1
|
Jackstadt R, van Hooff SR, Leach JD, Cortes-Lavaud X, Lohuis JO, Ridgway RA, Wouters VM, Roper J, Kendall TJ, Roxburgh CS, Horgan PG, Nixon C, Nourse C, Gunzer M, Clark W, Hedley A, Yilmaz OH, Rashid M, Bailey P, Biankin AV, Campbell AD, Adams DJ, Barry ST, Steele CW, Medema JP, Sansom OJ. Epithelial NOTCH Signaling Rewires the Tumor Microenvironment of Colorectal Cancer to Drive Poor-Prognosis Subtypes and Metastasis. Cancer Cell 2019; 36:319-336.e7. [PMID: 31526760 PMCID: PMC6853173 DOI: 10.1016/j.ccell.2019.08.003] [Citation(s) in RCA: 289] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/31/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022]
Abstract
The metastatic process of colorectal cancer (CRC) is not fully understood and effective therapies are lacking. We show that activation of NOTCH1 signaling in the murine intestinal epithelium leads to highly penetrant metastasis (100% metastasis; with >80% liver metastases) in KrasG12D-driven serrated cancer. Transcriptional profiling reveals that epithelial NOTCH1 signaling creates a tumor microenvironment (TME) reminiscent of poorly prognostic human CRC subtypes (CMS4 and CRIS-B), and drives metastasis through transforming growth factor (TGF) β-dependent neutrophil recruitment. Importantly, inhibition of this recruitment with clinically relevant therapeutic agents blocks metastasis. We propose that NOTCH1 signaling is key to CRC progression and should be exploited clinically.
Collapse
|
research-article |
6 |
289 |
2
|
Leslie J, Mackey JBG, Jamieson T, Ramon-Gil E, Drake TM, Fercoq F, Clark W, Gilroy K, Hedley A, Nixon C, Luli S, Laszczewska M, Pinyol R, Esteban-Fabró R, Willoughby CE, Haber PK, Andreu-Oller C, Rahbari M, Fan C, Pfister D, Raman S, Wilson N, Müller M, Collins A, Geh D, Fuller A, McDonald D, Hulme G, Filby A, Cortes-Lavaud X, Mohamed NE, Ford CA, Raffo Iraolagoitia XL, McFarlane AJ, McCain MV, Ridgway RA, Roberts EW, Barry ST, Graham GJ, Heikenwälder M, Reeves HL, Llovet JM, Carlin LM, Bird TG, Sansom OJ, Mann DA. CXCR2 inhibition enables NASH-HCC immunotherapy. Gut 2022; 71:gutjnl-2021-326259. [PMID: 35477863 PMCID: PMC9484388 DOI: 10.1136/gutjnl-2021-326259] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/17/2022] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is increasingly associated with non-alcoholic steatohepatitis (NASH). HCC immunotherapy offers great promise; however, recent data suggests NASH-HCC may be less sensitive to conventional immune checkpoint inhibition (ICI). We hypothesised that targeting neutrophils using a CXCR2 small molecule inhibitor may sensitise NASH-HCC to ICI therapy. DESIGN Neutrophil infiltration was characterised in human HCC and mouse models of HCC. Late-stage intervention with anti-PD1 and/or a CXCR2 inhibitor was performed in murine models of NASH-HCC. The tumour immune microenvironment was characterised by imaging mass cytometry, RNA-seq and flow cytometry. RESULTS Neutrophils expressing CXCR2, a receptor crucial to neutrophil recruitment in acute-injury, are highly represented in human NASH-HCC. In models of NASH-HCC lacking response to ICI, the combination of a CXCR2 antagonist with anti-PD1 suppressed tumour burden and extended survival. Combination therapy increased intratumoural XCR1+ dendritic cell activation and CD8+ T cell numbers which are associated with anti-tumoural immunity, this was confirmed by loss of therapeutic effect on genetic impairment of myeloid cell recruitment, neutralisation of the XCR1-ligand XCL1 or depletion of CD8+ T cells. Therapeutic benefit was accompanied by an unexpected increase in tumour-associated neutrophils (TANs) which switched from a protumour to anti-tumour progenitor-like neutrophil phenotype. Reprogrammed TANs were found in direct contact with CD8+ T cells in clusters that were enriched for the cytotoxic anti-tumoural protease granzyme B. Neutrophil reprogramming was not observed in the circulation indicative of the combination therapy selectively influencing TANs. CONCLUSION CXCR2-inhibition induces reprogramming of the tumour immune microenvironment that promotes ICI in NASH-HCC.
