Stacy Moulder’s research while affiliated with Eli Lilly and Company 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 (126)


Abstract 6438: Decoding the archetypes and ecotypes of triple-negative breast cancer in responses to chemotherapy
  • Article

April 2025

·

9 Reads

Yun Yan

·

Yiyun Lin

·

Tapsi Kumar

·

[...]

·

Triple-negative Breast Cancer (TNBC) is an aggressive subtype of breast cancer. The pillar of treatment is chemotherapy, but only half of the patients have a complete response and good survival. To resolve inter- and intra-tumoral heterogeneity and determine their clinical associations, we performed single-cell RNA-sequencing and spatial transcriptomics methods including Visium and Xenium on treatment-naïve samples of TNBC patients in the ARTEMIS clinical trial. We find that TNBC was classified into 4 major archetypes at patient level: luminal secretory-associated, basal-associated, immunoreactive, and luminal androgen receptor. At cell level, cancer cells exhibited intratumoral heterogeneity in 13 gene expression metaprograms. The TNBC tumor microenvironment (TME) consisted of 49 distinct immune and stromal cell states, many of which were reprogrammed relative to normal breast tissues from disease-free women. We further identified 8 ecotypes of cancer cells and TME cell states that co-occurred among patients and were associated with specific archetypes and chemotherapy response groups. In contrast to previous work on T-cells, our data showed the importance of macrophage cell states and cancer cell metaprograms for interferon signaling, HLA expression and cell cycle activity that were associated with chemotherapy response. To facilitate a clinical application, we developed a 13-gene-based model to predict response. Collectively, this study provides new insights into the natural biology of untreated TNBC tumors and their association with chemotherapy response. Citation Format Yun Yan, Yiyun Lin, Tapsi Kumar, Shanshan Bai, Aatish Thennavan, Jianzhuo Li, Tuan Tran, Min Hu, Mitchell Rao, Anna Casasent, Elizabeth Ravenberg, Gaiane Margishvili Rauch, Alyson Clayborn, Debu Tripathy, Alastair Thompson, Bora Lim, Lei Huo, Stacy Moulder, Clinton Yam, Nicholas Navin. Decoding the archetypes and ecotypes of triple-negative breast cancer in responses to chemotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2025; Part 1 (Regular Abstracts); 2025 Apr 25-30; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2025;85(8_Suppl_1):Abstract nr 6438.


