Network


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

Hotspot


Dive into the research topics where Thomas Januario is active.

Publication


Featured researches published by Thomas Januario.


Nature | 2008

A paracrine requirement for hedgehog signalling in cancer

Robert L. Yauch; Stephen E. Gould; Suzie J. Scales; Tracy Tang; Hua Tian; Christina P. Ahn; Derek Marshall; Ling Fu; Thomas Januario; Dara Y. Kallop; Michelle Nannini-Pepe; Karen Kotkow; James C. Marsters; Lee L. Rubin; Frederic J. de Sauvage

Ligand-dependent activation of the hedgehog (Hh) signalling pathway has been associated with tumorigenesis in a number of human tissues. Here we show that, although previous reports have described a cell-autonomous role for Hh signalling in these tumours, Hh ligands fail to activate signalling in tumour epithelial cells. In contrast, our data support ligand-dependent activation of the Hh pathway in the stromal microenvironment. Specific inhibition of Hh signalling using small molecule inhibitors, a neutralizing anti-Hh antibody or genetic deletion of smoothened (Smo) in the mouse stroma results in growth inhibition in xenograft tumour models. Taken together, these studies demonstrate a paracrine requirement for Hh ligand signalling in the tumorigenesis of Hh-expressing cancers and have important implications for the development of Hh pathway antagonists in cancer.


Science | 2009

Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma.

Robert L. Yauch; Gerrit J. P. Dijkgraaf; Bruno Alicke; Thomas Januario; Christina P. Ahn; Thomas Holcomb; Kanan Pujara; Jeremy Stinson; Christopher A. Callahan; Tracy Tang; J. Fernando Bazan; Zhengyan Kan; Somasekar Seshagiri; Christine L. Hann; Stephen E. Gould; Jennifer A. Low; Charles M. Rudin; Frederic J. de Sauvage

A Smooth(ened) Path to Drug Resistance The Hedgehog (Hh) signaling pathway has emerged as a key contributor to the growth of medulloblastoma, an aggressive brain tumor. GDC-0449, a drug that ramps down this signaling pathway by binding to the Hh pathway component Smoothened, was recently shown to induce rapid and dramatic tumor regression in a patient with metastatic medulloblastoma, but the tumor eventually developed resistance to the drug. Yauch et al. (p. 572, published online 3 September) show that resistance arose because the tumor acquired a mutation in Smoothened that disrupts binding of the drug. Identification of this resistance mechanism may facilitate the design of next-generation drugs for this type of cancer. A mutation that prevents binding of a promising drug lead to its target protein confers resistance in a human brain tumor. The Hedgehog (Hh) signaling pathway is inappropriately activated in certain human cancers, including medulloblastoma, an aggressive brain tumor. GDC-0449, a drug that inhibits Hh signaling by targeting the serpentine receptor Smoothened (SMO), has produced promising anti-tumor responses in early clinical studies of cancers driven by mutations in this pathway. To evaluate the mechanism of resistance in a medulloblastoma patient who had relapsed after an initial response to GDC-0449, we determined the mutational status of Hh signaling genes in the tumor after disease progression. We identified an amino acid substitution at a conserved aspartic acid residue of SMO that had no effect on Hh signaling but disrupted the ability of GDC-0449 to bind SMO and suppress this pathway. A mutation altering the same amino acid also arose in a GDC-0449–resistant mouse model of medulloblastoma. These findings show that acquired mutations in a serpentine receptor with features of a G protein–coupled receptor can serve as a mechanism of drug resistance in human cancer.


Clinical Cancer Research | 2005

Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients.

Robert L. Yauch; Thomas Januario; David A. Eberhard; Guy Cavet; Wenjing Zhu; Ling Fu; Thinh Q. Pham; Robert Soriano; Jeremy Stinson; Somasekar Seshagiri; Zora Modrusan; Chin Yu Lin; Vincent O'Neill; Lukas Amler

