Klothilda Lim’s research while affiliated with Dana-Farber Cancer Institute and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (34)


Fig. 1. 3D cultures of HER2+ breast cancer and brain metastasis stromal cells. (A) Representative images of MDA-MB-453 cells cultured in 5% matrigel in 3D in monoculture or cocultured with indicated BMSCs for 5 d. (Scale bar, 100 μm.) (B) Heatmap showing the effect of coculture with BMSC on the viability of HER2+ breast cancer cells following treatment with the indicated treatments. Protection (pink) or sensitization (purple) to treatment is calculated by the difference between the AUC of monoculture vs. coculture with different brain stroma cells [(AUC monoculture -AUC coculture )/AUC monoculture ]. (C) Relative cell viability of the HER2+ breast cancer cells treated with increasing doses of neratinib in monoculture or in coculture with BMSCs (two-way ANOVA, mean, and SEM). (D) Schematic representation of the SynNotch reporter model (created with BioRender.com). (E) Representative flow cytometry plots depicting gating strategy. BMSCs were excluded by gating out GFP + cells. No-contact cancer cells were sorted by gating for GFP − mCherry + BFP − cells, while BMSC contact cancer cells were GFP − mCherry + BFP + .
Fig. 4. An increased glycocalyx decreases neratinib sensitivity. (A) MUC1 immunoblot analysis of cells transduced with empty vector (EV) or MUC1ΔCT. Calnexin was used as loading control. (B) Relative cell viability of cancer cells expressing EV or MUC1ΔCT construct in the presence of increasing concentration of neratinib (two-way ANOVA, mean, and SEM). (C) Immunoblot analysis of EGFR/HER2 signaling pathway components in EV or MUC1ΔCT expressing cancer cells in the presence or absence of 50 nM neratinib. (D) Schematic of experimental brain metastasis induced by cardiac injection of MDA-MB-453 cells transduced with EV or MUC1ΔCT. Expression of MUC1ΔCT was induced by doxycycline in drinking water three days after cardiac injection. Mice were treated with daily gavage of 40 mg/kg of neratinib for 1 wk, starting 3 wk after cell injection. (E) Head radiance (photon/s/cm 2 /sr) of mice at day 24 (pretreatment) and day 31 (posttreatment) treated with vehicle or neratinib (n = 5 to 6 mice per group, Mann-Whitney between pre-and posttreatment). (F) Bioluminescence images at day 24 (pretreatment) and day 31 (posttreatment).
Cancer-stromal cell interactions in breast cancer brain metastases induce glycocalyx-mediated resistance to HER2-targeting therapies
  • Article
  • Full-text available

May 2024

·

73 Reads

·

8 Citations

Proceedings of the National Academy of Sciences

Marie-Anne Goyette

·

Laura E Stevens

·

Carolyn R DePinho

·

[...]

·

Kornelia Polyak

Brain metastatic breast cancer is particularly lethal largely due to therapeutic resistance. Almost half of the patients with metastatic HER2-positive breast cancer develop brain metastases, representing a major clinical challenge. We previously described that cancer-associated fibroblasts are an important source of resistance in primary tumors. Here, we report that breast cancer brain metastasis stromal cell interactions in 3D cocultures induce therapeutic resistance to HER2-targeting agents, particularly to the small molecule inhibitor of HER2/EGFR neratinib. We investigated the underlying mechanisms using a synthetic Notch reporter system enabling the sorting of cancer cells that directly interact with stromal cells. We identified mucins and bulky glycoprotein synthesis as top-up-regulated genes and pathways by comparing the gene expression and chromatin profiles of stroma-contact and no-contact cancer cells before and after neratinib treatment. Glycoprotein gene signatures were also enriched in human brain metastases compared to primary tumors. We confirmed increased glycocalyx surrounding cocultures by immunofluorescence and showed that mucinase treatment increased sensitivity to neratinib by enabling a more efficient inhibition of EGFR/HER2 signaling in cancer cells. Overexpression of truncated MUC1 lacking the intracellular domain as a model of increased glycocalyx-induced resistance to neratinib both in cell culture and in experimental brain metastases in immunodeficient mice. Our results highlight the importance of glycoproteins as a resistance mechanism to HER2-targeting therapies in breast cancer brain metastases.