Collapse
|
research-article |
3 |
99 |
3
|
Cristóbal I, Garcia-Orti L, Cirauqui C, Cortes-Lavaud X, García-Sánchez MA, Calasanz MJ, Odero MD. Overexpression of SET is a recurrent event associated with poor outcome and contributes to protein phosphatase 2A inhibition in acute myeloid leukemia. Haematologica 2011; 97:543-50. [PMID: 22133779 DOI: 10.3324/haematol.2011.050542] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Protein phosphatase 2A is a novel potential therapeutic target in several types of chronic and acute leukemia, and its inhibition is a common event in acute myeloid leukemia. Upregulation of SET is essential to inhibit protein phosphatase 2A in chronic myeloid leukemia, but its importance in acute myeloid leukemia has not yet been explored. DESIGN AND METHODS We quantified SET expression by real time reverse transcriptase polymerase chain reaction in 214 acute myeloid leukemia patients at diagnosis. Western blot was performed in acute myeloid leukemia cell lines and in 16 patients' samples. We studied the effect of SET using cell viability assays. Bioinformatics analysis of the SET promoter, chromatin immunoprecipitation, and luciferase assays were performed to evaluate the transcriptional regulation of SET. RESULTS SET overexpression was found in 60/214 patients, for a prevalence of 28%. Patients with SET overexpression had worse overall survival (P<0.01) and event-free survival (P<0.01). Deregulation of SET was confirmed by western blot in both cell lines and patients' samples. Functional analysis showed that SET promotes proliferation, and restores cell viability after protein phosphatase 2A overexpression. We identified EVI1 overexpression as a mechanism involved in SET deregulation in acute myeloid leukemia cells. CONCLUSIONS These findings suggest that SET overexpression is a key mechanism in the inhibition of PP2A in acute myeloid leukemia, and that EVI1 overexpression contributes to the deregulation of SET. Furthermore, SET overexpression is associated with a poor outcome in acute myeloid leukemia, and it can be used to identify a subgroup of patients who could benefit from future treatments based on PP2A activators.
Collapse
|
Research Support, Non-U.S. Gov't |
14 |
99 |
4
|
Corry SM, McCorry AM, Lannagan TR, Leonard NA, Fisher NC, Byrne RM, Tsantoulis P, Cortes-Lavaud X, Amirkhah R, Redmond KL, McCooey AJ, Malla SB, Rogan E, Sakhnevych S, Gillespie MA, White M, Richman SD, Jackstadt RF, Campbell AD, Maguire S, McDade SS, Longley DB, Loughrey MB, Coleman HG, Kerr EM, Tejpar S, Maughan T, Leedham SJ, Small DM, Ryan AE, Sansom OJ, Lawler M, Dunne PD. Activation of innate-adaptive immune machinery by poly(I:C) exposes a therapeutic vulnerability to prevent relapse in stroma-rich colon cancer. Gut 2022; 71:2502-2517. [PMID: 35477539 PMCID: PMC9664095 DOI: 10.1136/gutjnl-2021-326183] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/12/2022] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Stroma-rich tumours represent a poor prognostic subtype in stage II/III colon cancer (CC), with high relapse rates and limited response to standard adjuvant chemotherapy. DESIGN To address the lack of efficacious therapeutic options for patients with stroma-rich CC, we stratified our human tumour cohorts according to stromal content, enabling identification of the biology underpinning relapse and potential therapeutic vulnerabilities specifically within stroma-rich tumours that could be exploited clinically. Following human tumour-based discovery and independent clinical validation, we use a series of in vitro and stroma-rich in vivo models to test and validate the therapeutic potential of elevating the biology associated with reduced relapse in human tumours. RESULTS By performing our analyses specifically within the stroma-rich/high-fibroblast (HiFi) subtype of CC, we identify and validate the clinical value of a HiFi-specific prognostic signature (HPS), which stratifies tumours based on STAT1-related signalling (High-HPS v Low-HPS=HR 0.093, CI 0.019 to 0.466). Using in silico, in vitro and in vivo models, we demonstrate that the HPS is associated with antigen processing and presentation within discrete immune lineages in stroma-rich CC, downstream of double-stranded RNA and viral response signalling. Treatment with the TLR3 agonist poly(I:C) elevated the HPS signalling and antigen processing phenotype across in vitro and in vivo models. In an in vivo model of stroma-rich CC, poly(I:C) treatment significantly increased systemic cytotoxic T cell activity (p<0.05) and reduced liver metastases (p<0.0002). CONCLUSION This study reveals new biological insight that offers a novel therapeutic option to reduce relapse rates in patients with the worst prognosis CC.