Significant improvements in the outcome of non–small cell lung carcinoma (NSCLC) have been reported in patients treated with the epidermal growth factor receptor (EGFR) inhibitor, erlotinib. To discover biomarkers for the enrichment of patients who might benefit from treatment, a pharmacogenomic approach was used to identify gene signatures that may predict erlotinib activity using in vitro model systems. Erlotinib sensitivity in a panel of 42 NSCLC cell lines was determined by EGFR-mediated proliferative potential, EGFR mutations, and/or EGFR gene amplification, thus supporting an underlying biological mechanism of receptor activation. A strong multigene signature indicative of an epithelial to mesenchymal transition (EMT) was identified as a determinant of insensitivity to erlotinib through both supervised and unsupervised gene expression approaches. This observation was further supported by expression analysis of classic EMT marker proteins, including E-cadherin and vimentin. To investigate the clinical relevance of these findings, we examined expression of the epithelial marker E-cadherin by immunohistochemistry on primary tumor samples from subjects enrolled in a randomized NSCLC clinical trial in which erlotinib in combination with chemotherapy previously failed to show clinical activity. The majority (75%) of the 87 subjects tested showed strong E-cadherin staining and exhibited a significantly longer time to progression (hazard ratio, 0.37; log rank P = 0.0028) and a nonsignificant trend toward longer survival with erlotinib plus chemotherapy treatment versus chemotherapy alone. These data support a potential role for EMT as a determinant of EGFR activity in NSCLC tumor cells and E-cadherin expression as a novel biomarker predicting clinical activity of the EGFR inhibitor erlotinib in NSCLC patients.


Nature Medicine | 2007

Death-receptor O-glycosylation controls tumor-cell sensitivity to the proapoptotic ligand Apo2L/TRAIL

Klaus W Wagner; Elizabeth Punnoose; Thomas Januario; David A. Lawrence; Robert M. Pitti; Kate Lancaster; Dori Lee; Melissa von Goetz; Sharon Yee; Klara Totpal; Ling Huw; Viswanatham Katta; Guy Cavet; Sarah G. Hymowitz; Lukas Amler; Avi Ashkenazi

Apo2L/TRAIL stimulates cancer cell death through the proapoptotic receptors DR4 and DR5, but the determinants of tumor susceptibility to this ligand are not fully defined. mRNA expression of the peptidyl O-glycosyltransferase GALNT14 correlated with Apo2L/TRAIL sensitivity in pancreatic carcinoma, non–small-cell lung carcinoma and melanoma cell lines, and up to 30% of samples from various human malignancies showed GALNT14 overexpression. RNA interference of GALNT14 reduced cellular Apo2L/TRAIL sensitivity, whereas overexpression increased responsiveness. Biochemical analysis of DR5 identified several ectodomain O-(N-acetyl galactosamine–galactose–sialic acid) structures. Sequence comparison predicted conserved extracellular DR4 and DR5 O-glycosylation sites; progressive mutation of the DR5 sites attenuated apoptotic signaling. O-glycosylation promoted ligand-stimulated clustering of DR4 and DR5, which mediated recruitment and activation of the apoptosis-initiating protease caspase-8. These results uncover a new link between death-receptor O-glycosylation and apoptotic signaling, providing potential predictive biomarkers for Apo2L/TRAIL-based cancer therapy.


Cancer Research | 2011

Small molecule inhibition of GDC-0449 refractory Smoothened mutants and downstream mechanisms of drug resistance

Gerrit J. P. Dijkgraaf; Bruno Alicke; Lasse Weinmann; Thomas Januario; Kristina West; Zora Modrusan; Dan Burdick; Richard Goldsmith; Kirk Robarge; Dan Sutherlin; Suzie J. Scales; Stephen E. Gould; Robert L. Yauch; Frederic J. de Sauvage

Inappropriate Hedgehog (Hh) signaling has been directly linked to medulloblastoma (MB), a common malignant brain tumor in children. GDC-0449 is an Hh pathway inhibitor (HPI) currently under clinical investigation as an anticancer agent. Treatment of a MB patient with GDC-0449 initially regressed tumors, but this individual ultimately relapsed with a D473H resistance mutation in Smoothened (SMO), the molecular target of GDC-0449. To explore the role of the mutated aspartic acid residue in SMO function, we substituted D473 with every amino acid and found that all functional mutants were resistant to GDC-0449, with positively charged residues conferring potential oncogenic properties. Alanine scan mutagenesis of SMO further identified E518 as a novel prospective mutation site for GDC-0449 resistance. To overcome this form of acquired resistance, we screened a panel of chemically diverse HPIs and identified several antagonists with potent in vitro activity against these GDC-0449-resistant SMO mutants. The bis-amide compound 5 was of particular interest, as it was able to inhibit tumor growth mediated by drug resistant SMO in a murine allograft model of MB. However, focal amplifications of the Hh pathway transcription factor Gli2 and the Hh target gene cyclin D1 (Ccnd1) were observed in two additional resistant models, indicating that resistance may also occur downstream of SMO. Importantly, these HPI resistant MB allografts retained their sensitivity to PI3K inhibition, presenting additional opportunities for the treatment of such tumors.


