Honors & Awards


  • Young Researcher Travel Grant, Training Unit, Institute Curie (2019)
  • Horizon 2020 Marie Skłodowska-Curie actions-COFUND PhD Fellowship, European Commision (2017-2020)
  • Second-Best Masters’ thesis award, Indian Institute of Science Education and Research, Kolkata (2017)
  • INSPIRE Scholarship, Department of Science and Technology, Govt. of India (2012-2017)

Boards, Advisory Committees, Professional Organizations


  • Co-Chair, Stanford University Postdoctoral Association (SURPAS) (2023 - Present)
  • Associate Member, American Association for Cancer Research (2023 - Present)
  • Associate Trainee Member, Stanford Cancer Institute (2023 - Present)

Professional Education


  • B.S - M.S Dual Degree, Indian Institute of Science Education and Research (IISER), Kolkata, India, Biological Sciences (2017)
  • Ph.D., University of Paris-Saclay & Institut Curie - Research Center, Paris, France, Cancer Biology, Medicine and Health (2021)

Stanford Advisors


Current Research and Scholarly Interests


- Exploring the crosstalk between DNA repair mechanisms and protein arginine methyltransferases in triple-negative breast cancer
- Understanding the role of DNA repair enzymes in the context of breast cancer

Lab Affiliations


All Publications


  • Therapeutic Advantage of Targeting PRMT5 in Combination with Chemotherapies or EGFR/HER2 Inhibitors in Triple-Negative Breast Cancers. Breast cancer (Dove Medical Press) Dakroub, R., Huard, S., Hajj-Younes, Y., Suresh, S., Badran, B., Fayyad-Kazan, H., Dubois, T. 2023; 15: 785-799

    Abstract

    Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subgroup characterized by a high risk of resistance to chemotherapies and high relapse potential. TNBC shows inter-and intra-tumoral heterogeneity; more than half expresses high EGFR levels and about 30% are classified as HER2-low breast cancers. High PRMT5 mRNA levels are associated with poor prognosis in TNBC and inhibiting PRMT5 impairs the viability of subsets of TNBC cell lines and delays tumor growth in TNBC mice models. TNBC patients may therefore benefit from a treatment targeting PRMT5. The aim of this study was to assess the therapeutic benefit of combining a PRMT5 inhibitor with different chemotherapies used in the clinics to treat TNBC patients, or with FDA-approved inhibitors targeting the HER family members.The drug combinations were performed using proliferation and colony formation assays on TNBC cell lines that were sensitive or resistant to EPZ015938, a PRMT5 inhibitor that has been evaluated in clinical trials. The chemotherapies analyzed were cisplatin, doxorubicin, camptothecin, and paclitaxel. The targeted therapies tested were erlotinib (EGFR inhibitor), neratinib (EGFR/HER2/HER4 inhibitor) and tucatinib (HER2 inhibitor).We found that PRMT5 inhibition synergized mostly with cisplatin, and to a lesser extent with doxorubicin or camptothecin, but not with paclitaxel, to impair TNBC cell proliferation. PRMT5 inhibition also synergized with erlotinib and neratinib in TNBC cell lines, especially in those overexpressing EGFR. Additionally, a synergistic interaction was observed with neratinib and tucatinib in a HER2-low TNBC cell line as well as in a HER2-positive breast cancer cell line. We noticed that synergy can be obtained in TNBC cell lines that were resistant to PRMT5 inhibition alone.Altogether, our data highlight the therapeutic potential of targeting PRMT5 using combinatorial strategies for the treatment of subsets of TNBC patients.

    View details for DOI 10.2147/BCTT.S430513

    View details for PubMedID 37954171

    View details for PubMedCentralID PMC10637385

  • Expression, Localization and Prognosis Association of MEP50 in Breast Cancer. Cancers Suresh, S., Vinet, M., Dakroub, R., Lesage, L., Ye, M., Fayyad-Kazan, H., Nicolas, A., Meseure, D., Dubois, T. 2022; 14 (19)

    Abstract

    Breast cancer is composed of distinct subgroups, triple-negative breast cancer (TNBC), human epidermal growth factor receptor-2 (HER2), luminal A, and luminal B, which are associated with different prognosis. MEP50 is the main partner of the arginine methyltransferase PRMT5 required for its enzymatic activity. Here, we examined MEP50 expression in the different breast cancer subgroups from the transcriptomic data obtained on human breast cancer samples and on normal breast tissues in two cohorts (Curie, n = 141; The Cancer Genome Atlas-TCGA, n = 788). We observed higher levels of MEP50 mRNA in TNBC (Curie, n = 41; TCGA, n = 106) compared to the other breast cancer subgroups and normal breast tissues. Using an online KM-plotter database, which allows survival analyses in a larger number of breast cancer patients, we found that high MEP50 mRNA levels were associated with a more favorable recurrence-free survival (RFS) in TNBC (n = 953, p = 1.2 × 10-4) and luminal B (n = 1353, p = 0.013) tumors, whereas high PRMT5 mRNA levels were associated with worse RFS in these two subgroups (TNBC: n = 442, p = 1.0 × 10-4; luminal B: n = 566, p = 6.8 × 10-3). We next determined the expression and the subcellular localization of MEP50 protein by immunohistochemistry (IHC) in our Curie cohort of breast cancer (n = 94) and normal tissues (n = 7) using a validated MEP50 antibody. MEP50 was more expressed in breast tumors compared to normal breast tissues (p = 0.02). MEP50 was more localized to the cytosol in breast cancer cells compared to normal breast tissue (p = 4 × 10-4), and was more found at the plasma membrane in normal tissues compared to breast tumors (p = 0.01). We also evaluated PRMT5 activity by IHC in our Curie cohort using a validated antibody (H4R3me2s) detecting histone H4 symmetrically dimethylated on Arg3. High levels of H4R3me2s were found in normal breast tissues, whereas the lowest levels of H4R3me2s were observed in TNBC and HER2 breast cancer subgroups. Altogether, our study reports the expression of the PRMT5 cofactor (MEP50) and substrate (H4R3me2s) in breast cancer and highlights the association of PRMT5 and MEP50 mRNA with prognosis in luminal B and TNBC breast cancer subgroups and certain TNBC subtypes.