Download

Combination Therapies to Improve the Efficacy of Immunotherapy in Triple-negative Breast Cancer

September 2023

·

74 Reads

·

3 Citations

Molecular Cancer Therapeutics

Immune checkpoint inhibition combined with chemotherapy is currently approved as first-line treatment for patients with advanced PD-L1-positive triple-negative breast cancer (TNBC). However, a significant proportion of metastatic TNBC is PD-L1-negative and, in this population, chemotherapy alone largely remains the standard-of-care and novel therapeutic strategies are needed to improve clinical outcomes. Here, we describe a triple combination of anti-PDL-1 immune checkpoint blockade, epigenetic modulation thorough BET bromodomain inhibition (BBDI), and chemotherapy with paclitaxel that effectively inhibits both primary and metastatic tumor growth in two different syngeneic murine models of TNBC. Detailed cellular and molecular profiling of tumors from single and combination treatment arms revealed increased T and B cell infiltration and macrophage reprogramming from M1 to a M2 phenotype in mice treated with triple combination. Triple combination also had a major impact on gene expression and chromatin profiles shifting cells to a more immunogenic and senescent state. Our results provide strong preclinical evidence to justify clinical testing of BBDI, paclitaxel, and immune checkpoint blockade combination.


Endocrine Therapy Synergizes with SMAC Mimetics to Potentiate Antigen Presentation and Tumor Regression in Hormone Receptor–Positive Breast Cancer

July 2023

·

81 Reads

·

9 Citations

Cancer Research

Immunotherapies have yet to demonstrate significant efficacy in the treatment of hormone receptor–positive (HR+) breast cancer. Given that endocrine therapy (ET) is the primary approach for treating HR+ breast cancer, we investigated the effects of ET on the tumor immune microenvironment (TME) in HR+ breast cancer. Spatial proteomics of primary HR+ breast cancer samples obtained at baseline and after ET from patients enrolled in a neoadjuvant clinical trial (NCT02764541) indicated that ET upregulated β2-microglobulin and influenced the TME in a manner that promotes enhanced immunogenicity. To gain a deeper understanding of the underlying mechanisms, the intrinsic effects of ET on cancer cells were explored, which revealed that ET plays a crucial role in facilitating the chromatin binding of RelA, a key component of the NF-κB complex. Consequently, heightened NF-κB signaling enhanced the response to interferon-gamma, leading to the upregulation of β2-microglobulin and other antigen presentation-related genes. Further, modulation of NF-κB signaling using a SMAC mimetic in conjunction with ET augmented T-cell migration and enhanced MHC-I-specific T-cell–mediated cytotoxicity. Remarkably, the combination of ET and SMAC mimetics, which also blocks prosurvival effects of NF-κB signaling through the degradation of inhibitors of apoptosis proteins, elicited tumor regression through cell autonomous mechanisms, providing additional support for their combined use in HR+ breast cancer. Significance Adding SMAC mimetics to endocrine therapy enhances tumor regression in a cell autonomous manner while increasing tumor immunogenicity, indicating that this combination could be an effective treatment for HR+ patients with breast cancer.


A Distinct Chromatin State Drives Therapeutic Resistance in Invasive Lobular Breast Cancer

August 2022

·

68 Reads

·

14 Citations

Cancer Research

Most invasive lobular breast cancers (ILC) are of the luminal A subtype and are strongly hormone receptor-positive. Yet, ILC is relatively resistant to tamoxifen and associated with inferior long-term outcomes compared with invasive ductal cancers (IDC). In this study, we sought to gain mechanistic insights into these clinical findings that are not explained by the genetic landscape of ILC and to identify strategies to improve patient outcomes. A comprehensive analysis of the epigenome of ILC in preclinical models and clinical samples showed that, compared with IDC, ILC harbored a distinct chromatin state linked to gained recruitment of FOXA1, a lineage-defining pioneer transcription factor. This resulted in an ILC-unique FOXA1-estrogen receptor (ER) axis that promoted the transcription of genes associated with tumor progression and poor outcomes. The ILC-unique FOXA1-ER axis led to retained ER chromatin binding after tamoxifen treatment, which facilitated tamoxifen resistance while remaining strongly dependent on ER signaling. Mechanistically, gained FOXA1 binding was associated with the autoinduction of FOXA1 in ILC through an ILC-unique FOXA1 binding site. Targeted silencing of this regulatory site resulted in the disruption of the feed-forward loop and growth inhibition in ILC. In summary, ILC is characterized by a unique chromatin state and FOXA1-ER axis that is associated with tumor progression, offering a novel mechanism of tamoxifen resistance. These results underscore the importance of conducting clinical trials dedicated to patients with ILC in order to optimize treatments in this breast cancer subtype. Significance: A unique FOXA1-ER axis in invasive lobular breast cancer promotes disease progression and tamoxifen resistance, highlighting a potential therapeutic avenue for clinical investigations dedicated to this disease. See related commentary by Blawski and Toska, p. 3668.