Science Signaling | 2011

TRPS1 Targeting by miR-221/222 Promotes the Epithelial-to-Mesenchymal Transition in Breast Cancer

Susanna Stinson; Mark R. Lackner; Alex T. Adai; Nancy Yu; Hyo-Jin Kim; Carol O’Brien; Jill M. Spoerke; Suchit Jhunjhunwala; Zachary Boyd; Thomas Januario; Robert J. Newman; Peng Yue; Richard Bourgon; Zora Modrusan; Howard M. Stern; Søren Warming; Frederic J. de Sauvage; Lukas Amler; Ru-Fang Yeh; David Dornan

The microRNAs miR-221 and miR-222 promote a phenotype associated with metastasis and are found in a clinically aggressive form of breast cancer. Parsing Breast Cancer Subtype with MicroRNAs MicroRNAs (miRNAs), short noncoding RNAs that bind to and silence target mRNAs, have emerged as playing crucial regulatory roles not only in normal cellular processes but also in pathological conditions, such as cancer. Stinson et al. analyzed miRNA expression in different types of human breast cancer and found that miR-221 and miR-222 (miR-221/222) abundance was increased in the clinically aggressive basal-like subtype compared to the less aggressive luminal subtype. They determined that signaling through the epidermal growth factor receptor (EGFR)–RAS–extracellular signal–regulated kinase (ERK) pathway increased miR-221/222 transcription, and they defined a transcriptional regulatory pathway through which miR-221/222 promoted a phenotype associated with cancer cell invasion and metastasis. Their data suggest that combining inhibition of the EGFR-RAS-ERK pathway with standard chemotherapy could, by limiting miR-221/222 production, provide a strategy to combat metastasis in the basal-like subtype of breast cancer. The basal-like subtype of breast cancer has an aggressive clinical behavior compared to that of the luminal subtype. We identified the microRNAs (miRNAs) miR-221 and miR-222 (miR-221/222) as basal-like subtype–specific miRNAs and showed that expression of miR-221/222 decreased expression of epithelial-specific genes and increased expression of mesenchymal-specific genes, and increased cell migration and invasion in a manner characteristic of the epithelial-to-mesenchymal transition (EMT). The transcription factor FOSL1 (also known as Fra-1), which is found in basal-like breast cancers but not in the luminal subtype, stimulated the transcription of miR-221/222, and the abundance of these miRNAs decreased with inhibition of the epidermal growth factor receptor (EGFR) or MEK (mitogen-activated or extracellular signal–regulated protein kinase kinase), placing miR-221/222 downstream of the RAS pathway. Furthermore, miR-221/222–mediated reduction in E-cadherin abundance depended on their targeting the 3′ untranslated region of the GATA family transcriptional repressor TRPS1 (tricho-rhino-phalangeal syndrome type 1), which inhibited EMT by decreasing ZEB2 (zinc finger E-box–binding homeobox2) expression. We conclude that by promoting EMT, miR-221/222 may contribute to the more aggressive clinical behavior of basal-like breast cancers.


Cancer Cell | 2015

Genomic Analysis of Smoothened Inhibitor Resistance in Basal Cell Carcinoma

Hayley Sharpe; Gregoire Pau; Gerrit J. P. Dijkgraaf; Nicole Basset-Seguin; Zora Modrusan; Thomas Januario; Vickie Tsui; Alison B. Durham; Andrzej A. Dlugosz; Peter M. Haverty; Richard Bourgon; Jean Y. Tang; Kavita Y. Sarin; Luc Dirix; David C. Fisher; Charles M. Rudin; Howard Sofen; Michael R. Migden; Robert L. Yauch; Frederic J. de Sauvage

Smoothened (SMO) inhibitors are under clinical investigation for the treatment of several cancers. Vismodegib is approved for the treatment of locally advanced and metastatic basal cell carcinoma (BCC). Most BCC patients experience significant clinical benefit on vismodegib, but some develop resistance. Genomic analysis of tumor biopsies revealed that vismodegib resistance is associated with Hedgehog (Hh) pathway reactivation, predominantly through mutation of the drug target SMO and to a lesser extent through concurrent copy number changes in SUFU and GLI2. SMO mutations either directly impaired drug binding or activated SMO to varying levels. Furthermore, we found evidence for intra-tumor heterogeneity, suggesting that a combination of therapies targeting components at multiple levels of the Hh pathway is required to overcome resistance.


