Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Daisy Huynh is active.

Publication


Featured researches published by Daisy Huynh.


Biochimica et Biophysica Acta | 2014

Regulation of microRNAs in cancer metastasis.

Juliette M.C. Bouyssou; Salomon Manier; Daisy Huynh; Samar Issa; Aldo M. Roccaro; Irene M. Ghobrial

Metastasis is a phenomenon of crucial importance in defining prognosis in patients with cancer and is often responsible for cancer-related mortality. It is known that several steps are necessary for clonal cells to disseminate from their primary tumor site and colonize distant tissues, thus originating metastatic lesions. Therefore, investigating the molecular actors regulating this process may provide helpful insights in the development of efficient therapeutic responses. Recent evidences have indicated the role of microRNAs (miRNAs) in modulating the metastatic process in solid tumors. miRNAs are small regulatory non-coding RNAs that bind to specific target mRNAs, leading to translational repression. miRNAs are known to act as negative regulators of gene expression and are involved in the regulation of biological processes, including cell growth, differentiation and apoptosis, both in physiological conditions and during diseases, such as tumors. In the specific field of tumorigenesis, miRNAs play an important role in mediating oncogenesis and favoring tumor progression, as a result of their ability to modulate epithelial-to-mesenchymal transition (EMT) and other series of events facilitating the formation of metastasis. The role of miRNAs in cancer development has been widely studied and has helped elucidate events such as the change in expression of oncogenes, tumor-suppressors and cancer-related proteins. This review focuses on the mechanisms underlying the role of miRNAs as part of the metastatic process.


Cancer Research | 2015

Metabolic Signature Identifies Novel Targets for Drug Resistance in Multiple Myeloma

Patricia Maiso; Daisy Huynh; Michele Moschetta; Antonio Sacco; Yosra Aljawai; Yuji Mishima; John M. Asara; Aldo M. Roccaro; Alec C. Kimmelman; Irene M. Ghobrial

Drug resistance remains a major clinical challenge for cancer treatment. Multiple myeloma is an incurable plasma cell cancer selectively localized in the bone marrow. The main cause of resistance in myeloma is the minimal residual disease cells that are resistant to the original therapy, including bortezomib treatment and high-dose melphalan in stem cell transplant. In this study, we demonstrate that altered tumor cell metabolism is essential for the regulation of drug resistance in multiple myeloma cells. We show the unprecedented role of the metabolic phenotype in inducing drug resistance through LDHA and HIF1A in multiple myeloma, and that specific inhibition of LDHA and HIF1A can restore sensitivity to therapeutic agents such as bortezomib and can also inhibit tumor growth induced by altered metabolism. Knockdown of LDHA can restore sensitivity of bortezomib resistance cell lines while gain-of-function studies using LDHA or HIF1A induced resistance in bortezomib-sensitive cell lines. Taken together, these data suggest that HIF1A and LDHA are important targets for hypoxia-driven drug resistance. Novel drugs that regulate metabolic pathways in multiple myeloma, specifically targeting LDHA, can be beneficial to inhibit tumor growth and overcome drug resistance.


Cell Reports | 2015

CXCR4 regulates extra-medullary myeloma through epithelial-mesenchymal transition-like transcriptional activation

Aldo M. Roccaro; Yuji Mishima; Antonio Sacco; Michele Moschetta; Yu-Tzu Tai; Jiantao Shi; Yong Zhang; Michaela R. Reagan; Daisy Huynh; Yawara Kawano; Ilyas Sahin; Marco Chiarini; Salomon Manier; Michele Cea; Yosra Aljawai; Siobhan Glavey; Elizabeth A. Morgan; Chin Pan; Franziska Michor; Pina M. Cardarelli; Michelle Kuhne; Irene M. Ghobrial

Extra-medullary disease (EMD) in multiple myeloma (MM) is associated with poor prognosis and resistance to chemotherapy. However, molecular alterations that lead to EMD have not been well defined. We developed bone marrow (BM)- and EMD-prone MM syngeneic cell lines; identified that epithelial-to-mesenchymal transition (EMT) transcriptional patterns were significantly enriched in both clones compared to parental cells, together with higher levels of CXCR4 protein; and demonstrated that CXCR4 enhanced the acquisition of an EMT-like phenotype in MM cells with a phenotypic conversion for invasion, leading to higher bone metastasis and EMD dissemination in vivo. In contrast, CXCR4 silencing led to inhibited tumor growth and reduced survival. Ulocuplumab, a monoclonal anti-CXCR4 antibody, inhibited MM cell dissemination, supported by suppression of the CXCR4-driven EMT-like phenotype. These results suggest that targeting CXCR4 may act as a regulator of EMD through EMT-like transcriptional modulation, thus representing a potential therapeutic strategy to prevent MM disease progression.