A Distinct Chromatin State Drives Therapeutic Resistance in Invasive Lobular Breast Cancer

March 2022

·

37 Reads

Most invasive lobular breast cancers (ILC) are of the luminal A subtype and strongly hormone receptor positive. Yet, they are relatively resistant to tamoxifen and are associated with inferior long-term outcomes compared to invasive ductal cancers (IDC). In this study, we sought to gain mechanistic insights into these clinical findings that are not explained by the genetic landscape of ILC and to identify strategies to improve patient outcomes. Through a comprehensive analysis of the epigenome of ILC in pre-clinical models and clinical samples we found that compared to IDC, ILC has a distinct chromatin state that is linked to gained recruitment of FOXA1, a lineage-defining pioneer transcription factor. This results in an ILC-unique FOXA1-estrogen receptor (ER) axis that promotes the transcription of genes associated with tumor progression and poor outcomes. The ILC-unique FOXA1-ER axis leads to retained ER chromatin binding after tamoxifen treatment thereby facilitating tamoxifen resistance while remaining strongly dependent on ER signaling. Mechanistically, gained FOXA1 binding was associated with the auto-induction of FOXA1 in ILC through an ILC-unique FOXA1 binding site. Targeted silencing of this regulatory site resulted in the disruption of the feed-forward loop and growth inhibition in ILC. In summary, we show that ILC is characterized by a unique cell state and FOXA1-ER axis that dictate tumor progression and offer a novel mechanism of tamoxifen resistance. These results underscore the importance of conducting clinical trials dedicated to patients with ILC to optimize endocrine treatments in this breast cancer subtype.


BCOR and BCORL1 Mutations Drive Epigenetic Reprogramming and Oncogenic Signaling by Unlinking PRC1.1 from Target Genes

December 2021

·

64 Reads

·

28 Citations

Blood Cancer Discovery

Polycomb repressive epigenetic complexes are recurrently dysregulated in cancer. Unlike polycomb repressive complex 2 (PRC2), the role of PRC1 in oncogenesis and therapy resistance is not well-defined. Here, we demonstrate that highly recurrent mutations of the PRC1 subunits BCOR and BCORL1 in leukemia disrupt assembly of a noncanonical PRC1.1 complex, thereby selectively unlinking the RING-PCGF enzymatic core from the chromatin-targeting auxiliary subcomplex. As a result, BCOR-mutated PRC1.1 is localized to chromatin but lacks repressive activity, leading to epigenetic reprogramming and transcriptional activation at target loci. We define a set of functional targets that drive aberrant oncogenic signaling programs in PRC1.1-mutated cells and primary patient samples. Activation of these PRC1.1 targets in BCOR-mutated cells confers acquired resistance to treatment while sensitizing to targeted kinase inhibition. Our study thus reveals a novel epigenetic mechanism that explains PRC1.1 tumor-suppressive activity and identifies a therapeutic strategy in PRC1.1-mutated cancer. Significance: We demonstrate that BCOR and BCORL1 mutations in leukemia unlink PRC1.1 repressive function from target genes, resulting in epigenetic reprogramming and activation of aberrant cell signaling programs that mediate treatment resistance. Our study provides mechanistic insights into the pathogenesis of PRC1.1-mutated leukemia that inform novel therapeutic approaches. This article is highlighted in the In This Issue feature, p. 85.