Clinical activity of L-NMMA combined with docetaxel in chemorefractory MpBC patients
A Waterfall Plot showing change in target lesion/tumor volume, best response (RECIST), and baseline iNOS H-score expression status in the 13 patients with available data from post-baseline assessments. PD progressive disease, SD stable disease, PR partial response, and CR complete response. iNOS H-Score Metric (0–50: None, 51–100: low, 101–150: Moderate, >150: High) B Representative mammogram images at baseline (BL) and end of cycle 6 for Patient 100-048 (CR), showing shrinkage in target lesion size. C Hematoxylin and Eosin staining of BL tumor from 100-048 showing metaplastic squamous differentiation and end-of-treatment (EOT) tissue showing residual keratinized and fibrotic tissue. Scale bars represent 100 µM. Representative images of n = 3 tissue samples. D BL IHC staining of iNOS, PTEN, and phospho-Akt of tumor tissue from 100-048 and associated EOT iNOS, pAkt, and PTEN staining in residual tissue. Scale bars represent 100 µM. E iNOS H-score for BL and EOT tumors in indicated responders and non-responders. BL and EOT H-scores are indicated by red and blue box and whisker plots, respectively. R2-R4 indicates responder patients, and NR1-NR6 indicate non-responder patients. H-score analysis of 4–6 images per slide captured from tissues to cover entire tissue bed. The center bar indicates median, bounds of box represent lower and upper quartiles, and the whiskers indicate minimum and maximum of the dataset for each group. Statistical analysis by two-sided Student’s t test. FNOS2 alteration frequencies in various cancers from cBioPortal Combined Study Dataset (n = 179290). GNOS2 normalized gene expression in MpBC/metaplastic-like TNBC (n = 41, red dots) and non-MpTNBC (n = 137, blue dots) from the ARTEMIS dataset. The dot and error bars represent mean ± SD. Statistical analysis by two-sided Student’s t test. H Kaplan-Meier metastasis-free survival analysis of MpBC and non-MpTNBC tumors based on expression status [high (red line)/low (blue line)] of NOS2 from ARTEMIS dataset. I Gene set enrichment analysis of the top represented upregulated hallmark gene sets based on normalized enrichment score (NES) from RNA-sequencing data from TCGA in human MpBC tumors (n = 14) compared to invasive ductal carcinoma tumors (n = 814).
Co-expression of iNOS and phospho-Akt is predominant in MpBC
A Network of protein interactions with iNOS generated by STRING analysis. Each network node represents one gene. Red line highlight interaction between NOS2 and AKT1. B Mutual exclusivity analysis of cBioPortal combined dataset of all cancers (n = 179,290) demonstrated a significant tendency toward co-occurrence for NOS2 with PIK3CA, AKT1, PTEN, and RPL39 genomic alterations. mRNA expression and protein/phosphoprotein data were selected in analysis. Statistical analysis using one-sided Fisher’s Exact Test. C Immunoblotting analysis of iNOS, PTEN, phospho-Akt (Thr308 and Ser473) in a panel of breast cancer cell lines with known PIK3CA/PIK3R1 and PTEN mutation status. HSP90 was used as a loading control. Human mammary epithelial cell line (MCF-10A), ER+ breast cancer, HER2+ breast cancer, non-metaplastic TNBC, and metaplastic TNBC cell lines are indicated in black, red, blue, green, and purple, respectively. Blots shown are representative images of n = 3 biological replicates. D Comparison of normalized iNOS and phospho-Akt (Thr308 and Ser473) protein levels among MpBC, TNBC, ER+, and HER2+ breast cancer cell lines. Each dot represents a cell line showing a representative experiment, statistical analysis by two-sided Student’s t test comparing differences in protein expression ratios of MpBC to TNBC cell lines. Bars and error bars represent Mean ± SD. E Droplet digital PCR analysis of RPL39 A14V and PIK3CA hotspot mutations (E542, E545K, H1047L, H1047R) and iNOS/phospho-Akt (Ser473) immunohistochemical expression status in PDXs of TNBC (n = 12), ER+ (n = 5), HER2+ (n = 3), and MpBC (n = 6). Green bars represent PDX models that express the specifically indicated mutation in the column. Blue bars (depending on gradient) represent the expression of phospho-Akt from low to high expression, a marker of PI3K/Akt activation. Orange bars (depending on gradient) represent low to high iNOS expression. F Results of two-sided Fisher’s exact test comparing MpBC mutation status of RPL39 A14V and PIK3CA hotspot mutations in other breast cancer subtypes. G mRNA expression of NOS2 in all breast cancer PDX models. Values were compared to ΔCT value from PDX 4913 (TNBC) as a control that was set to 1 and represent the mean ± SD of three biological replicates.
Pan-NOS inhibitor L-NMMA acts synergistically with isoform α-specific PI3K inhibitor alpelisib in MpBC cell lines with PIK3CA/PIK3R1 mutations
A Four MpBC cell lines were treated with dimethyl sulfoxide (DMSO), or increasing concentrations of L-NMMA, alpelisib, or combination for 72 hours. Cell growth was evaluated using Sulforhodamine B (SRB) assay. Sensitivity of MpBC cell lines to L-NMMA alone, alpelisib alone, or L-NMMA combined with alpelisib was compared to vehicle control treated MpBC cells. Cell viability (left) and the combination index (right) are shown for each of these four cell lines and determined by CalcuSyn software. Fa, fraction affected. Bars and error bars represent mean ± SD of three biological replicates. B Protein levels of iNOS, phospho-Akt (Ser473/Thr308), total Akt, and GAPDH in SUM159 control and different NOS2 knockout (NOS2KO) clones. NOS2KO clones were developed using iNOS Double Nickase CRISPR plasmids. Blots shown are representative images of n = 2 biological replicates. C Cell Glo Titer Cell Viability Assay results of SUM159 control and NOS2KO clones treated with alpelisib at varied concentrations for 72 hours. IC50 values were determined by GraphPad Prism software. D Immunoblotting of iNOS and PI3K signaling markers in SUM159 (PIK3CA mutated), Hs578T (PIK3R1 mutated), BT549 (PTEN-deleted) cell lines treated for 24 hours with DMSO control, 4 mM L-NMMA, 5 µM alpelisib, and L-NMMA combined with alpelisib. Blots shown are representative images of n = 3 biological replicates. Densitometry quantification values were determined using ImageLab software (Biorad) and found in Supplementary Fig. 3. E Immunoblotting of S-nitrosoglutathione reductase (GSNOR) and tubulin loading control of MpBC cell lines HCC1806, BT549, Hs578T, and SUM159 with densitometry analysis indicated in a bar graph. The blots shown are representative images of n = 2 biological replicates. F Extent of DNA damage, quantified by the comet tail moment in the neutral comet assay. Statistical analysis by two-sided Student’s t test. n = 3 biological replicates per condition, 40 comets counted per biological replicate.
NOS inhibition augments PI3K inhibitor and taxane treatment in vivo
A Schematics representing the MpBC PDX (BCM-3807, BCM-4664, PIM-010, and PIM-084) experimental design. PDXs derived from human MpBCs were transplanted into cleared mammary fat-pad of female NSG mice. When tumors reached 150–200 mm³, mice were randomized to receive vehicle control, NOS inhibition therapy (L-NMMA [400 mg/kg oral gavage on day 1, 200 mg/kg oral gavage on days 2–5] + amlodipine [10 mg/kg intraperitoneal injection on days 1–5]), PI3K inhibitor alpelisib (35 mg/kg oral gavage on days 1–5), or the combination of both therapies as indicated. Caliper measurements were taken twice a week. Days in which mice were treated with therapies are indicated in red and rest days are indicated in green. B–E Mean tumor volume and corresponding waterfall plots demonstrating maximal treatment response to single-agent or combination therapy in four MpBC PDX models ([BCM-3807, n = 6], [PIM-010, n = 7], [PIM-084, n = 5], and [BCM-4664, n = 6]). Average tumor volume [0.5 × (mm long dimension) × (mm short dimension)²] and data points are mean tumor volume ± SEM. Statistical analysis for b–e by two-sided Student’s t test (*p ≤ 0.05, **p ≤ 0.01). Each bar in waterfall is derived from the maximal response of a single tumor-bearing mouse to therapy. Lines and bars in the plots indicated in black represent vehicle control, blue represent L-NMMA single-agent therapy, red represent alpelisib single-agent therapy, and gray indicate dual-agent therapy. P values: BCM-3807 (control vs L-NMMA+alpelisib [p = 0.0211], L-NMMA vs L-NMMA+alpelisib [p = 0.0398], alpelisib vs L-NMMA+alpelisib [p = 0.0456]), PIM-010 (control vs L-NMMA+alpelisib [p = 0.006], L-NMMA vs L-NMMA+alpelisib [p = 0.0231], alpelisib vs L-NMMA+alpelisib [p = 0.0079]), PIM-084 (control vs L-NMMA+alpelisib [p = 0.0231], alpelisib vs L-NMMA+alpelisib [p = 0.016]), BCM-4664 (control vs L-NMMA+alpelisib [p = 0.0052], L-NMMA vs L-NMMA+alpelisib [p = 0.0254], alpelisib vs L-NMMA+alpelisib [p = 0.1275]). F, G Tumor volumes of BCM-4664 (F) and BCM-3807 (G) tumors treated with vehicle control (black), docetaxel (purple), or combination therapy (docetaxel + NOS inhibition therapy [blue], docetaxel + alpelisib [green], and docetaxel + NOS inhibition therapy + alpelisib [red]). When tumors reached 150–200 mm³, they were randomized into the respective treatment arms. Each graph line represents a replicate/treatment arm. H, I Kaplan–Meier survival curves of model BCM-4664 (H) and BCM-3807 (I) treated with vehicle control, docetaxel, or combination therapy (dual/triple combination). An event was scored when a tumor reached 1200 mm³ or from death. Statistical analysis using Log-rank (Mantel–Cox) test. P values: BCM-4664 (docetaxel+alpelisib vs triple combination [p = 0.0432], docetaxel+L-NMMA vs triple combination [p = 0.0007], BCM-3807 (docetaxel vs. triple combination [p = 0.0192].
NOS inhibition induces epithelial-to-mesenchymal transition reversal in MpBC
ATop GSEA pathways by normalized enrichment score (NES) from the Hallmark and Reactome collections, enriched in control (blue) and L-NMMA+alpelisib (red) treated PDX tumors. Light blue bars indicate non-significant pathways. B Representative immunofluorescence images of BCM-3807 tumors evaluated for C E-cadherin and Zeb1 protein expression. n = 3 biological replicates per condition. Five images at ×10 magnification per biological replicate covering the complete tissue bed were utilized for analysis. Scale bars represent 200 µM. Black, blue, red, and gray bars represent vehicle control, L-NMMA, alpelisib, and L-NMMA+alpelisib, respectively. D Immunoblotting of EMT markers in Hs578T (PIK3R1 mutated) cell lines treated with DMSO control, L-NMMA, alpelisib, and L-NMMA+alpelisib for 4–24 hours. E Morphology of SUM159 control cells and NOS2KO clone cells. ×20 magnification and scale bars represent 200 µM. G Volcano Plot representing global transcriptional changes comparing SUM159 control cells and NOS2KO clone cells. Each data point represents a gene regulated by AP-1 transcription factor family. Differentially expressed genes (p < 0.05) with a log2 fold change >1 are upregulated genes (red dots), and less than −1 are downregulated genes (green dots). Statistical analysis was performed using the Wald test. H Significantly differentially expressed genes clustered by their gene ontology (GO) with an adjusted P value < 0.05, tested using two-sided Fisher exact test (GeneSCF v1.1-p2). mRNA expression of TGFB1I and LCN2J from Parental SUM159 cells and NOS2KO clone cells and TGFB1L and LCN2M in SUM159 treated with non-targeting control siRNA and siRNAs specific to XBP1, CREB3, FOS, and JUN. Immunoblots of F EMT and iNOS-associated proteins, K LCN2, TGFβ [active form], N phospho-c-Jun (Ser63/Ser73), O S-nitrosylation of JNK (SNO-JNK), and HSP90 loading control in Parental SUM159 cells and NOS2KO clone cells. For C, I, J, K–O, Statistical analysis by two-sided Student’s t test. Bars and error bars represent the mean ± SD of three biological replicates. For all Blots, images shown are representative of n = 3 biological replicates, and graph represents SNO-JNK/JNK protein expression ratios from n = 3 biological replicates.