Science Signaling | 2011

miR-221/222 Targeting of Trichorhinophalangeal 1 (TRPS1) Promotes Epithelial-to-Mesenchymal Transition in Breast Cancer

Susanna Stinson; Mark R. Lackner; Alex T. Adai; Nancy Yu; Hyo-Jin Kim; Carol O’Brien; Jill M. Spoerke; Suchit Jhunjhunwala; Zachary Boyd; Thomas Januario; Robert J. Newman; Peng Yue; Richard Bourgon; Zora Modrusan; Howard M. Stern; Søren Warming; Frederic J. de Sauvage; Lukas Amler; Ru-Fang Yeh; David Dornan

MicroRNAs miR-221 and miR-222 are associated with a clinically aggressive form of breast cancer and promote epithelial-to-mesenchymal transition. Compared with the luminal subtype, the basal-like subtype of breast cancer has an aggressive clinical behavior, but the reasons for this difference between the two subtypes are poorly understood. We identified microRNAs (miRNAs) miR-221 and miR-222 (miR-221/222) as basal-like subtype-specific miRNAs that decrease expression of epithelial-specific genes and increase expression of mesenchymal-specific genes. In addition, expression of these miRNAs increased cell migration and invasion, which collectively are characteristics of the epithelial-to-mesenchymal transition (EMT). The basal-like transcription factor FOSL1 (also known as Fra-1) directly stimulated the transcription of miR-221/222, and the abundance of these miRNAs decreased with inhibition of MEK (mitogen-activated or extracellular signal–regulated protein kinase kinase), placing miR-221/222 downstream of the RAS pathway. The miR-221/222–mediated reduction in E-cadherin abundance depended on their targeting of the 3′ untranslated region (3′UTR) of TRPS1 (trichorhinophalangeal syndrome type 1), which is a member of the GATA family of transcriptional repressors. TRPS1 inhibited EMT by directly repressing expression of ZEB2 (Zinc finger E-box–binding homeobox 2). Therefore, miR-221/222 may contribute to the aggressive clinical behavior of basal-like breast cancers.


Science Translational Medicine | 2011

CD40 Pathway Activation Status Predicts Response to CD40 Therapy in Diffuse Large B Cell Lymphoma

Bart Burington; Peng Yue; Xiaoyan Shi; Ranjana H. Advani; Jeffrey Lau; Jenille Tan; Susanna Stinson; Jeremy Stinson; Thomas Januario; Sven de Vos; Stephen M. Ansell; Andres Forero-Torres; Grazyna Fedorowicz; Teddy T C Yang; Kristi Elkins; Changchun Du; Sankar Mohan; Nancy Yu; Zora Modrusan; Somasekar Seshagiri; Shang Fan Yu; Ajay Pandita; Hartmut Koeppen; Dorothy French; Andrew G. Polson; Rienk Offringa; Nancy Whiting; Allen Ebens; David Dornan

A 15-gene expression signature predicts whether a patient with diffuse large B cell lymphoma will respond to dacetuzumab, a therapeutic antibody. Matching Treatment to Tumor If physicians could predict the future, it would take the guess work out of designing the right treatment regimen for every patient’s cancer. The results presented by Burington et al. move us closer to clearing the crystal ball for diffuse large B cell lymphomas, a common type of non-Hodgkin’s lymphoma in which a cell surface receptor, CD40, presents a seemingly attractive target for therapy. Although stimulation of CD40 by ligand binding can cause apoptosis of B cells—a trait that one would predict to be desirable for a B cell lymphoma drug—it can also induce undesirable proliferation of some lymphoma cells. This murky paradox makes it unclear when to prescribe dacetuzumab, a CD40-targeted therapeutic monoclonal antibody. The authors have now identified a 15-gene expression signature that signals the biochemical status of a lymphoma, thus clarifying whether it can be subdued by anti-CD40 therapy. The authors collected an array of cell lines derived from non-Hodgkin’s lymphomas that show a range of sensitivity to anti-CD40 therapy. By assessing gene expression before and after CD40 stimulation and creating a score that reflected CD40 pathway activation, Burington et al. found that cell lines with higher baseline activation of the CD40 pathway tended to be unresponsive to anti-CD40 stimulation. The researchers then identified a group of 15 active genes whose expression in formalin-fixed tissue (as would be obtained from patients) correctly predicted susceptibility to anti-CD40 treatment 77% of the time, a result verified in another set of cells lines and by quantitative polymerase chain reaction (PCR). Next, in a real-world test of the utility of this 15-gene predictor, tumor tissue samples from 39 patients who had been treated with dacetuzumab were scored for CD40 pathway activation with the new gene signature. A large majority (88%) of the patients predicted by the gene signature to be resistant to therapy in fact did not respond to therapy, showing a median progression-free survival of 40 days; 67% of those predicted to respond to dacetuzumab did so, with median progression-free survival extended to 169 days. These results encourage further testing in a prospective clinical trial designed to examine the ability of the 15-gene signature to predict which lymphoma patients will benefit from dacetuzumab treatment. If this index proves useful, it can be added to the catalog of prognostic tools at the service of the oncologist as they match drug to patient—without the need of a crystal ball. The primary function of B cells, critical components of the adaptive immune response, is to produce antibodies against foreign antigens, as well as to perform isotype class switching, which changes the heavy chain of an antibody so that it can interact with different repertoires of effector cells. CD40 is a member of the tumor necrosis factor superfamily of cell surface receptors that transmits survival signals to B cells. In contrast, in B cell cancers, stimulation of CD40 signaling results in a heterogeneous response in which cells can sometimes undergo cell death in response to treatment, depending on the system studied. We found an association between sensitivity to CD40 stimulation and mutation of the tumor suppressor p53 in a panel of non-Hodgkin’s lymphoma cell lines. Consistent with p53’s tumor suppressor role, we found that higher levels of intrinsic DNA damage and increased proliferation rates, as well as higher levels of BCL6, a transcriptional repressor proto-oncogene, were associated with sensitivity to CD40 stimulation. In addition, CD40 treatment–resistant cell lines were sensitized to CD40 stimulation after the introduction of DNA-damaging agents. Using gene expression analysis, we also showed that resistant cell lines exhibited a preexisting activated CD40 pathway and that an mRNA expression signature comprising CD40 target genes predicted sensitivity and resistance to CD40-activating agents in cell lines and mouse xenograft models. Finally, the gene signature predicted tumor shrinkage and progression-free survival in patients with diffuse large B cell lymphoma treated with dacetuzumab, a monoclonal antibody with partial CD40 agonist activity. These data show that CD40 pathway activation status may be useful in predicting the antitumor activity of CD40-stimulating therapeutic drugs.