Abstract P108: In vivo CRISPR screens identify E3 ligase COP1 as a modulator of macrophage infiltration and cancer immunotherapy target

December 2021

·

39 Reads

·

1 Citation

Molecular Cancer Therapeutics

Despite remarkable clinical efficacies of immune checkpoint blockade (ICB) in cancer treatment, ICB benefits in triple-negative breast cancer (TNBC) remain limited. Through pooled in vivo CRISPR knockout (KO) screens in syngeneic TNBC mouse models, we found that inhibition of the E3 ubiquitin ligase COP1 in cancer cells decreases the secretion of macrophage-associated chemokines, reduces tumor macrophage infiltration, enhances tumor immunity and ICB response. Transcriptomics, epigenomics, and proteomics analyses revealed COP1 functions through proteasomal degradation of the C/ebpδ protein. COP1 substrate TRIB2 functions as a scaffold linking COP1 and C/ebpδ, which leads to polyubiquitination of C/ebpδ. COP1 inhibition stabilizes C/ebpδ to suppress the expression of macrophage chemoattractant genes. Our integrated approach implicates COP1 as a target for improving cancer immunotherapy efficacy by regulating chemokine secretion and macrophage infiltration in the TNBC tumor microenvironment. Citation Format: Xiaoqing Wang, Collin Tokheim, Shengqing S. Gu, Binbin Wang, Qin Tang, Yihao Li, Nicole Traugh, Zexian Zeng, Yi Zhang, Boning Zhang, Jingxin Fu, Tengfei Xiao, Wei Li, Clifford Meyer, Peng Jiang, Paloma Cejas, Klothilda Lim, Henry Long, Myles Brown, X. Shirley Liu. In vivo CRISPR screens identify E3 ligase COP1 as a modulator of macrophage infiltration and cancer immunotherapy target [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr P108.


FGFR-inhibitor-mediated dismissal of SWI/SNF complexes from YAP-dependent enhancers induces adaptive therapeutic resistance

November 2021

·

282 Reads

·

30 Citations

Nature Cell Biology

How cancer cells adapt to evade the therapeutic effects of drugs targeting oncogenic drivers is poorly understood. Here we report an epigenetic mechanism leading to the adaptive resistance of triple-negative breast cancer (TNBC) to fibroblast growth factor receptor (FGFR) inhibitors. Prolonged FGFR inhibition suppresses the function of BRG1-dependent chromatin remodelling, leading to an epigenetic state that derepresses YAP-associated enhancers. These chromatin changes induce the expression of several amino acid transporters, resulting in increased intracellular levels of specific amino acids that reactivate mTORC1. Consistent with this mechanism, addition of mTORC1 or YAP inhibitors to FGFR blockade synergistically attenuated the growth of TNBC patient-derived xenograft models. Collectively, these findings reveal a feedback loop involving an epigenetic state transition and metabolic reprogramming that leads to adaptive therapeutic resistance and provides potential therapeutic strategies to overcome this mechanism of resistance. Li et al. define an adaptive resistance mechanism against FGFR inhibitor treatment in breast cancer attributed to loss of BRG1 chromatin recruitment, reactivation of YAP-dependent enhancers and upregulation of amino acid-induced mTORC1 activity.