Collapse
|
research-article |
3 |
6 |
5
|
Maicas M, Vazquez I, Vicente C, Cortes-Lavaud X, Marcotegui N, Urquiza L, Calasanz MJ, Odero MD. Abstract 85: RUNX1 and ELK1 directly regulate the transcription of EVI1 during megakaryocytic differentiation. Cancer Res 2012. [DOI: 10.1158/1538-7445.am2012-85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
High expression of EVI1 is a negative prognostic indicator of survival in patients with acute myeloid leukemia (AML). The EVI1 gene (3q26) codes for a transcription factor indispensable for the proliferation and self-renewal of HSCs during embryogenesis and in the adult, and is downregulated upon differentiation; however, the regulation of EVI1 in megakaryocytic differentiation should be different, as it is highly expressed in megakaryocytes and platelets, but not in other committed hematopoietic cells. Our aim was to identify transcription factors involved in the regulation of EVI1. Site-directed mutagenesis and ChIP assays identified RUNX1 and ELK1 as putative transcription factors of EVI1. Furthermore, knockdown of RUNX1 and ELK1 led to EVI1 downregulation, and their overexpression to upregulation of EVI1. Of note, in a series of patient samples with AML at diagnosis (n=46), we found a significant positive correlation between EVI1 and RUNX1, and EVI1 and ELK1 mRNA expression levels. RUNX1 has been reported as a key transcription factor in megakaryocytic development. To investigate whether RUNX1 could regulate EVI1 in this process, we treated K562 cells with TPA. After the treatment, we observed an increase in RUNX1, as previously described, and in ELK1 and EVI1. Knockdown of RUNX1 resulted in a marked decrease of the megakaryocytic markers ITGA2B and ITGB3, and of EVI1. Moreover, EVI1 knockdown led to the same effects by blocking the differentiation process, suggesting that the role of EVI1 in megakaryocytic differentiation would be through RUNX1. Interestingly, chromatin immunoprecipitation assay (ChIP) showed that RUNX1 regulates the transcription of EVI1 by acetylation of the histone H3 on its promoter region. In conclusion, we demonstrate that RUNX1 and ELK1, two proteins with essential functions in hematopoiesis, regulate EVI1 in AML. Moreover, our results suggest that these transcription factors could regulate EVI1 expression during megakaryocytic differentiation, and that RUNX1 would activate EVI1 expression through acetylation of the histone H3 on its promoter region. This study opens new directions to further understand the mechanisms of EVI1 overexpressing leukemias.
Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 85. doi:1538-7445.AM2012-85
Collapse
|
|
13 |
|
6
|
Maicas M, Vazquez I, Cortes-Lavaud X, Garcia-Sanchez MA, Marcotegui N, Urquiza L, Cristobal I, Calasanz MJ, Odero MD. Abstract 2205: The EVI1 human protein regulates its own transcription. Role of the different isoforms. Cancer Res 2012. [DOI: 10.1158/1538-7445.am2012-2205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
The EVI1 transcription factor has an essential role in development and leukemogenesis. High expression of EVI1 is a negative prognostic indicator of survival in acute myeloid leukemia (AML) patients, irrespective of the presence of 3q26 rearrangements; however, the only known mechanisms that lead to EVI1 overexpression are 3q aberrations, and MLL oncoproteins, which activate the transcription of EVI1 in hematopoietic stem cells. Our aim was to study mechanisms of EVI1 expression at transcriptional level, and to determine the role of the different isoforms of this protein in leukemogenesis. Bioinformatic analysis of the promoter of the EVI1 gene identified 8 EVI1-potential binding sites. ChIP assay confirmed that EVI1 directly binds to two of these sites, suggesting that EVI1 could act as a regulator of its own expression. Luciferase assays showed that the EVI1-full length (145KDa) protein binds to the EVI1 promoter as a transcriptional activator. The EVI1 human locus codes for at least 4 different proteins: EVI1-145KDa; MDS1EVI1, with an additional PR domain; delta324, that lacks the zinc fingers 6 and 7; and RP-9 that lacks 9 amino-acids from the repressor domain. The best studied is the EVI1-145KDa protein; however, the functional significance of the other isoforms is unclear. This prompted us to determine whether the other isoforms had the same ability than EVI1-145KDa. We observed that the RP-9, delta324 and MDS1EVI1 isoforms repressed the transcription of EVI1 by binding to a more distal promoter region. To confirm these results we transfected HEK293T with different expression vectors. EVI1-145KDa overexpression increased the EVI1-1A, -1B and -1D transcripts. However, RP-9 decreased M/E, EVI1-1A, -1B and -1D, and delta324 decreased EVI1-1D. These results support the data obtained with the luciferase experiments; however, we found no differences at protein level. In addition, we analyzed at mRNA and protein level 16 cell lines and 18 samples of patients with AML. We found a positive correlation between the expression of EVI1-145KDa and MDS1EVI1 at mRNA and protein level. However, some of the patients with the same protein levels expressed different delta324 transcript levels, suggesting either a regulation of the protein expression at post-transcriptional level or protein stability. Cycloheximide experiments demonstrated that the delta324 isoform is more stable than the MDS1EVI1 and EVI1-145KDa proteins. Preliminary results show that knockdown of EVI1 caused a decrease in proliferation with an increase in apoptosis. Further experiments are in progress to determine the role of each isoform. In conclusion, our results suggest that the EVI1 isoforms oppositely regulate the expression of the EVI1 human gene by specific binding to the promoter motifs. This study opens new directions to further understand the mechanisms of EVI1 overexpressing leukemias.
Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2205. doi:1538-7445.AM2012-2205
Collapse
|
|
13 |
|
7
|
Pennel KAF, Steele CW, Quinn JA, Roseweir AK, Jackstadt R, Nixon C, Cortes-Lavaud X, Roxburgh CSD, Van Wyk HC, McMillan DC, Horgan PG, Sansom O, Edwards J. Effect of phenotype on outcome in synchronously resected primary colorectal cancer and matched liver metastases. J Clin Oncol 2020. [DOI: 10.1200/jco.2020.38.4_suppl.221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
221 Background: 5-year survival of patients with resectable colorectal liver metastases is 25-40%. Mechanisms of disease progression are heterogenous and do not follow a clearly defined pathway from genotype to phenotype. In stage I-III colorectal cancer (CRC), patients with high tumor stroma exhibit poor prognosis, while those with high immune cell infiltrate do well following resection. We hypothesise that stromally-dense phenotypes lead to T cell exclusion, myeloid cell accumulation and aggressive metastatic progression. Here, we examine relationships between histological tumor phenotype, cellular infiltrate and outcomes in metastatic CRC. Methods: A unique cohort of synchronously resected primary CRC and matched liver metastases (n = 46) were assessed for immune cell infiltration (CD3, CD4, CD8, CD68, CD66b), inflammatory signalling (CXCR2, PDL-1, MMP9) and hypoxia (CAIX) using immunohistochemistry. Tumors were phenotypically subtyped using immune infiltrate (Klintrup-Makinen Grade (KM)), stromal invasion (tumor-stroma percentage (TSP)) and proliferation (Ki67). Results: Phenotypic subtype of primary tumors was predictive of metastatic subtype (rho = 0.522, p = 0.003). Immune phenotypes were associated with good prognosis and stromal phenotypes with poor prognosis (p = 0.004). Infiltration of macrophages and granulocytes associated with poor outcomes (p = 0.018) and increased CXCR2 expression (p = 0.03) at both sites. Increased CXCR2+ cells and macrophages at both sites associated with stromal phenotype (p = 0.02), tumour budding (p = 0.002), low KM grade (p = 0.05) and poor prognosis (p = 0.002). Macrophage and MMP9 levels increased in metastases compared to primary tumour, but no changes were seen in lymphocyte infiltration, CXCR2 and CD66b. Conclusions: Density of immune cell infiltrate, in the primary and metastatic niche, conferred good prognosis. In contrast, stromal, myeloid rich tumors convey poor prognosis. This clinically relevant and histologically efficient process permits segregation of disease and supports further study of relationships in the tumour microenvironment of CRC in the context of chemotherapy to better target therapeutics to individual patients.