+2

NOS inhibition sensitizes metaplastic breast cancer to PI3K inhibition and taxane therapy via c-JUN repression
  • Article
  • Full-text available

December 2024

·

83 Reads

·

1 Citation

Metaplastic breast cancer (MpBC) is a highly chemoresistant subtype of breast cancer with no standardized therapy options. A clinical study in anthracycline-refractory MpBC patients suggested that nitric oxide synthase (NOS) inhibitor NG-monomethyl-l-arginine (L-NMMA) may augment anti-tumor efficacy of taxane. We report that NOS blockade potentiated response of human MpBC cell lines and tumors to phosphoinositide 3-kinase (PI3K) inhibitor alpelisib and taxane. Mechanistically, NOS blockade leads to a decrease in the S-nitrosylation of c-Jun NH2-terminal kinase (JNK)/c-Jun complex to repress its transcriptional output, leading to enhanced tumor differentiation and associated chemosensitivity. As a result, combined NOS and PI3K inhibition with taxane targets MpBC stem cells and improves survival in patient-derived xenograft models relative to single-/dual-agent therapy. Similarly, biopsies from MpBC tumors that responded to L-NMMA+taxane therapy showed a post-treatment reversal of epithelial-to-mesenchymal transition and decreased stemness. Our findings suggest that combined inhibition of iNOS and PI3K is a unique strategy to decrease chemoresistance and improve clinical outcomes in MpBC.

