ricarda M. Hoffmann, Patrycja Gazinska, Kristina M. Ilieva, James Opzoomer, and Anthony Cheung
- Abstract
Supplementary figures
- Published
- 2023
- Full Text
- View/download PDF
49. Supplementary Figures legends from RORγt+ Innate Lymphoid Cells Promote Lymph Node Metastasis of Breast Cancers
- Author
-
Tony Ng, Andrew N.J. Tutt, Paul Ellis, Sergio A. Quezada, Borivoj Vojnovic, Peter Lane, David R. Withers, Uzma Hasan, Leo M. Carlin, Frank McCaughan, Simon Ameer-Beg, Simon P. Poland, Cheryl E. Gillett, Sarah E. Pinder, Stewart G. Martin, Julie Owen, Manuel Rodriguez-Justo, Dominic Patel, Natalie Woodman, Paul R. Barber, Gilbert O. Fruhwirth, Anita Grigoriadis, Felix Wong, Farzana Noor, Patrycja Gazinska, Anthony Cheung, Peter Gordon, Victoria Male, Rachel Evans, James Monypenny, Katherine Lawler, Fabian Flores-Borja, and Sheeba Irshad
- Abstract
Supplementary Figure legends
- Published
- 2023
- Full Text
- View/download PDF
50. Supplementary Figures 1-10, Supplementary Materials and Methods, Supplementary References from Repurposing Tin Mesoporphyrin as an Immune Checkpoint Inhibitor Shows Therapeutic Efficacy in Preclinical Models of Cancer
- Author
-
James N. Arnold, James F. Spicer, Sandra S. Diebold, Shahram Kordasti, Joy M. Burchell, Stephen F. Madden, Cheryl E. Gillett, Anita Grigoriadis, Andrew Tutt, Francesco Dazzi, Mieke Van Hemelrijck, Sharanpreet Lall, Paris Kosti, Mary Okesola, Jonathan Caron, James W. Opzoomer, and Tamara Muliaditan
- Abstract
Supplementary Figure S1â",HO-1 expression in MMTV-PyMT tumors; Supplementary Figure S2â",Effects of SnMP and chemotherapy combination therapy; Supplementary Figure S3â",HO-1+ TAMs in murine models of breast cancer; Supplementary Figure S4â",Genetic inactivation and therapeutic inhibition of myeloid HO-1 in the MMTV-PyMT model; Supplementary Figure S5â",HO-1 in human cancer; Supplementary Figure S6â",tSNE clustering identifies stromal and epithelial cell types from breast cancer patient single-cell RNA sequencing data; Supplementary Figure 7â", Single cell RNA-Seq gene expression for checkpoint molecules grouped by inferred cell type; Supplementary Figure S8â",HO-1 protein in myeloid cells in human cancer; Supplementary Figure S9â", Comparative efficacies of therapeutically targeting HO-1 and PD-1 alongside paclitaxel treatment in 4T1 tumors; Supplementary Figure S10â",Improving checkpoint blockade therapy using SnMP.
- 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.