700 Increasing MHC-I expression to potentiate immune checkpoint blockade therapy

November 2021

·

68 Reads

·

1 Citation

Background Cancer immunotherapy, especially immune checkpoint blockade (ICB) therapy, is leading to a paradigm shift in cancer treatment, as a small percentage of cancer patients have obtained durable remission following ICB treatment. Successful ICB responses rely on cancer cells presenting antigens to the cell surface via the major histocompatibility complex (MHC), which activates antigen-specific T-lymphocytes to kill cancer cells. Type-I MHC (MHC-I) is wildly expressed in all cell types and mediates the interaction with cytotoxic CD8 T cells. However, over 65% of cancer patients are estimated to show defects in MHC-I-mediated antigen presentation, including downregulation of its expression that can lead to primary or acquired resistance to ICB therapy, and therapeutic strategies to effectively restore or boost MHC-I are limited. Methods Here, we employed a CRISPR screening approach with dual-marker FACS sorting to identify factors that decouple the regulation of MHC-I and PD-L1. The experimentally validated target was used to generate a KO differential expression signature. Using this signature, we analyzed transcriptome data from drug perturbation studies to identify drugs that regulate MHC-I but not PD-L1. Finally, we validated the effect of the identified drug to enhance ICB response in a T-cell-dependent manner in vivo. Results CRISPR screens identified TRAF3, a suppressor of the NF-κB pathway, as a negative regulator of MHC-I but not PD-L1. The Traf3-knockout (Traf3-KO) gene expression signature is associated with better survival in ICB-naive cancer patients and better ICB response. We then screened for drugs with similar transcriptional effects as this signature and identified SMAC mimetics. We experimentally validated that the SMAC mimetic birinapant upregulates MHC-I, sensitizes cancer cells to T-cell-dependent killing, and adds to ICB efficacy. However, in cancer cells with high NF-κB activity, the effect of birinapant on MHC-I is weak, indicating context-dependent MHC-I regulation. Conclusions In summary, Traf3 deletion specifically upregulates MHC-I without inducing PD-L1 in response to various cytokines and sensitizes cancer cells to T-cell-driven cytotoxicity. The SMAC mimetic birinapant phenocopies Traf3-knockout and sensitizes MHC-I-low melanoma to ICB therapy. Further studies are needed to elucidate the context-dependencies of MHC-I regulation. Our findings provide preclinical rationale for treating some tumors expressing low MHC-I with SMAC mimetics to enhance sensitivity to immunotherapy. The approach used in this study can be generalized to identify other drugs that enhance immunotherapy efficacy. Acknowledgements This study was supported by grants from the NIH (R01CA234018 to XSL, R01AI137337 to BEG, P50CA101942-12 and P50CA206963 to GJF), Breast Cancer Research Foundation (BCRF-19-100 to XSL), Burroughs Wellcome Career Award in Medical Sciences (to BEG), and Sara Elizabeth O'Brien Trust Fellowship (to SG).We thank Drs. Kai Wucherpfennig and Deng Pan for their insightful suggestions on this study. Ethics Approval All mice were housed in standard cage in Dana-Farber Cancer Institute Animal Resources Facility (ARF). All animal procedures were carried out under the ARF Institutional Animal Care and Use Committee (IACUC) protocol and were in accordance with the IACUC standards for the welfare of animals.