Download

Fig. 1 | Study design. Schematic representation of the ARTEMIS clinical trial showing PDX models established at clinical presentation (Pre), after four cycles of Adriamycin and cyclophosphamide (AC, Mid), and after completion of NACT (Post). Patient IDs (PiD) and Patient in Mouse (PIM) IDs that were established at each time point are shown below. Color code denotes matched patient/PDX serial models.
Fig. 2 | Longitudinal analysis of drug response profiles. a Heatmap showing the pairwise-Pearson correlation of cell lines using the z-normalized AUCs of the filtered drug profile. Top bar identifies the timepoint (Time) and Patient ID (PiD) using the color code denoted in the figure. b Volcano plot of the mean difference in the AUC (Time x -T 0 ) by the log significance of the interaction determined from the linear model. The top 5 drugs are highlighted, in addition to components of the NACT regimen, which show an acquired resistance. Dotted lines show cut offs for À log 10 p À value À Á ≥ 1:3& absðmean ΔAUC ð Þ Þ≥ 0:05) used to prioritize the top compounds from the screen. c Heat map showing the shifts in AUC values for each matched pair. Y-axis shows the Patient ID number grouped by Mid-to-Pre and Postto-Pre comparisons. X-axis is grouped according to pre-established mechanistic class and annotated by target.
Fig. 3 | Longitudinal analysis of transcriptomic profiles. a Heatmap of the pairwise-Pearson correlation of cell lines using the z-normalized ComBat-adjusted TPM. Top bar denotes the timepoint and Patient ID using the color code denoted in the figure. b Heterogeneous network representation generated by performing genepathway enrichment analysis using pathfindR. Significantly altered genes are represented by circles, while pathway annotations are shown as squares, with connecting lines to member genes. Pathways and genes related to protein homeostasis are highlighted in yellow, while those related to RNA homeostasis are in green.
Fig. 5 | NEDD8 and SUMO1 tissue labeling. Representative images of NEDD8 (a) and SUMO1 (b) IHC in PDX tumor sections. Images shown are ×10 magnification, insets are ×20 magnification, and scale bars are 100 µm. c Bar chart showing PDX response to pevonedistat via normalized tumor to control ratios (T/C). Bars represent mean ± standard deviation. H-scores for NEDD8 (d) and SUMO1 (e) intensity, combining percent of tumor cells staining and intensity of staining. Images were quantified from three tumors per PDX model. Bars represent the mean ± SEM. f Dot plot showing the correlation of pevonedistat response (1-T/C) of SUMO1 H-Score (black) or NEDD8 H-score (Red). r = Spearman correlation coefficient, p = p value calculated from a two-tailed test.
Fig. 6 | Perturbational response to pevonedistat in vivo. a Schematic of the FNA time series design. b Heatmap of the top ssGSEA pathways that showed a significant (p < 0.001) difference in the time series of the responsive class but not (p > 0.05) in the vehicle-treated controls. Top bar indicates class of pevonedistat response (responsive, non-responsive), PDX_ID, timepoint, and treatment.
Targeting neddylation and sumoylation in chemoresistant triple negative breast cancer

May 2024

·

42 Reads

·

2 Citations

npj Breast Cancer

Triple negative breast cancer (TNBC) accounts for 15–20% of breast cancer cases in the United States. Systemic neoadjuvant chemotherapy (NACT), with or without immunotherapy, is the current standard of care for patients with early-stage TNBC. However, up to 70% of TNBC patients have significant residual disease once NACT is completed, which is associated with a high risk of developing recurrence within two to three years of surgical resection. To identify targetable vulnerabilities in chemoresistant TNBC, we generated longitudinal patient-derived xenograft (PDX) models from TNBC tumors before and after patients received NACT. We then compiled transcriptomes and drug response profiles for all models. Transcriptomic analysis identified the enrichment of aberrant protein homeostasis pathways in models from post-NACT tumors relative to pre-NACT tumors. This observation correlated with increased sensitivity in vitro to inhibitors targeting the proteasome, heat shock proteins, and neddylation pathways. Pevonedistat, a drug annotated as a NEDD8-activating enzyme (NAE) inhibitor, was prioritized for validation in vivo and demonstrated efficacy as a single agent in multiple PDX models of TNBC. Pharmacotranscriptomic analysis identified a pathway-level correlation between pevonedistat activity and post-translational modification (PTM) machinery, particularly involving neddylation and sumoylation targets. Elevated levels of both NEDD8 and SUMO1 were observed in models exhibiting a favorable response to pevonedistat compared to those with a less favorable response in vivo. Moreover, a correlation emerged between the expression of neddylation-regulated pathways and tumor response to pevonedistat, indicating that targeting these PTM pathways may prove effective in combating chemoresistant TNBC.


Abstract 6933: Decoding the archetypes and eco-hubs of triple-negative breast cancer in responses to chemotherapy

March 2024

·

14 Reads

Triple-negative breast cancer (TNBC) is a highly aggressive type of breast cancer commonly treated with neoadjuvant chemotherapy (NAC). While about 50% of patients achieve a pathological complete response (pCR), the remainder often develop resistance and metastasis, leading poor survivals. Currently, the factors linking pre-treatment cancer cells and the tumor microenvironment (TME) to NAC response are unclear. To investigate this question, we conducted single-cell RNA sequencing on fresh pre-treatment core biopsy samples collected from patients in the ARTEMIS clinical trial. We identified 13 gene expression meta-programs of cancer cells and about 50 cell states of immune and stromal cell types. Based on the cancer cells alone, we identified four TNBC archetypes at patient-level: luminal secretory-associated (LSA), basal-associated (BA), immunoreactive (IR), and luminal androgen receptor (LAR). Notably, the archetype BA and IR were associated with non-pCR and pCR, respectively. Additionally, we found the TNBC ecosystem was composed of eight eco-hubs, reflecting the co-occurrence patterns of cancer and TME cell states. These eco-hubs revealed different cell community across archetypes and NAC outcomes. For example, an eco-hub with a co-occurrence of interferon response-related cancer cells and immune cells was prevalent in the archetype IR and pCR patients. To facilitate a clinical application, we further developed a 13-gene-based machine learning model to predict NAC response. In summary, these results provide new insights into the cellular determinants of TNBC biology and chemotherapeutic response. Citation Format: Yun Yan, Yiyun Lin, Tapsi Kumar, Shanshan Bai, Jianzhuo Li, Tuan Tran, Min Hu, Elizabeth Ravenberg, Maia Rauch, Alyson Clayborn, Alastair Thompson, Bora Lim, Lei Huo, Stacy Moulder, Clinton Yam, Nicholas Navin. Decoding the archetypes and eco-hubs of triple-negative breast cancer in responses to chemotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 6933.