Proceedings of the National Academy of Sciences of the United States of America | 2017

PRC2-mediated repression of SMARCA2 predicts EZH2 inhibitor activity in SWI/SNF mutant tumors

Thomas Januario; Xiaofen Ye; Russell Bainer; Bruno Alicke; Tunde Smith; Benjamin Haley; Zora Modrusan; Stephen E. Gould; Robert L. Yauch

Significance Targeting epigenetic dependencies caused by mutations in chromatin-modifying enzymes represents a novel therapeutic approach in cancer. Notably, cancers harboring mutations in the SNF5 subunit of the SWI/SNF chromatin remodeling complex have been shown to be susceptible to small-molecule inhibitors of the EZH2 histone methyltransferase that are currently in clinical development. We demonstrate that EZH2 inhibition can be effective in SMARCA4 mutant cancers that concurrently transcriptionally silence the paralog helicase SMARCA2. SMARCA2 is directly suppressed by EZH2, and SMARCA2 expression levels predict EZH2 inhibitor activity in other SWI/SNF mutant contexts, including ARID1A mutant tumors. These data provide insight into the utility of EZH2 inhibitors in SWI/SNF mutant tumors and have important implications regarding predictive diagnostics. Subunits of the SWI/SNF chromatin remodeling complex are frequently mutated in human cancers leading to epigenetic dependencies that are therapeutically targetable. The dependency on the polycomb repressive complex (PRC2) and EZH2 represents one such vulnerability in tumors with mutations in the SWI/SNF complex subunit, SNF5; however, whether this vulnerability extends to other SWI/SNF subunit mutations is not well understood. Here we show that a subset of cancers harboring mutations in the SWI/SNF ATPase, SMARCA4, is sensitive to EZH2 inhibition. EZH2 inhibition results in a heterogenous phenotypic response characterized by senescence and/or apoptosis in different models, and also leads to tumor growth inhibition in vivo. Lower expression of the SMARCA2 paralog was associated with cellular sensitivity to EZH2 inhibition in SMARCA4 mutant cancer models, independent of tissue derivation. SMARCA2 is suppressed by PRC2 in sensitive models, and induced SMARCA2 expression can compensate for SMARCA4 and antagonize PRC2 targets. The induction of SMARCA2 in response to EZH2 inhibition is required for apoptosis, but not for growth arrest, through a mechanism involving the derepression of the lysomal protease cathepsin B. Expression of SMARCA2 also delineates EZH2 inhibitor sensitivity for other SWI/SNF complex subunit mutant tumors, including SNF5 and ARID1A mutant cancers. Our data support monitoring SMARCA2 expression as a predictive biomarker for EZH2-targeted therapies in the context of SWI/SNF mutant cancers.

Collaboration


Dive into the Thomas Januario's collaboration.

Researchain Logo
Decentralizing Knowledge