NE carcinomas share a common chromatin state independent of their anatomical origin
a Principal component analysis (PCA) of ATAC-seq data of NECs including Merkel cell carcinoma (MCC), neuroendocrine prostate cancer (NEPC), gastrointestinal neuroendocrine carcinoma (GINE), and small-cell lung cancer (SCLC). The plot also includes prostate adenocarcinoma (PDX models and TCGA primary tissues) and lung adenocarcinoma (TCGA primary tissues). b Hierarchical clustering of the pairwise Pearson’s correlation of the ATAC-seq signal across the distinct tumor types. c Heatmap representation of the differential regions between representative ADs and NECs. Each row is a peak location and each column is a sample. Shown above each column are the composite tag density plots for the AD sites (blue) and NE sites (green). d Gene Ontology enrichment using a binomial test¹⁷ showing the pathways enriched in genes with nearby NE-specific accessible regions shown in c. e Top results from motif analysis of the NE-specific accessible regions. f Public ChIP-seq data sets showing the highest overlap with the NE-specific accessible regions as determined by CistromeDB toolkit annotated by tissue type. The TFs are ordered by the top scoring data set of each type. g Expression of NE markers and bHLH TFs across all the NEC samples in our study displayed as a heatmap. Source data are provided as a Source Data file.
NEPC shows tumor subtypes based on the differential expression of the transcription factors ASCL1 and NEUROD1
a Principal component analysis of ATAC-seq data from NEPC and ADPC PDXs. Samples are color coded by the dominant TF expressed in that sample. b The left side of the heatmap (red) displays the differential ATAC-seq regions identified between NEPC subtypes. There are 12,751 NEUROD1-specific regions (top) and 8950 ASCL1-specific (bottom) ATAC sites. The right side of the heatmap shows the ChIP-seq data at the same sites for ASCL1 (green) and NEUROD1 (blue) for the indicated samples. c Association between differential ATAC-accessible sites and differential gene expression. Each volcano plot depicts RNA-seq log2-fold change (x-axis) and p-value adjusted for multiple hypothesis testing calculated by DESeq2 using a Wald’s test (y-axis). Each dot represents one gene: green indicates a differential ATAC peak is within 50 kB of the gene and orange indicates there is no such peak. Left: ASCL1-specific accessible regions and genes upregulated in ASCL1 subtype; (right) NEUROD1-specific accessible regions and genes upregulated in NEUROD1 subtype. d GSEA pathway analysis of genes enriched in the ASCL1 subtype (green) and the NEUROD1 subtype (blue) (**q-value < 0.001, *q-value < 0.05). e Signal distribution of H3K27ac marked enhancers from representative cases of the ASCL1 subtype (top) and NEUROD1 subtype (bottom). The bars in the lower right of each plot identify the subset of enhancers known as super-enhancers defined by the ROSE algorithm; 693 were identified in LuCaP 93 (ASCL1) and 766 in LuCaP 173.1 (NEUROD1). Super-enhancers nearby selected genes are indicated by the arrows. f Representative IGV tracks at the ASCL1 and NEUROD1 gene loci. ATAC-seq tracks are in red, ASCL1 ChIP-seq in green, NEUROD1 ChIP-seq in blue, and H3K27ac in gray. The loci are marked by subtype-specific super-enhancers with preferential binding of their respective TF. g Circuits of lineage transcription factors specific for the ASCL1 subtype (green) and NEUROD1 subtype (blue). Source data are provided as a Source Data file.
Single-cell analysis reveals that NEPC subtypes co-exist in human metastasis and contribute to inter- and intra-tumoral heterogeneity
a Plot of ASCL1 and NEUROD1 expression in NEPC tissues from a clinical cohort¹⁶. TPM: transcripts per million. b Representative immunostaining of FLM3 (ASCL1 staining in the top panel and NEUROD1 staining in the middle panel) showing intratumor heterogeneity. c Hematoxylin and eosin staining of the same field illustrates the distinct histologies for the two subpopulations. d Combined analysis of the scATAC-seq and snRNA-seq in FLM3 (left). Markers specific for normal cell populations enabled assignment of clusters: 1, vascular cells; 2, stromal cells; 3, hepatic cells; 4, monocytes. Accessibility at the top 30 differential ATAC-seq regions between ASCL1 and NEUROD1 subtypes identified by bulk analysis (top right). Analysis of ASCL1 and NEUROD1 expression in the snRNA-seq analysis (bottom right). This analysis matches cells with TF expression and the corresponding differential DNA accessibility for each subtype. e tSNE analysis of the combined FLM3 (blue) and FLM5 (black) scATAC-seq data (left). The other three plots show accessibility at INSM1 promoter (NE marker) and the differential accessibility at ASCL1 promoter and NEUROD1 promoter. f Projection of the aggregated scATAC-seq clusters for FLM3 and 5 (light brown dots) within the PCA space defined in Fig. 2a. Source data are provided as a Source Data file.
The NEPC subtypes are distinct clones
a Genome-wide CNV profiles inferred from the scATAC-seq clusters in FLM3 and FLM5. Black dots are values in 1 MB regions and the red line is the result of running a segmentation algorithm on the data (see “Methods”). Arrows point to differences seen in CNVs across the clusters. b Sample pairwise Pearson’s correlation of the CNV profiles. c Summary heatmap of the scATAC-seq-inferred CNV alterations across all of the patient samples (blue represents losses and red represents gains). d Heatmap of the single-cell CNV analysis of FLM3 where each column is a 2 MB bin tiled across the genome and the rows are individual cells that have been clustered with K-means. Arrows point to CNV differences observed here and in the cluster level analysis. e tSNE plot of FLM3 scATAC-seq data colored by the cluster each cell was partitioned into by the inferred CNV alterations. Those three clusters clearly correspond to NEUROD1 (blue), ASCL1 (green), and normal cells (gray). Source data are provided as a Source Data file.
Subtype heterogeneity and epigenetic convergence in neuroendocrine prostate cancer