Abstract 6620: PDX models of TNBC established from pre- and post-therapy tumors identify vulnerabilities of resistant disease

March 2024

·

12 Reads

Chemotherapy, along with the PD1 inhibitor pembrolizumab, is the recommended standard of care for patients with primary triple negative breast cancer (TNBC). Nearly half of TNBC patients treated with standard neoadjuvant chemotherapy (NACT) have excellent responses. However, those patients with significant residual cancer burden (RCB) after NACT are at high risk of recurrence or metastatic relapse within two years. Due to the challenges posed by the inter- and intratumoral heterogeneity of TNBC, it is critical to develop appropriate models for predicting and overcoming therapy resistance. We established a collection of 92 orthotopic patient-derived xenograft (PDX) models of TNBC from 84 patients before, during, and after NACT while they were enrolled in the ARTEMIS trial (NCT02276443) at MD Anderson Cancer Center. Serial biopsies were obtained from patients prior to treatment (pre-NACT), from poorly responsive disease after four cycles of Adriamycin and cyclophosphamide (AC, mid-NACT), after a 3-month course of additional chemotherapy and/or different experimental therapies in cases of AC resistance (post-NACT), and from the metastatic lesions of two patients. The collection includes 12 longitudinal sets. Models were established from chemo-sensitive and -resistant tumors, but engraftment success was higher from those cancers that proved to be treatment resistant (RCB II/III). The PDX collection includes models encompassing a broad heterogeneity of chemotherapy responses, histologic features, and molecular TNBC subtypes. In addition, the majority of models develop spontaneous lung metastases. Whole exome sequencing demonstrated conservation of mutations between patient tumors and corresponding PDX models, with TP53 being the most commonly mutated gene. A similar comparison of patient and PDX transcriptomes revealed conservation of signaling pathway signatures, with those related to immune and stromal interactions being the most variable, as anticipated when adapting human tumors to growth in mice. The subclonal architecture of the tumors exhibited little overall change throughout NACT, suggesting that selection for resistant subclones is not likely to be a significant contributor to resistance. Tumor cells from the longitudinal PDX collections were subjected to high throughput drug susceptibility profiling to identify targetable vulnerabilities associated with resistance to NACT. We found that post-NACT tumors exhibited enhanced sensitivity to drugs targeting protein homeostasis pathways. Preclinical studies with pevonedistat validated the neddylation pathway as an effective target in chemoresistant TNBC. Citation Format: Amanda L. Rinkenbaugh, Yuan Qi, Reid T. Powell, Shirong Cai, Jiansu Shao, Faiza Baameur Hancock, Lei Guo, Xiaomei Zhang, Sabrina Jeter-Jones, Chunxiao Fu, Rebekah Gould, Jason B. White, Clifford Stephan, Gloria V. Echeverria, Peter J. Davies, Stacy Moulder, W. Fraser Symmans, Jeffrey T. Chang, Helen Piwnica-Worms. PDX models of TNBC established from pre- and post-therapy tumors identify vulnerabilities of resistant disease [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 6620.


PTEN in triple-negative breast carcinoma: protein expression and genomic alteration in pretreatment and posttreatment specimens

August 2023

·

104 Reads

·

2 Citations

Background Recent advances have been made in targeting the phosphoinositide 3-kinase pathway in breast cancer. Phosphatase and tensin homolog (PTEN) is a key component of that pathway. Objective To understand the changes in PTEN expression over the course of the disease in patients with triple-negative breast cancer (TNBC) and whether PTEN copy number variation (CNV) by next-generation sequencing (NGS) can serve as an alternative to immunohistochemistry (IHC) to identify PTEN loss. Methods We compared PTEN expression by IHC between pretreatment tumors and residual tumors in the breast and lymph nodes after neoadjuvant chemotherapy in 96 patients enrolled in a TNBC clinical trial. A correlative analysis between PTEN protein expression and PTEN CNV by NGS was also performed. Results With a stringent cutoff for PTEN IHC scoring, PTEN expression was discordant between pretreatment and posttreatment primary tumors in 5% of patients (n = 96) and between posttreatment primary tumors and lymph node metastases in 9% (n = 33). A less stringent cutoff yielded similar discordance rates. Intratumoral heterogeneity for PTEN loss was observed in 7% of the patients. Among pretreatment tumors, PTEN copy numbers by whole exome sequencing (n = 72) were significantly higher in the PTEN-positive tumors by IHC compared with the IHC PTEN-loss tumors (p < 0.0001). However, PTEN-positive and PTEN-loss tumors by IHC overlapped in copy numbers: 14 of 60 PTEN-positive samples showed decreased copy numbers in the range of those of the PTEN-loss tumors. Conclusion Testing various specimens by IHC may generate different PTEN results in a small proportion of patients with TNBC; therefore, the decision of testing one versus multiple specimens in a clinical trial should be defined in the patient inclusion criteria. Although a distinct cutoff by which CNV differentiated PTEN-positive tumors from those with PTEN loss was not identified, higher copy number of PTEN may confer positive PTEN, whereas lower copy number of PTEN would necessitate additional testing by IHC to assess PTEN loss. Trial registration NCT02276443.


