Unveiling Molecular Insights in Cancer with Notable Spatial Transcriptomics Datasets

Shreyasi Chandra
April 24, 2024

Spatial transcriptomics technologies have revolutionized our understanding of complex diseases by enabling researchers to map the cellular composition of tissues in a spatially resolved manner. This dataset roundup delves into two significant studies that utilize spatial transcriptomics to explore breast cancer and oral squamous cell carcinoma (OSCC), offering groundbreaking insights into their cellular ecosystems.

Notable Spatial Transcriptomics Datasets in Cancer

Dataset 1

Dataset ID: 4739739_CID44971
Year of Publication: 2021
Disease: Breast Cancer
Experiment Type: Spatial Transcriptomics
Total Samples: 6
Total cells: 1.05k
Organism: Homo sapiens
Reference Link: Publication

Why This Dataset Matters: Unraveling the Complexities of Breast Cancer

This dataset is crucial for advancing our understanding of breast cancer because it provides detailed single-cell and spatially resolved transcriptomics analysis. This comprehensive examination reveals the cellular heterogeneity within breast tumors, offering insights into the various cell types and their interactions, which can influence disease progression and treatment response. The study's immunophenotyping using CITE-seq uncovers immune cell profiles, including novel macrophage populations linked to clinical outcomes. By examining stromal-immune niches, the dataset enhances our knowledge of antitumor immune regulation and the spatial organization of different cell types in the tumor microenvironment. Additionally, it identifies nine distinct clusters (ecotypes) with unique cellular compositions and clinical outcomes, contributing valuable information for personalized treatment strategies. Overall, this transcriptional atlas of breast cancer cellular architecture is a foundational resource for understanding the complex ecosystem of the disease and informing future research and therapeutic approaches.

Decoding Impacts:

  • High-Resolution Cellular Landscape: The analysis of 26 primary breast tumors using scRNA-seq provides a detailed high-resolution cellular landscape, offering insights into the diverse cellular compositions across different clinical subtypes (ER+, HER2+, TNBC).
  • Heterogeneous Tumor Subtypes: SCSubtype revealed substantial heterogeneity within breast tumors, with many tumors exhibiting a mix of molecular subtypes, including luminal, HER2E, and basal-like profiles.
  • Validation Through Bulk RNA-seq: The approach's accuracy was validated through bulk whole-transcriptome RNA-seq, confirming strong concordance between SCSubtype calls and bulk tumor profiles.
  • Recurrent Gene Modules: The analysis identified recurrent gene modules driving neoplastic cell heterogeneity, providing insights into the key biological pathways and functions influencing intratumor transcriptional heterogeneity.
Notable Spatial Transcriptomics Datasets in Cancer
Mapping breast cancer heterogeneity using spatially resolved transcriptomics. Images of TNBC CID44971 sample depict distinct tissue regions including normal ductal (purple), stromal and adipose (blue), lymphocyte aggregates (yellow), ductal carcinoma in situ (DCIS, orange), and invasive cancer (red) (Picture taken from publication)
Notable Spatial Transcriptomics Datasets in Cancer
Pearson correlation heatmap in TNBC CID44971 reveals correlations between gene modules (GMs) with two-sided correlation coefficients (Benjamini–Hochberg-adjusted P < 0.05) (Picture taken from publication)
Notable Spatial Transcriptomics Datasets in Cancer
Heatmap of spatial proximity between selected CAF T cell signaling molecules, highlighting interaction scores for top 10% of tissue spots enriched for iCAFs and CD4+/CD8+ T cells (Picture taken from publication)

Insights into Breast Cancer Data:

  • Spatial Mapping of Heterogeneity: Spatial transcriptomics enabled the mapping of cellular heterogeneity within breast cancers, allowing researchers to visualize the spatial organization of different cell types in tumors.
  • Distinguishing Tumor Regions: The study revealed distinct spatial patterns for various gene modules (GMs) associated with different biological functions such as EMT, proliferation, and hormone receptor activity across TNBC and ER+ cases.
  • Mutually Exclusive Cancer Phenotypes: Spatial analysis showed that certain gene modules were mutually exclusive, suggesting distinct cancer phenotypes occurred in different regions of the same breast cancer tumor.
  • CAF and Immune Cell Interactions: Spatial mapping highlighted the relationship between different subtypes of cancer-associated fibroblasts (CAFs) and immune cells. Myofibroblast-like CAFs were found in invasive cancer regions, while iCAFs were dispersed across invasive cancer, stroma, and TIL-aggregate regions.
  • Immunoregulatory Niches: The study identified potential mediators of CAF-lymphocyte interactions in specific tumor regions, including signaling pathways involving chemokines, the complement pathway, and transforming growth factor-β (TGF-β).
  • Implications for Patient Survival: By examining cellular compositions and frequencies across tumors, spatial transcriptomics offered insights into breast tumor ecotypes that could be associated with patient survival and response to treatment.