October 2021

·

474 Reads

·

104 Citations

Neuroendocrine carcinomas (NEC) are tumors expressing markers of neuronal differentiation that can arise at different anatomic sites but have strong histological and clinical similarities. Here we report the chromatin landscapes of a range of human NECs and show convergence to the activation of a common epigenetic program. With a particular focus on treatment emergent neuroendocrine prostate cancer (NEPC), we analyze cell lines, patient-derived xenograft (PDX) models and human clinical samples to show the existence of two distinct NEPC subtypes based on the expression of the neuronal transcription factors ASCL1 and NEUROD1. While in cell lines and PDX models these subtypes are mutually exclusive, single-cell analysis of human clinical samples exhibits a more complex tumor structure with subtypes coexisting as separate sub-populations within the same tumor. These tumor sub-populations differ genetically and epigenetically contributing to intra- and inter-tumoral heterogeneity in human metastases. Overall, our results provide a deeper understanding of the shared clinicopathological characteristics shown by NECs. Furthermore, the intratumoral heterogeneity of human NEPCs suggests the requirement of simultaneous targeting of coexisting tumor populations as a therapeutic strategy.


Citations (25)


... The majority of patients with advanced BM are treated with multiple lines of therapy and experience several problems, such as drug resistance, which greatly increases the difficulty of treatment (89). Combination therapy is particularly important. ...

Reference:

Anti‑HER2‑targeted therapies for the treatment of advanced HER2‑positive breast cancer with brain metastases (Review)
Cancer-stromal cell interactions in breast cancer brain metastases induce glycocalyx-mediated resistance to HER2-targeting therapies

Proceedings of the National Academy of Sciences

... To investigate cellular niches and intercellular crosstalk across development and cancer progression of the breast tissue we used the MMTV-PyMT mouse model, which mimics the different stages of human ductal breast cancer including hyperplasia, neoplasia, early and advanced carcinoma and metastasis. This model displays immune infiltration, progressive loss of estrogen and progesterone receptors and despite the overexpression of HER2, it is widely used as a triple-negative breast cancer model 4,14 . We first explored the processes inducing physiological development of the mammary gland, by purifying single cells from oncogene-carrier (PyMT + ) and noncarrier control (PyMT − ) tissues, during the developmental stages: pre-puberty (10 days, 10 d) and puberty (3 weeks, 3 w). ...

Combination Therapies to Improve the Efficacy of Immunotherapy in Triple-negative Breast Cancer

Molecular Cancer Therapeutics

... Immunotherapies aimed at reactivating exhausted T cells in the tumor microenvironment have revolutionized cancer treatment, but their success has been limited for solid tumors, including mild response rates in endocrineresistant HR+ metastatic BC. [22][23][24][25][26] Although immune checkpoint blockade has shown moderate success in triple-negative BC, it has only yielded mild survival benefits for patients with HR+BC, likely because their tumors have low numbers of tumor-infiltrating lymphocytes (TILs) and weak T cell activation, rendering them less responsive to this immunotherapeutic approach. 27 Robust antitumor responses by CD8+T cells require tumor-associated antigens or neoantigens presented on MHC class I. Previous studies have linked steroid HRs, such as glucocorticoid receptor (GR) and ER, to APP/MHC class I downregulation in pancreatic cancer 28 and BC, respectively. 29 Given the structural similarities between GR and PR, we sought to investigate whether progesterone regulates APP/MHC class I through PR. ...

Endocrine Therapy Synergizes with SMAC Mimetics to Potentiate Antigen Presentation and Tumor Regression in Hormone Receptor–Positive Breast Cancer

Cancer Research

... Tamoxifen is a selective estrogen receptor modulator, which is widely used in the treatment of ER-positive breast cancer [240]. However, recent studies have pointed out that invasive lobular breast cancer (ILC) has higher tamoxifen resistance than invasive ductal carcinoma (IDC) [241]. Agostina Nardone et al.'s experiment suggests that this resistance is due to the abnormal impact of ILC's unique chromatin state on the FOXA1-ER axis. ...

