2020
DOI: 10.1038/s41588-020-0630-5
|View full text |Cite
|
Sign up to set email alerts
|

Unraveling tumor–immune heterogeneity in advanced ovarian cancer uncovers immunogenic effect of chemotherapy

Abstract: In metastatic cancer, the role of heterogeneity at the tumor-immune microenvironment, its molecular underpinnings and clinical relevance remain largely unexplored. To understand tumor-immune dynamics at baseline and upon chemotherapy treatment, we performed unbiased pathway and cell type-specific immunogenomics analysis of treatment-naive (38 5 samples from 8 patients) and paired chemotherapy treated (80 paired samples from 40 patients) high-grade serous ovarian cancer (HGSOC) samples. Whole transcriptome anal… Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

1
90
0

Year Published

2020
2020
2023
2023

Publication Types

Select...
7
1

Relationship

1
7

Authors

Journals

citations
Cited by 141 publications
(91 citation statements)
references
References 126 publications
1
90
0
Order By: Relevance
“…However, BPPNM tumors were unresponsive to single-agent PD-L1 and only produced long-term survival when ICB was combined with Olaparib, reflecting reported clinical data (46,49,63). These data suggest that PD-L1 expression and inflammatory status alone may not be sufficient on their own predict the success of ICB in these tumors and that immune-stimulating alterations to the tumor microenvironment incurred by adjuvant treatments such as chemotherapy may be necessary for a significant response to checkpoint inhibitors (43,64). Nonetheless, more generally, targeted therapy based on inhibiting the DNA damage response pathway in combination with ICB therapy in ovarian cancer offers the potential to tailor treatment for patients with BRCA1 mutant HGSC.…”
Section: Discussionmentioning
confidence: 63%
See 1 more Smart Citation
“…However, BPPNM tumors were unresponsive to single-agent PD-L1 and only produced long-term survival when ICB was combined with Olaparib, reflecting reported clinical data (46,49,63). These data suggest that PD-L1 expression and inflammatory status alone may not be sufficient on their own predict the success of ICB in these tumors and that immune-stimulating alterations to the tumor microenvironment incurred by adjuvant treatments such as chemotherapy may be necessary for a significant response to checkpoint inhibitors (43,64). Nonetheless, more generally, targeted therapy based on inhibiting the DNA damage response pathway in combination with ICB therapy in ovarian cancer offers the potential to tailor treatment for patients with BRCA1 mutant HGSC.…”
Section: Discussionmentioning
confidence: 63%
“…Altogether, the scRNA seq, flow cytometry, and ascitic cytokine data combined to describe distinct immunosuppressive microenvironments across the spectrum of the tumor-cell genotypes compared here, this being reminiscent of the heterogeneity seen among tumors borne by HGSC patients (42,43).…”
Section: Hgsc Tumors With Different Genotypes Evoke Distinct Immune Mmentioning
confidence: 91%
“…The tumouricidal treatment causes cell death, incurring multiple changes in the TME. After neoadjuvant chemotherapy (NACT), researchers adopted immunogenic analysis in HGSOC tumors and found increased NK cell infiltration and oligoclonal expansion of T cells, suggesting chemotherapy can potentiate immunogenicity of the primary tumor (Jiménez-Sánchez et al, 2020). Another gene expression analysis revealed ovarian cancer patients treated with paclitaxel had an enriched gene signature linked to M1 TAM activation (Wanderley et al, 2018).…”
Section: Treatment Impact and Response Prediction In Ovarian Cancer Tmementioning
confidence: 99%
“…We studied the differences in the molecular signaling pathways between the low-risk and high-risk patient groups using the 50 hallmark gene sets [26], extracted using single sample gene-set enrichment (ssgsea) analysis of the RNA samples [25]. The signaling pathways were categorized into immune, oncogenic, stromal, cellular, and other [24]. Patients were dichotimized using the median value (cluDiss = 68.6) of cluDiss (low-risk<median and vice versa).…”
Section: Correlation Of Cludiss To Biological Processesmentioning
confidence: 99%
“…The aims of this study were to (i) validate cluDiss as a predictor of outcomes using an internal and external multi-institutional cohort, and (ii) evaluate whether an integrated iRCG measure of HGSOC outcomes was more accurate than average heterogeneity radiomic (aRCG) and conventional imaging (CCG) measures. Finally, we attempted to establish the biological basis of the prognostic radiomics measures by studying their correlation with underlying biological processes characterized by a well-defined molecular HALLMARK gene set pathways, stromal and immune scores of the tumor microenvironment (TME), and established 18 cell types of the TME extracted using the consensus TME method [23,24] by using patient-level single sample gene set analysis (ssgsea) [25] from RNA-sequencing data.…”
Section: Introductionmentioning
confidence: 99%