Collapse
|
|
5 |
|
8
|
Fetit R, McLaren AS, White M, Mills ML, Falconer J, Cortes-Lavaud X, Gilroy K, Lannagan TR, Ridgway RA, Nixon C, Naiker V, Njunge R, Clarke CJ, Whyte D, Kirschner K, Jackstadt R, Norman J, Carlin LM, Campbell AD, Sansom OJ, Steele CW. Characterizing Neutrophil Subtypes in Cancer Using scRNA Sequencing Demonstrates the Importance of IL1β/CXCR2 Axis in Generation of Metastasis-specific Neutrophils. CANCER RESEARCH COMMUNICATIONS 2024; 4:588-606. [PMID: 38358352 PMCID: PMC10903300 DOI: 10.1158/2767-9764.crc-23-0319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/08/2023] [Accepted: 02/08/2024] [Indexed: 02/16/2024]
Abstract
Neutrophils are a highly heterogeneous cellular population. However, a thorough examination of the different transcriptional neutrophil states between health and malignancy has not been performed. We utilized single-cell RNA sequencing of human and murine datasets, both publicly available and independently generated, to identify neutrophil transcriptomic subtypes and developmental lineages in health and malignancy. Datasets of lung, breast, and colorectal cancer were integrated to establish and validate neutrophil gene signatures. Pseudotime analysis was used to identify genes driving neutrophil development from health to cancer. Finally, ligand-receptor interactions and signaling pathways between neutrophils and other immune cell populations in primary colorectal cancer and metastatic colorectal cancer were investigated. We define two main neutrophil subtypes in primary tumors: an activated subtype sharing the transcriptomic signatures of healthy neutrophils; and a tumor-specific subtype. This signature is conserved in murine and human cancer, across different tumor types. In colorectal cancer metastases, neutrophils are more heterogeneous, exhibiting additional transcriptomic subtypes. Pseudotime analysis implicates IL1β/CXCL8/CXCR2 axis in the progression of neutrophils from health to cancer and metastasis, with effects on T-cell effector function. Functional analysis of neutrophil-tumoroid cocultures and T-cell proliferation assays using orthotopic metastatic mouse models lacking Cxcr2 in neutrophils support our transcriptional analysis. We propose that the emergence of metastatic-specific neutrophil subtypes is driven by the IL1β/CXCL8/CXCR2 axis, with the evolution of different transcriptomic signals that impair T-cell function at the metastatic site. Thus, a better understanding of neutrophil transcriptomic programming could optimize immunotherapeutic interventions into early and late interventions, targeting different neutrophil states. SIGNIFICANCE We identify two recurring neutrophil populations and demonstrate their staged evolution from health to malignancy through the IL1β/CXCL8/CXCR2 axis, allowing for immunotherapeutic neutrophil-targeting approaches to counteract immunosuppressive subtypes that emerge in metastasis.
Collapse
|
research-article |
1 |
|