    View details for DOI 10.3390/cancers14194766

    View details for PubMedID 36230689

  • PRMT1 Regulates EGFR and Wnt Signaling Pathways and Is a Promising Target for Combinatorial Treatment of Breast Cancer. Cancers Suresh, S., Huard, S., Brisson, A., Némati, F., Dakroub, R., Poulard, C., Ye, M., Martel, E., Reyes, C., Silvestre, D. C., Meseure, D., Nicolas, A., Gentien, D., Fayyad-Kazan, H., Le Romancer, M., Decaudin, D., Roman-Roman, S., Dubois, T. 2022; 14 (2)

    Abstract

    Identifying new therapeutic strategies for triple-negative breast cancer (TNBC) patients is a priority as these patients are highly prone to relapse after chemotherapy. Here, we found that protein arginine methyltransferase 1 (PRMT1) is highly expressed in all breast cancer subtypes. PRMT1 depletion decreases cell survival by inducing DNA damage and apoptosis in various breast cancer cell lines. Transcriptomic analysis and chromatin immunoprecipitation revealed that PRMT1 regulates the epidermal growth factor receptor (EGFR) and the Wnt signaling pathways, reported to be activated in TNBC. PRMT1 enzymatic activity is also required to stimulate the canonical Wnt pathway. Type I PRMT inhibitors decrease breast cancer cell proliferation and show anti-tumor activity in a TNBC xenograft model. These inhibitors display synergistic interactions with some chemotherapies used to treat TNBC patients as well as erlotinib, an EGFR inhibitor. Therefore, targeting PRMT1 in combination with these chemotherapies may improve existing treatments for TNBC patients.

    View details for DOI 10.3390/cancers14020306

    View details for PubMedID 35053470

    View details for PubMedCentralID PMC8774276

  • CARM1/PRMT4: Making Its Mark beyond Its Function as a Transcriptional Coactivator. Trends in cell biology Suresh, S., Huard, S., Dubois, T. 2021; 31 (5): 402-417

    Abstract

    Coactivator-associated arginine methyltransferase 1 (CARM1), identified 20 years ago as a coregulator of transcription, is an enzyme that catalyzes arginine methylation of proteins. Beyond its well-established involvement in the regulation of transcription, the physiological functions of CARM1 are still poorly understood. However, recent studies have revealed novel roles of CARM1 in autophagy, metabolism, paraspeckles, and early development. In addition, CARM1 is emerging as an attractive therapeutic target and a drug response biomarker for certain types of cancer. Here, we provide a comprehensive overview of the structure of CARM1 and its post-translational modifications, its various functions, apart from transcriptional coactivation, and its involvement in cancer.

    View details for DOI 10.1016/j.tcb.2020.12.010

    View details for PubMedID 33485722

  • Protein arginine methyltransferase 5: A novel therapeutic target for triple-negative breast cancers. Cancer medicine Vinet, M., Suresh, S., Maire, V., Monchecourt, C., Némati, F., Lesage, L., Pierre, F., Ye, M., Lescure, A., Brisson, A., Meseure, D., Nicolas, A., Rigaill, G., Marangoni, E., Del Nery, E., Roman-Roman, S., Dubois, T. 2019; 8 (5): 2414-2428

    Abstract

    TNBC is a highly heterogeneous and aggressive breast cancer subtype associated with high relapse rates, and for which no targeted therapy yet exists. Protein arginine methyltransferase 5 (PRMT5), an enzyme which catalyzes the methylation of arginines on histone and non-histone proteins, has recently emerged as a putative target for cancer therapy. Potent and specific PRMT5 inhibitors have been developed, but the therapeutic efficacy of PRMT5 targeting in TNBC has not yet been demonstrated. Here, we examine the expression of PRMT5 in a human breast cancer cohort obtained from the Institut Curie, and evaluate the therapeutic potential of pharmacological inhibition of PRMT5 in TNBC. We find that PRMT5 mRNA and protein are expressed at comparable levels in TNBC, luminal breast tumors, and healthy mammary tissues. However, immunohistochemistry analyses reveal that PRMT5 is differentially localized in TNBC compared to other breast cancer subtypes and to normal breast tissues. PRMT5 is heterogeneously expressed in TNBC and high PRMT5 expression correlates with poor prognosis within this breast cancer subtype. Using the small-molecule inhibitor EPZ015666, we show that PRMT5 inhibition impairs cell proliferation in a subset of TNBC cell lines. PRMT5 inhibition triggers apoptosis, regulates cell cycle progression and decreases mammosphere formation. Furthermore, EPZ015666 administration to a patient-derived xenograft model of TNBC significantly deters tumor progression. Finally, we reveal potentiation between EGFR and PRMT5 targeting, suggestive of a beneficial combination therapy. Our findings highlight a distinctive subcellular localization of PRMT5 in TNBC, and uphold PRMT5 targeting, alone or in combination, as a relevant treatment strategy for a subset of TNBC.

    View details for DOI 10.1002/cam4.2114

    View details for PubMedID 30957988

    View details for PubMedCentralID PMC6537044