Blood | 2014

Pyk2 promotes tumor progression in multiple myeloma.

Yu Zhang; Michele Moschetta; Daisy Huynh; Yu-Tzu Tai; Yong Zhang; Wenjing Zhang; Yuji Mishima; Jennifer E. Ring; Winnie F. Tam; Qunli Xu; Patricia Maiso; Michaela R. Reagan; Ilyas Sahin; Antonio Sacco; Salomon Manier; Yosra Aljawai; Siobhan Glavey; Nikhil C. Munshi; Kenneth C. Anderson; Jonathan A. Pachter; Aldo M. Roccaro; Irene M. Ghobrial

Proline-rich tyrosine kinase 2 (Pyk2) is a member of the focal adhesion kinase family that has been recently linked to tumor development. However, its role in modulating multiple myeloma (MM) biology and disease progression remains unexplored. We first demonstrated that patients with MM present with higher expression of Pyk2 compared with healthy individuals. By using loss-of-function approaches, we found that Pyk2 inhibition led to reduction of MM tumor growth in vivo as well as decreased cell proliferation, cell-cycle progression, and adhesion ability in vitro. In turn, overexpression of Pyk2 promoted the malignant phenotype, substantiated by enhanced tumor growth and reduced survival. Mechanistically, inhibition of Pyk2 reduced activation of Wnt/β-catenin signaling by destabilizing β-catenin, leading to downregulation of c-Myc and Cyclin D1. Furthermore, treatment of MM cells with the FAK/Pyk2 inhibitor VS-4718 effectively inhibited MM cell growth both in vitro and in vivo. Collectively, our findings describe the tumor-promoting role of Pyk2 in MM, thus providing molecular evidence for a novel tyrosine kinase inhibitor as a new therapeutic option in MM.


Blood | 2017

Prognostic role of circulating exosomal miRNAs in multiple myeloma

Salomon Manier; Chia-Jen Liu; Hervé Avet-Loiseau; Jihye Park; Jiantao Shi; Federico Campigotto; Karma Salem; Daisy Huynh; Siobhan Glavey; Bradley Rivotto; Antonio Sacco; Aldo M. Roccaro; Juliette M.C. Bouyssou; Stéphane Minvielle; Philippe Moreau; Thierry Facon; Xavier Leleu; Edie Weller; Lorenzo Trippa; Irene M. Ghobrial

Exosomes, secreted by several cell types, including cancer cells, can be isolated from the peripheral blood and have been shown to be powerful markers of disease progression in cancer. In this study, we examined the prognostic significance of circulating exosomal microRNAs (miRNAs) in multiple myeloma (MM). A cohort of 156 patients with newly diagnosed MM, uniformly treated and followed, was studied. Circulating exosomal miRNAs were isolated and used to perform a small RNA sequencing analysis on 10 samples and a quantitative reverse transcription polymerase chain reaction (qRT-PCR) array on 156 samples. We studied the relationship between miRNA levels and patient outcomes, including progression-free survival (PFS) and overall survival (OS). We identified miRNAs as the most predominant small RNAs present in exosomes isolated from the serum of patients with MM and healthy controls by small RNA sequencing of circulating exosomes. We then analyzed exosomes isolated from serum samples of 156 patients using a qRT-PCR array for 22 miRNAs. Two of these miRNAs, let-7b and miR-18a, were significantly associated with both PFS and OS in the univariate analysis and were still statistically significant after adjusting for the International Staging System and adverse cytogenetics in the multivariate analysis. Our findings support the use of circulating exosomal miRNAs to improve the identification of patients with newly diagnosed MM with poor outcomes. The results require further validation in other independent prospective MM cohorts.