Trial design and study cohort. Biomarkers including sTIL, PD-L1, AR and Ki-67 were assessed in pretreatment tumors. Patients underwent imaging to assess treatment response after AC. Those who had a chemo-sensitive disease after 4 cycles of AC were recommended to proceed with standard taxane-based chemotherapy. Those who were predicted to be chemo-insensitive were offered therapy in clinical trials using targeted therapy in combination with chemotherapy based on the biomarker results of their tumors. The experimental trials were as follows: a phase II trial of neoadjuvant nab-paclitaxel and atezolizumab (NCT02530489); phase II trial of neoadjuvant liposomal doxorubicin, bevacizumab and everolimus (DAT) in TNBC insensitive to standard neoadjuvant chemo (NCT02456857); phase II trial of panitumumab, carboplatin and paclitaxel (PaCT) in localized TNBC insensitive to NACT (NCT02593175); and phase IIB neoadjuvant enzalutamide plus paclitaxel for AR+ TNBC (NCT02689427). AC, doxorubicin and cyclophosphamide; AR, androgen receptor; ARTEMIS, A Robust TNBC Evaluation fraMework to Improve Survival; NACT, neoadjuvant chemotherapy; pCR: pathologic complete response; sTIL: stromal tumor-infiltrating lymphocytes; TNBC: triple-negative breast cancer.
pCR rates in sTIL groups among 408 patients. The total number of patients in each sTIL group is shown at the top of the bars. Recursive partitioning analysis identified ≥20% as the value for defining high sTIL in association with pCR. pCR, pathologic complete response; sTIL, stromal tumor-infiltrating lymphocytes.
Patient clinicopathologic characteristics.
Computed response score (testing set, n = 204).
Predictive Roles of Baseline Stromal Tumor-Infiltrating Lymphocytes and Ki-67 in Pathologic Complete Response in an Early-Stage Triple-Negative Breast Cancer Prospective Trial

June 2023

·

104 Reads

·

13 Citations

Simple Summary High stromal tumor-infiltrating lymphocytes (sTILs) are associated with improved pathologic complete response (pCR) in triple-negative breast cancer (TNBC). In this study of 408 patients enrolled in a prospective early-stage TNBC neoadjuvant chemotherapy trial, we aimed to identify clinicopathologic features that could be combined with sTILs to better predict pCR. Applying a training set and a testing set, we found that integrating high Ki-67 (cutoff > 35%) and high sTIL (cutoff ≥ 20%) in a model of computed response scores could predict a pCR rate of 65%. This model may refine the selection of early-stage TNBC patients for neoadjuvant clinical trials evaluating de-escalation strategies. Abstract High stromal tumor-infiltrating lymphocytes (sTILs) are associated with improved pathologic complete response (pCR) in triple-negative breast cancer (TNBC). We hypothesize that integrating high sTILs and additional clinicopathologic features associated with pCR could enhance our ability to predict the group of patients on whom treatment de-escalation strategies could be tested. In this prospective early-stage TNBC neoadjuvant chemotherapy study, pretreatment biopsies from 408 patients were evaluated for their clinical and demographic features, as well as biomarkers including sTILs, Ki-67, PD-L1 and androgen receptor. Multivariate logistic regression models were developed to generate a computed response score to predict pCR. The pCR rate for the entire cohort was 41%. Recursive partitioning analysis identified ≥20% as the optimal cutoff for sTILs to denote 35% (143/408) of patients as having high sTILs, with a pCR rate of 59%, and 65% (265/408) of patients as having low sTILs, with a pCR rate of 31%. High Ki-67 (cutoff > 35%) was identified as the only predictor of pCR in addition to sTILs in the training set. This finding was verified in the testing set, where the highest computed response score encompassing both high sTILa and high Ki-67 predicted a pCR rate of 65%. Integrating Ki67 and sTIL may refine the selection of early stage TNBC patients for neoadjuvant clinical trials evaluating de-escalation strategies.



Abstract 2147: Decoding the natural biology of triple-negative breast cancer and response to chemotherapy by single-cell transcriptomics

April 2023

·

48 Reads

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks the expression of estrogen receptor (ER), progesterone receptor (PR) and HER2 and therefore have limited hormonal treatment options. Neoadjuvant chemotherapy (NAC) is backbone of treatment for TNBC, and about 50% of patients respond well leading to pathological complete response (pCR). However, the remaining patients develop resistance to NAC and progress to metastatic disease and poor survival in 1-2 years after the initial treatment. Previous studies have performed bulk RNA expression profiling of TNBC patients and identified 5-6 subgroups of patients, however these studies could not resolve expression programs at single cell resolution to distinguish between the tumor cells and different components of the tumor microenvironment (TME). Here we performed scRNA-seq of pre-treatment fresh core biopsy tissue samples from TNBC patients in the ARTEMIS clinical trial and compared these data between pCR and non-pCR patients to identify programs associated with response to NAC. We also compared these data to scRNA-seq data from patients with disease-free breast tissue to understand the basic biology of TNBC and identify cell types that are reprogrammed in malignant disease. Using the single cell tumor cell data, we identified 4 archetypes of TNBC which represent patient-level intertumor expression programs: luminal secretory-like (LS), basal/luminal-like (BL), immunoregulatory (IM), and luminal androgen receptor (LAR). Notably, the archetype BL was associated with non-pCR, while IM was associated with pCR. We further identified 13 metatraits, which are unique intratumoral expression programs that are shared across patients. Across the cancer cells, we identified 13 metatraits such as cell cycling, stress, hypoxia, interferon response, HLA, partial epithelial-mesenchymal transition, and endoplasmic reticulum stress, many of which corresponded to NAC response. In the immune compartment, we found 15 myeloid cell states, 14 T/NK cell states, and 6 B cell states, several of which corresponded to pCR/non-pCR. Similarly, in the stromal compartment, there were 4 fibroblast cell states, 4 pericyte cell states, and 7 endothelial cell subtypes, of which several cell states were associated with NAC response. Overall, these data report the natural biology of TNBC patients and malignant cell states that are reprogrammed in malignant disease, as well as their correspondence to NAC response, providing new data to predict which TNBC patients are likely to respond to chemotherapy. Citation Format: Tapsi Kumar, Yiyun Lin, Yun Yan, Shanshan Bai, Jianzhuo Li, Tuan Tran, Min Hu, Elizabeth Ravenberg, Maia Rauch, Alyson Clayborn, Alastair Thompson, Lei Huo, Stacy Moulder, Clinton Yam, Nicholas Navin. Decoding the natural biology of triple-negative breast cancer and response to chemotherapy by single-cell transcriptomics [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2147.