Dataset 2

Dataset ID: GSE220978_GSM6833484
Year of Publication: 2024
Disease: Oral Squamous Cell Carcinoma
Experiment Type: Spatial Transcriptomics
Total Samples: 4
Total cells: 4.2k
Organism: Homo sapiens
Reference Link: Publication

Why This Dataset Matters: Deciphering the Progression of Oral Sub mucous Fibrosis to OSCC

The dataset being presented in this study integrates Spatial Transcriptomics (ST) and Spatial Metabolomics (SM) techniques to provide a comprehensive and detailed landscape of oral squamous cell carcinoma (OSCC) tissues, specifically those derived from oral submucous fibrosis (OSF). By examining the transcriptomic and metabolomic profiles within OSCC tissues, the study reveals the complex interactions and progression patterns of the disease, from in situ carcinoma to partial epithelial-mesenchymal transition (pEMT), and ultimately, a cancer-associated fibroblast-like phenotype.

Through this integration, the dataset uncovers the intricate relationships between tumor cells, fibroblasts, and immune cells, as well as their impact on the tumor microenvironment.

One of the most significant findings of the dataset is the identification of major metabolic reprogramming in OSCC, particularly within the polyamine metabolism pathway. The abnormal expression of key enzymes involved in polyamine metabolism is shown to drive this metabolic reprogramming, which may subsequently reshape the tumor microenvironment and influence disease progression. This dataset is essential for advancing our understanding of OSCC and the complex molecular events underpinning its development, as well as for guiding the discovery of potential therapeutic targets and interventions.

Decoding Impacts:

  • Complex Progression of OSF-derived OSCC: The study examines the progression of oral squamous cell carcinoma (OSCC) originating from oral submucous fibrosis (OSF), often induced by prolonged betel nut chewing.
  • Spatial Analysis Reveals Cellular Dynamics: Using spatial transcriptomics (ST) and spatial metabolomics (SM) analysis, researchers uncover the cellular diversity and molecular events involved in the malignant transformation of OSCC.
  • Identification of Distinct Clusters: The study identifies 11 distinct clusters within the ST data, including tumor, adjacent epithelium, immune, and stroma subtypes, highlighting the diversity and evolution of cell types in OSCC.
  • Malignant Transformation Trajectory: The research maps the malignant trajectory from in situ carcinoma (ISC) through partial epithelial-mesenchymal transition (pEMT) to a cancer-associated fibroblast (CAF)-like phenotype.
  • Role of Transcription Factors: Transcription factors such as FOSL1 and TCF4 are identified as regulators of key downstream genes in the EMT and CAF pathways, influencing tumor progression and migration.
  • Impact on Tumor Microenvironment: The progression of epithelial cells to mesenchymal and CAF-like states impacts the tumor microenvironment, facilitating tumor invasion and migration.
  • Potential Therapeutic Targets: By focusing on the evolution and regulation of OSCC, the study opens avenues for potential therapeutic strategies targeting key molecular drivers in OSF-derived OSCC.
Notable Spatial Transcriptomics Datasets in Cancer
Fig: A) Spatial transcriptomics (ST) reveals distribution of immune cells in oral squamous cell carcinoma (OSCC) from oral submucous fibrosis (OSF). Immune cells, including B, T/NK, macrophages, dendritic, and other myeloid cells, show distinct spatial patterns. B) ST shows the locations of these immune cell types within tissue. (Picture taken from publication)
Notable Spatial Transcriptomics Datasets in Cancer
Fig: C) Dot plots present key ligand-receptor pairs expressed in immune cells. D) ST feature plots visualize expression and spatial distribution of ligand-receptor pairs among immune cells, epithelial cells, and cancer-associated fibroblasts (CAFs) (Picture taken from Publication)
Notable Spatial Transcriptomics Datasets in Cancer
Fig: E) TLS-like regions, suggesting enhanced immune responses, are identified in tissue sections. F) Immunofluorescence demonstrates the spatial organisation of T-cell (CD3) and B-cell (CD20) markers within TLS-like structures (Picture taken from Publication)

Insights into OSCC

  • Interplay Among Cells:
    • Immune cells interact with different types of fibroblasts, affecting EMT and fibrosis processes.
    • Immune cells may induce pEMT and acquire CAF-like phenotypes in OSCC through ligand-receptor pairs.
  • Prognostic Value:
    • The presence of TLSs and specific ligand-receptor pairs correlates with prognosis in OSCC patients.
  • Spatial Distribution:
    • Immune cells show significant aggregation in the stroma and tumor regions of the tissue sections.
    • Distribution patterns suggest interactions among immune cells, tumor cells, and fibroblasts
  • Significance:
    • The complex interactions between immune cells, epithelial cells, and fibroblasts create a unique immune microenvironment in OSF-derived OSCC.
    • Understanding these interactions could guide therapeutic strategies for OSCC.

The Path Forward

These datasets not only deepen our understanding of complex diseases like breast cancer and OSCC but also illustrate the power of spatial transcriptomics in uncovering novel insights that can drive therapeutic innovations. As we continue to unravel the spatial and molecular intricacies of diseases, we pave the way for more effective and personalised treatment strategies.

Connect with us to explore how Polly can expedite your research journey or reach us at info@elucidata.io to learn more.
Request Demo