Leukemia | 2015

Phase I/II trial of everolimus in combination with bortezomib and rituximab (RVR) in relapsed/refractory Waldenstrom macroglobulinemia

Irene M. Ghobrial; Robert Redd; Philippe Armand; Ranjit Banwait; Erica N Boswell; Stacey Chuma; Daisy Huynh; Antonio Sacco; Aldo M. Roccaro; Adriana Perilla-Glen; Kimberly Noonan; M MacNabb; Houry Leblebjian; D Warren; P Henrick; Jorge J. Castillo; Paul G. Richardson; Jeffrey Matous; Edie Weller; Steven P. Treon

We examined the combination of the mammalian target of rapamycin inhibitor everolimus with bortezomib and rituximab in patients with relapsed/refractory Waldenstrom macroglobulinemia (WM) in a phase I/II study. All patients received six cycles of the combination of everolimus/rituximab or everolimus/bortezomib/rituximab followed by maintenance with everolimus until progression. Forty-six patients were treated; 98% received prior rituximab and 57% received prior bortezomib. No dose-limiting toxicities were observed in the phase I. The most common treatment-related toxicities of all grades were fatigue (63%), anemia (54%), leucopenia (52%), neutropenia (48%) and diarrhea (43%). Thirty-six (78%) of the 46 patients received full dose therapy (FDT) of the three drugs. Of these 36, 2 (6%) had complete response (90% confidence interval (CI): 1–16). In all, 32/36 (89%) of patients experienced at least a minimal response (90% CI: 76–96%). The observed partial response or better response rate was 19/36 (53, 90 CI: 38–67%). For the 36 FDT patients, the median progression-free survival was 21 months (95% CI: 12–not estimable). In summary, this study demonstrates that the combination of everolimus, bortezomib and rituximab is well tolerated and achieved 89% response rate even in patients previously treated, making it a possible model of non-chemotherapeutic-based combination therapy in WM.


Leukemia | 2017

The LIN28B/let-7 axis is a novel therapeutic pathway in multiple myeloma

Salomon Manier; John T. Powers; Antonio Sacco; Siobhan Glavey; Daisy Huynh; Michaela R. Reagan; Karma Salem; Michele Moschetta; Jiantao Shi; Yuji Mishima; Catherine Roche-Lestienne; Xavier Leleu; Aldo M. Roccaro; George Q. Daley; Irene M. Ghobrial

MYC is a major oncogenic driver of multiple myeloma (MM) and yet almost no therapeutic agents exist that target MYC in MM. Here we report that the let-7 biogenesis inhibitor LIN28B correlates with MYC expression in MM and is associated with adverse outcome. We also demonstrate that the LIN28B/let-7 axis modulates the expression of MYC, itself a let-7 target. Further, perturbation of the axis regulates the proliferation of MM cells in vivo in a xenograft tumor model. RNA-sequencing and gene set enrichment analyses of CRISPR-engineered cells further suggest that the LIN28/let-7 axis regulates MYC and cell cycle pathways in MM. We provide proof of principle for therapeutic regulation of MYC through let-7 with an LNA-GapmeR (locked nucleic acid-GapmeR) containing a let-7b mimic in vivo, demonstrating that high levels of let-7 expression repress tumor growth by regulating MYC expression. These findings reveal a novel mechanism of therapeutic targeting of MYC through the LIN28B/let-7 axis in MM that may impact other MYC-dependent cancers as well.


Endocrinology | 2017

Metformin Affects Cortical Bone Mass and Marrow Adiposity in Diet-Induced Obesity in Male Mice

Sheila Bornstein; Michele Moschetta; Yawara Kawano; Antonio Sacco; Daisy Huynh; Daniel J. Brooks; Salomon Manier; Heather Fairfield; Carolyne Falank; Aldo M. Roccaro; Kenichi Nagano; Roland Baron; Mary Bouxein; Calvin P.H. Vary; Irene M. Ghobrial; Clifford J. Rosen; Michaela R. Reagan

&NA; Obesity during maturation can affect the growing skeleton directly and indirectly, although these effects and the mechanisms behind them are not fully understood. Our objective was to determine how a high‐fat diet with or without metformin treatment affects skeletal development. We also sought to characterize changes that occur in white adipose tissue, circulating metabolites, lipids, and gut microbiota. A diet‐induced obesity C57BL/6J mouse model was used to test the effects of obesity and metformin on bone using bone histomorphometry and microcomputed tomography. Bone marrow adipose tissue was quantified with osmium tetroxide microcomputed tomography and histology. Dual‐energy x‐ray absorptiometry was used to analyze body composition. Hematoxylin and eosin staining was used to assess changes in white adipose depots, mass spectrometry was used for circulating lipids and protein metabolite analysis, and ribosomal RNA sequencing was used for gut microbiome analysis. Mice fed a high fat‐diet since wean displayed increased medullary areas and decreased osteoblast numbers in the long bones; this phenotype was partially normalized by metformin. Marrow and inguinal adipose expansion was also noted in obese mice, and this was partially normalized by metformin. A drug‐by‐diet interaction was noted for circulating lipid molecules, protein metabolites, and gut microbiome taxonomical units. Obesity was not detrimental to trabecular bone in growing mice, but bone marrow medullary expansion was observed, likely resulting from inhibition of osteoblastogenesis, and this was partially reversed by metformin treatment.