Abstract 3447: NOS inhibition reverses epithelial-to-mesenchymal transition and synergizes with alpelisib in metaplastic breast cancer

April 2023

·

44 Reads

Metaplastic breast cancer (MpBC) is a rare and highly chemoresistant breast cancer subtype, with a median survival of 8 months after metastatic disease, and no standard therapeutic options. MpBCs are enriched for epithelial-to-mesenchymal transition (EMT)/cancer stem cell (CSC) markers, produce high levels of nitric oxide (NO), and have a hyperactive phosphoinositide 3-kinase (PI3K) signaling pathway. NO activates multiple oncogenic pathways, such as PI3K and transforming growth factor beta (TGFβ), a regulator of EMT. Therefore, we hypothesized that pan-NOS inhibitor NG-monomethyl-l-arginine (L-NMMA) could augment the efficacy of α-specific PI3K inhibitor alpelisib in MpBC in vitro and in vivo models. Immunostaining analysis revealed that MpBC PDX tumors had elevated co-expression of iNOS and pAkt (60% vs 23%, p=0.04) relative to triple-negative breast cancer (TNBC) PDX tumors. MpBC PDX tumors had higher RPL39 A14V (66% vs 4.7%, p< 0.00) and PIK3CA hotspot mutation rates (50% vs 19.1%, p=0.31) than TNBC PDX tumors. L-NMMA was synergistic with alpelisib in MpBC cell lines with PIK3CA/PIK3R1 mutations and antagonistic in PIK3CA-wild type and PTEN-deleted models. In vivo evaluation using MpBC PDX tumors found that L-NMMA augmented the efficacy of alpelisib in reducing tumor volume in PIK3CA-mutated MpBC PDX models. Transcriptomic analysis found gene sets associated with EMT reversal, such as the formation of cornified envelope (NES = 2.04 Nom p<0.00) and keratinization pathway (NES = 2.06, Nom p<0.00), were enriched pathways in MpBC PDX tumors that responded to combination therapy. Pharmacological/genomic inhibition of iNOS reversed EMT in MpBC cells, by decreased expression of Zeb1, TGFβ, Snail, Vimentin, and increased expression of E-cadherin and ZO-1 in immunoblotting analysis. MpBC cells with NOS2 knockout acquired an epithelial-like cellular morphology, and this reversal of EMT rendered MpBC cells more sensitive to alpelisib and taxane-chemotherapy. MpBC PDX tumors that responded to combination therapy also exhibited a reversal in EMT, with a decrease in aldehyde dehydrogenase (ALDH1), a CSC marker. Combination therapy also reduced tumor-initiating ability, enhanced chemosensitivity, and improved overall survival in MpBC PDX models. These studies paralleled a phase 1b/2 clinical trial with L-NMMA+taxane chemotherapy in a cohort of anthracycline-refractory MpBC patients (NCT02834403). The clinical benefit rate was 40% (6/15), overall response rate was 20% (3/15), and one patient achieved a pathologic complete response. Relative to baseline tumors, the responder end-of-treatment tumors had undergone reversal of EMT, with decreased expression of iNOS and ALDH1. We find that combining L-NMMA and alpelisib is a novel therapeutic strategy to treat MpBC, and combination therapy is being tested in a first multicenter phase 2 study for patients with MpBC. Citation Format: Tejaswini P. Reddy, Akshjot Puri, Liliana Guzman-Rojas, Christoforos Thomas, Wei Qian, Jianying Zhou, Hong Zhao, Xiaoxian Li, Bijan Mahboubi, Adrian Oo, Cho Young-Jae, Baek Kim, Jose Thaiparambil, Maria Florencia Chervo, Roberto Rosato, Camila Ayerbe, Noah Giese, Stacy Moulder, Helen Piwnica-Worms, Funda Meric-Bernstam, Jenny C. Chang. NOS inhibition reverses epithelial-to-mesenchymal transition and synergizes with alpelisib in metaplastic breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3447.


Citations (57)


... Given the role of SUMOylation in regulating cell proliferation and maintaining genome integrity, evidence is accumulating for its crucial role in cancer. Interestingly, SUMOylation machinery components are highly overexpressed in breast cancer and are associated with poor patient outcomes [9,10]. Different reports have demonstrated that blocking the SUMOylation machinery induces cell death, inhibits the invasion of tumor cells, reduces the speed of cell migration [11,12] and even sensitizes cells to the cytotoxic effects of chemotherapeutic drugs [13]. ...

Reference:

SUMOylation regulates the aggressiveness of breast cancer-associated fibroblasts
Targeting neddylation and sumoylation in chemoresistant triple negative breast cancer

npj Breast Cancer

... The lack of specificity, sensitivity and inter-laboratory standardization in the determination of PTEN has led to controversy over its role as a prognostic and predictive biomarker for patients with breast cancer [28]. Lower expression of PTEN in breast cancer tissue indicates poor prognosis in patients with TNBC [29]. The HMG-box transcription factor 1/TIMP metallopeptidase inhibitor 3/PTEN axis inhibits the tumorigenesis of breast cancer and promotes the sensitivity of breast cancer to radiotherapy and hormone therapy [30]. ...

PTEN in triple-negative breast carcinoma: protein expression and genomic alteration in pretreatment and posttreatment specimens

... Differences between the pCR and non-pCR groups in age, body mass index, and longest tumor diameter were assessed using a t-test; differences in clinical stage, tumor category, and nodal category were assessed using the Pearson χ 2 test; and differences in stromal tumor-infiltrating lymphocytes and Ki-67 level were assessed using Fisher's exact test [32]. Statistical analyses were carried out using R version 4.0.3 ...

Predictive Roles of Baseline Stromal Tumor-Infiltrating Lymphocytes and Ki-67 in Pathologic Complete Response in an Early-Stage Triple-Negative Breast Cancer Prospective Trial

... By finding patterns in large numbers of data, like metabolomics data from plasma samples, XAI methods can help find specific biomarkers for disease diagnosis. Plasma samples provide an ideal biological material for being non-invasive and reflecting the general profile of circulating metabolites [16,17]. ...

Application of Artificial Intelligence to Plasma Metabolomics Profiles to Predict Response to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer

Frontiers in Artificial Intelligence

... A greater size and more irregular morphology of the tumor are linked to increased risks of metastasis and recurrence [11,12]. Consequently, there is a close correlation between tumor morphological features and treatment response and prognosis [13,14]. Despite the variety of features used to describe tumor morphology, many techniques essentially assess the same tumor characteristics, such as tumor size indicated by diameter and volume. ...

Abstract P3-03-06: Prediction of response to neoadjuvant systemic therapy in triple negative breast cancer using baseline tumor MRI characteristics and imaging patterns of response
  • Citing Article
  • February 2022

... The ratio of the lymphoid cells to stroma in each breast cancer tumor was recorded as a percentage [14]. Several recent studies identified the optimal cut-off value of 20% sTILs that was best associated with pathologic complete response in patients with TNBC [10,21]. In this study, the patients were categorized into high-sTILs (≥ 20%) and low-sTILs (< 20%) level groups (Fig. 2). ...

Prognostic Impact of High Baseline Stromal Tumor-Infiltrating Lymphocytes in the Absence of Pathologic Complete Response in Early-Stage Triple-Negative Breast Cancer

... Due to absence of ER, PR and HER2 expression, TNBC lacks effective targeted therapies, leaving neoadjuvant chemotherapy (NACT) as an initial treatment for early-stage disease [6,7], with the primary objectives of achieving pathologic complete response (pCR ypT0N0, with no residual invasive carcinoma in the breast or lymph nodes) [8,9] (i.e., complete absence of residual invasive carcinoma) or downstaging tumors before surgical intervention, potentially improving surgical outcomes and allowing for breast-conserving surgery in cases that might otherwise require mastectomy [2,3] [10]. Around 40-50% of patients achieve a pCR [3,[11][12][13][14][15], a critical surrogate endpoint strongly correlated with prolonged disease-free and overall survival [16]. Conversely, patients with residual disease (non-pCR) face higher rates of relapse and worse overall survival [6,17], highlighting the urgent need for NACT prediction response to effectively guide the clinical decision-making by stratifying patients into distinct prognostic and therapeutic pathways, tailor therapies and avoid unnecessary treatments [18][19][20]. ...

Early-stage Triple-negative Breast Cancer: Time to Optimize Personalized Strategies

The Oncologist

... Studies have highlighted the utility of US in evaluating tumor vascularity changes during NAC, which can serve as early indicators of treatment efficacy [22]. Additionally, advancements in US technology, such as contrast-enhanced ultrasound (CEUS), have shown promise in enhancing the visualization of tumor perfusion and assessing therapeutic response [23]. ...

Early ultrasound evaluation identifies excellent responders to neoadjuvant systemic therapy among patients with triple‐negative breast cancer

... In a previous cohort of 32 patients with TNBC undergoing NACT, no difference in terms of α-diversity between patients who achieved a pCR and those with residual disease was found. 19 However, some bacterial species were enriched in case of pCR. Another study of patients with TNBC including 30 women treated with NACT identified specific taxa that were abundant in patients with pCR; among patients with residual disease, those with partial response were enriched in Bacteroides caccae. ...

Abstract PS4-05: Prospective evaluation of the gut microbiome and response to neoadjuvant therapy (NAT) in early-stage triple negative breast cancer (TNBC)
  • Citing Conference Paper
  • February 2021

... pCR is then ascertained by pathologists following appropriate surgery according to the size of the residual tumor. To investigate if radiomic phenotypes can predict pCR, 390 radiomic features (first-order histogram features and second order grey-level-co-occurrence matrix) were extracted from each of 74 patients with Stage I-III TNBC who underwent ultrafast DCE-MRI at baseline in the ARTEMIS trial [20]. Given the relatively small sample size and large number of radiomic features, 3-fold cross-validation and a penalized logistic regression model with the elastic net penalty were used to build prediction models. ...

Abstract PD6-06: Radiomic phenotypes from dynamic contrast-enhanced MRI (DCE-MRI) parametric maps for early prediction of response to neoadjuvant systemic therapy (NAST) in triple negative breast cancer (TNBC) patients
  • Citing Conference Paper
  • February 2021