A Distinct Chromatin State Drives Therapeutic Resistance in Invasive Lobular Breast Cancer
  • Citing Article
  • August 2022

Cancer Research

... The advent of genome sequencing has significantly accelerated the understanding of the molecular pathogenesis of leukemia at an unprecedented rate [3,4]. In fact, mountains of evidence prove that pathogenesis of myeloid leukemia is largely attributed to chromosomal translocations and gene mutations, which result in the generation of abnormal genes known as oncogenes [5][6][7]. Notably, oncogenes are traditionally defined as genes with dominant traits that contribute to the transforming phenotype of cells through encoding abnormal proteins (i.e. oncogenic proteins) that impede normal differentiation, proliferation, and self-renewal of hematopoietic precursor cells [8,9]. ...

BCOR and BCORL1 Mutations Drive Epigenetic Reprogramming and Oncogenic Signaling by Unlinking PRC1.1 from Target Genes
  • Citing Article
  • December 2021

Blood Cancer Discovery

... 3. The current microenvironment recognition method is mainly established by single-cell transcriptome and antibody-dependent multidimensional imaging. However, immune cells are functionally regulated by numerous transcription factors (21), hence, other single-cell strategies such as scATAC-seq, scChIP-seq, and the emerging CRISPR screening (72,73) will be useful for breast TME analysis. 4. TME analysis needs to pay more attention to the combination of cell function and spatial distribution in the future. ...

Abstract P108: In vivo CRISPR screens identify E3 ligase COP1 as a modulator of macrophage infiltration and cancer immunotherapy target
  • Citing Conference Paper
  • December 2021

Molecular Cancer Therapeutics

... In addition, the loss or downregulation of MHC-I antigen presentation is a major reason for resistance to immune checkpoint therapy in cancer patients, with studies predicting that over 65% of cancer patients have MHC-I-related defects [191]. As MHC-I and the immune inhibitory signal protein PD-L1 are often regulated by common molecular signals, upregulation of MHC-I may also be accompanied by upregulation of PD-L1 and inhibition of immune response. ...

700 Increasing MHC-I expression to potentiate immune checkpoint blockade therapy

... Currently, the FDA has approved a variety of FGFR inhibitors on the market for the treatment of solid tumours (58,59). Although FGFR-targeted therapies have not yet been approved for the treatment of breast cancer, many trials have begun to use FGFR inhibitors for the treatment of breast cancer, including TNBC (7,42,60,61). FGFR inhibitors can be broadly classified into three categories: (i) smallmolecule TKIs that selectively target the kinase structure of the FGFRs; (ii) small-molecule, multi-targeted tyrosine kinase inhibitor TKIs that block tyrosine kinase activity; and (iii) monoclonal antibody mAbs that block FGFRs as well as carry their ligands. ...

FGFR-inhibitor-mediated dismissal of SWI/SNF complexes from YAP-dependent enhancers induces adaptive therapeutic resistance

Nature Cell Biology

... Chromatin landscape of t-NEPC was shown to exhibit similarities to SCLC with distinct subtypes based on the expression of the neuronal transcription factors ASCL1 and neurogenic differentiation factor 1 (NEUROD1). Single-cell sequencing of human clinical samples revealed a more intricate tumor structure, where these subtypes coexist as separate subpopulations within individual tumors [56]. Recent single-cell analyses have revealed more complex subpopulations, including cells co-expressing achaete-scute family BHLH transcription factor 2 (ASCL2) and POU class 2 homeobox 3 (POU2F3). ...

Subtype heterogeneity and epigenetic convergence in neuroendocrine prostate cancer

... In addition to the effects of TAMs on cancer cells, the latter reciprocally polarize TAMs towards states in favor of tumor progression by producing cytokines, chemokines, and metabolites. For instance, substances such as succinate, histamine, CSF1, E3 ubiquitin protein ligase COP1, and β-glucosylceramide released by cancer cells can modulate metabolic state and induce ER stress of TAMs, thereby escalating the generation of pro-tumor TAMs [58,[101][102][103][104][105]. In glioblastoma, periostin secreted by tumor stem cells recruited monocyte-derived macrophages from peripheral blood and polarized them into M2-like TAMs to promote tumorigenesis [106]. ...

In vivo CRISPR screens identify the E3 ligase Cop1 as a modulator of macrophage infiltration and cancer immunotherapy target
  • Citing Article
  • September 2021

Cell