Nature Communications | 2018

Whole-exome sequencing of cell-free DNA and circulating tumor cells in multiple myeloma

Salomon Manier; Jang-Ung Park; Marzia Capelletti; Mark Bustoros; Sam Freeman; Gavin Ha; Justin Rhoades; Chia-Jen Liu; Daisy Huynh; Sarah C. Reed; Gregory Gydush; Karma Salem; Denisse Rotem; C. Freymond; Amir Yosef; Adriana Perilla-Glen; Laurent Garderet; E. Van Allen; Shaji Kumar; J. C. Love; Gad Getz; Viktor A. Adalsteinsson; Irene M. Ghobrial

Liquid biopsies including circulating tumor cells (CTCs) and cell-free DNA (cfDNA) have enabled minimally invasive characterization of many cancers, but are rarely analyzed together. Understanding the detectability and genomic concordance of CTCs and cfDNA may inform their use in guiding cancer precision medicine. Here, we report the detectability of cfDNA and CTCs in blood samples from 107 and 56 patients with multiple myeloma (MM), respectively. Using ultra-low pass whole-genome sequencing, we find both tumor fractions correlate with disease progression. Applying whole-exome sequencing (WES) to cfDNA, CTCs, and matched tumor biopsies, we find concordance in clonal somatic mutations (~99%) and copy number alterations (~81%) between liquid and tumor biopsies. Importantly, analyzing CTCs and cfDNA together enables cross-validation of mutations, uncovers mutations exclusive to either CTCs or cfDNA, and allows blood-based tumor profiling in a greater fraction of patients. Our study demonstrates the utility of analyzing both CTCs and cfDNA in MM.Circulating tumor cells (CTCs) and cell-free DNA (cfDNA) enables characterization of a patient’s cancer. Here, the authors analyse CTCs, cfDNA, and tumor biopsies from multiple myeloma patients to show these approaches are complementary for mutation detection, together enabling a greater fraction of patient tumors to be profiled.


Journal of Clinical Investigation | 2018

Blocking IFNRA1 inhibits multiple myeloma-driven Treg expansion and immunosuppression

Yawara Kawano; Oksana Zavidij; Jihye Park; Michele Moschetta; Katsutoshi Kokubun; Tarek H. Mouhieddine; Salomon Manier; Yuji Mishima; Naoka Murakami; Mark Bustoros; Romanos Sklavenitis Pistofidis; Mairead Reidy; Yu J. Shen; Mahshid Rahmat; Pavlo Lukyanchykov; Esilida Sula Karreci; Shokichi Tsukamoto; Jiantao Shi; Satoshi Takagi; Daisy Huynh; Antonio Sacco; Yu-Tzu Tai; Marta Chesi; P. Leif Bergsagel; Aldo M. Roccaro; Jamil Azzi; Irene M. Ghobrial

Despite significant advances in the treatment of multiple myeloma (MM), most patients succumb to disease progression. One of the major immunosuppressive mechanisms that is believed to play a role in myeloma progression is the expansion of regulatory T cells (Tregs). In this study, we demonstrate that myeloma cells drive Treg expansion and activation by secreting type 1 interferon (IFN). Blocking IFN &agr; and &bgr; receptor 1 (IFNAR1) on Tregs significantly decreases both myeloma-associated Treg immunosuppressive function and myeloma progression. Using syngeneic transplantable murine myeloma models and bone marrow (BM) aspirates of MM patients, we found that Tregs were expanded and activated in the BM microenvironment at early stages of myeloma development. Selective depletion of Tregs led to a complete remission and prolonged survival in mice injected with myeloma cells. Further analysis of the interaction between myeloma cells and Tregs using gene sequencing and enrichment analysis uncovered a feedback loop, wherein myeloma-cell-secreted type 1 IFN induced proliferation and expansion of Tregs. By using IFNAR1-blocking antibody treatment and IFNAR1-knockout Tregs, we demonstrated a significant decrease in myeloma-associated Treg proliferation, which was associated with longer survival of myeloma-injected mice. Our results thus suggest that blocking type 1 IFN signaling represents a potential strategy to target immunosuppressive Treg function in MM.

Collaboration


Dive into the Daisy Huynh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge