Network


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

Hotspot


Dive into the research topics where Angela J. Waanders is active.

Publication


Featured researches published by Angela J. Waanders.


Nature Genetics | 2016

MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism.

Pratiti Bandopadhayay; Lori A. Ramkissoon; Payal Jain; Guillaume Bergthold; Jeremiah Wala; Rhamy Zeid; Steven E. Schumacher; Laura M. Urbanski; Ryan O'Rourke; William J. Gibson; Kristine Pelton; Shakti Ramkissoon; Harry J. Han; Yuankun Zhu; Namrata Choudhari; Amanda Silva; Katie Boucher; Rosemary E. Henn; Yun Jee Kang; David Knoff; Brenton R. Paolella; Adrianne Gladden-Young; Pascale Varlet; Mélanie Pagès; Peleg Horowitz; Alexander J. Federation; Hayley Malkin; Adam Tracy; Sara Seepo; Matthew Ducar

Angiocentric gliomas are pediatric low-grade gliomas (PLGGs) without known recurrent genetic drivers. We performed genomic analysis of new and published data from 249 PLGGs, including 19 angiocentric gliomas. We identified MYB-QKI fusions as a specific and single candidate driver event in angiocentric gliomas. In vitro and in vivo functional studies show that MYB-QKI rearrangements promote tumorigenesis through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression and hemizygous loss of the tumor suppressor QKI. To our knowledge, this represents the first example of a single driver rearrangement simultaneously transforming cells via three genetic and epigenetic mechanisms in a tumor.


Blood | 2015

MAPK signaling cascades mediate distinct glucocorticoid resistance mechanisms in pediatric leukemia

Courtney L. Jones; Christy M. Gearheart; Susan Fosmire; Cristina Delgado-Martin; Nikki A. Evensen; Karen L. Bride; Angela J. Waanders; Faye Pais; Jinhua Wang; Teena Bhatla; Danielle S. Bitterman; Simone R. de Rijk; Wallace Bourgeois; Smita Dandekar; Eugene Park; Tamara M. Burleson; Pillai Pallavi Madhusoodhan; David T. Teachey; Elizabeth A. Raetz; Michelle L. Hermiston; Markus Müschen; Mignon L. Loh; Stephen P. Hunger; Jinghui Zhang; Michael J. Garabedian; Christopher C. Porter; William L. Carroll

The outcome for pediatric acute lymphoblastic leukemia (ALL) patients who relapse is dismal. A hallmark of relapsed disease is acquired resistance to multiple chemotherapeutic agents, particularly glucocorticoids. In this study, we performed a genome-scale short hairpin RNA screen to identify mediators of prednisolone sensitivity in ALL cell lines. The incorporation of these data with an integrated analysis of relapse-specific genetic and epigenetic changes allowed us to identify the mitogen-activated protein kinase (MAPK) pathway as a mediator of prednisolone resistance in pediatric ALL. We show that knockdown of the specific MAPK pathway members MEK2 and MEK4 increased sensitivity to prednisolone through distinct mechanisms. MEK4 knockdown increased sensitivity specifically to prednisolone by increasing the levels of the glucocorticoid receptor. MEK2 knockdown increased sensitivity to all chemotherapy agents tested by increasing the levels of p53. Furthermore, we demonstrate that inhibition of MEK1/2 with trametinib increased sensitivity of ALL cells and primary samples to chemotherapy in vitro and in vivo. To confirm a role for MAPK signaling in patients with relapsed ALL, we measured the activation of the MEK1/2 target ERK in matched diagnosis-relapse primary samples and observed increased phosphorylated ERK levels at relapse. Furthermore, relapse samples have an enhanced response to MEK inhibition compared to matched diagnosis samples in xenograft models. Together, our data indicate that inhibition of the MAPK pathway increases chemosensitivity to glucocorticoids and possibly other agents and that the MAPK pathway is an attractive target for prevention and/or treatment of relapsed disease.


Cancer Cell | 2017

Integrated Molecular Meta-Analysis of 1,000 Pediatric High-Grade and Diffuse Intrinsic Pontine Glioma

Alan Mackay; Anna Burford; Diana Carvalho; Elisa Izquierdo; Janat Fazal-Salom; Kathryn R. Taylor; Lynn Bjerke; Matthew Clarke; Mara Vinci; Meera Nandhabalan; Sara Temelso; Sergey Popov; Valeria Molinari; Pichai Raman; Angela J. Waanders; Harry J. Han; Saumya Gupta; Lynley V. Marshall; Stergios Zacharoulis; Sucheta Vaidya; Henry Mandeville; Leslie R. Bridges; Andrew J. Martin; Safa Al-Sarraj; Christopher Chandler; Ho Keung Ng; Xingang Li; Kun Mu; Saoussen Trabelsi; Dorra H’mida-Ben Brahim

Summary We collated data from 157 unpublished cases of pediatric high-grade glioma and diffuse intrinsic pontine glioma and 20 publicly available datasets in an integrated analysis of >1,000 cases. We identified co-segregating mutations in histone-mutant subgroups including loss of FBXW7 in H3.3G34R/V, TOP3A rearrangements in H3.3K27M, and BCOR mutations in H3.1K27M. Histone wild-type subgroups are refined by the presence of key oncogenic events or methylation profiles more closely resembling lower-grade tumors. Genomic aberrations increase with age, highlighting the infant population as biologically and clinically distinct. Uncommon pathway dysregulation is seen in small subsets of tumors, further defining the molecular diversity of the disease, opening up avenues for biological study and providing a basis for functionally defined future treatment stratification.


Pediatric Blood & Cancer | 2016

Carboplatin Rechallenge After Hypersensitivity Reactions in Pediatric Patients With Low-Grade Glioma

Amish C. Shah; Jane E. Minturn; Yimei Li; Jean B. Belasco; Peter C. Phillips; Tammy I. Kang; Kristina A. Cole; Angela J. Waanders; Rosanna Pollack; Concetta DiDomenico; Cynthia Wildes; Michael J. Fisher

The high prevalence of carboplatin hypersensitivity reactions (HSR) significantly affects the treatment of pediatric patients with low‐grade glioma (LGG). Rechallenging patients is an option that must balance the risks of repeat allergic reaction to the benefits of retaining an effective anti‐tumor regimen.


Clinical Cancer Research | 2016

BRAF status in personalizing treatment approaches for pediatric gliomas

Aleksandra Olow; Sabine Mueller; Xiaodong Yang; Rintaro Hashizume; Justin G. Meyerowitz; William A. Weiss; Adam C. Resnick; Angela J. Waanders; Lukas J.A. Stalpers; Mitchel S. Berger; Nalin Gupta; C. David James; Claudia Petritsch; Daphne A. Haas-Kogan

Purpose: Alteration of the BRAF/MEK/MAPK pathway is the hallmark of pediatric low-grade gliomas (PLGGs), and mTOR activation has been documented in the majority of these tumors. We investigated combinations of MEK1/2, BRAFV600E and mTOR inhibitors in gliomas carrying specific genetic alterations of the MAPK pathway. Experimental Design: We used human glioma lines containing BRAFV600E (adult high-grade: AM-38, DBTRG, PLGG: BT40), or wild-type BRAF (pediatric high-grade: SF188, SF9427, SF8628) and isogenic systems of KIAA1549:BRAF-expressing NIH/3T3 cells and BRAFV600E-expressing murine brain cells. Signaling inhibitors included everolimus (mTOR), PLX4720 (BRAFV600E), and AZD6244 (MEK1/2). Proliferation was determined using ATP-based assays. In vivo inhibitor activities were assessed in the BT40 PLGG xenograft model. Results: In BRAFV600E cells, the three possible doublet combinations of AZD6244, everolimus, and PLX4720 exhibited significantly greater effects on cell viability. In BRAFWT cells, everolimus + AZD6244 was superior compared with respective monotherapies. Similar results were found using isogenic murine cells. In KIAA1549:BRAF cells, MEK1/2 inhibition reduced cell viability and S-phase content, effects that were modestly augmented by mTOR inhibition. In vivo experiments in the BRAFV600E pediatric xenograft model BT40 showed the greatest survival advantage in mice treated with AZD6244 + PLX4720 (P < 0.01). Conclusions: In BRAFV600E tumors, combination of AZD6244 + PLX4720 is superior to monotherapy and to other combinatorial approaches. In BRAFWT pediatric gliomas, everolimus + AZD6244 is superior to either agent alone. KIAA1549:BRAF-expressing tumors display marked sensitivity to MEK1/2 inhibition. Application of these results to PLGG treatment must be exercised with caution because the dearth of PLGG models necessitated only a single patient-derived PLGG (BT40) in this study. Clin Cancer Res; 22(21); 5312–21. ©2016 AACR.


Oncogene | 2017

CRAF gene fusions in pediatric low-grade gliomas define a distinct drug response based on dimerization profiles.

Payal Jain; T M Fierst; Harry J. Han; Tiffany Smith; Aesha Vakil; P J Storm; Adam C. Resnick; Angela J. Waanders

Pediatric low-grade gliomas (PLGGs) are commonly associated with BRAF gene fusions that aberrantly activate the mitogen-activated protein kinase (MAPK) signaling pathway. This has led to PLGG clinical trials utilizing RAF- and MAPK pathway-targeted therapeutics. Whole-genome profiling of PLGGs has also identified rare gene fusions involving another RAF isoform, CRAF/RAF1, in PLGGs and cancers occuring in adults. Whereas BRAF fusions primarily dysregulate MAPK signaling, the CRAF fusions QKI-RAF1 and SRGAP3-RAF1 aberrantly activate both the MAPK and phosphoinositide-3 kinase/mammalian target of rapamycin (PI3K/mTOR) signaling pathways. Although ATP-competitive, first-generation RAF inhibitors (vemurafenib/PLX4720, RAFi) cause paradoxical activation of the MAPK pathway in BRAF-fusion tumors, inhibition can be achieved with ‘paradox breaker’ RAFi, such as PLX8394. Here we report that, unlike BRAF fusions, CRAF fusions are unresponsive to both generations of RAFi, vemurafenib and PLX8394, highlighting a distinct responsiveness of CRAF fusions to clinically relevant RAFi. Whereas PLX8394 decreased BRAF-fusion dimerization, CRAF-fusion dimerization is unaffected primarily because of robust protein–protein interactions mediated by the N-terminal non-kinase fusion partner, such as QKI. The pan-RAF dimer inhibitor, LY3009120, could suppress CRAF-fusion oncogenicity by inhibiting dimer-mediated signaling. In addition, as CRAF fusions activate both the MAPK and PI3K/mTOR signaling pathways, we identify combinatorial inhibition of the MAPK/mTOR pathway as a potential therapeutic strategy for CRAF-fusion-driven tumors. Overall, we define a mechanistic distinction between PLGG-associated BRAF- and CRAF/RAF1 fusions in response to RAFi, highlighting the importance of molecularly classifying PLGG patients for targeted therapy. Furthermore, our study uncovers an important contribution of the non-kinase fusion partner to oncogenesis and potential therapeutic strategies against PLGG-associated CRAF fusions and possibly pan-cancer CRAF fusions.


Neuro-oncology | 2018

Pediatric low-grade gliomas: Next biologically driven steps

David T. W. Jones; Mark W. Kieran; Eric Bouffet; Sanda Alexandrescu; Pratiti Bandopadhayay; Miriam Bornhorst; David W. Ellison; Jason Fangusaro; Michael J. Fisher; Nicholas K. Foreman; Maryam Fouladi; Darren Hargrave; Cynthia Hawkins; Nada Jabado; Maura Massimino; Sabine Mueller; Giorgio Perilongo; Antoinette Y.N. Schouten Van Meeteren; Uri Tabori; Katherine Warren; Angela J. Waanders; David Walker; William A. Weiss; Olaf Witt; Karen Wright; Daniel C. Bowers; Stefan M. Pfister; Roger J. Packer

Despite the fact that they are not typically life-threatening, low-grade gliomas (LGGs) remain a significant clinical challenge in pediatric neuro-oncology due to comorbidities associated with these tumors and/or their treatments, and their propensity to multiply recurs. LGGs, in total the most common brain tumors arising in childhood, can often become a chronic problem requiring decades of management. The Second International Consensus Conference on Pediatric Low-Grade Gliomas held in Padua, Italy in 2016 was convened in an attempt to advance the pace of translating biological discoveries on LGGs into meaningful clinical benefit. Topics discussed included: the implications of our growing biological understanding of the genomics underlying these tumors; the assessment of the model systems available; the implications of the molecular and histopathologic differences between adult and pediatric diffuse gliomas; and steps needed to expedite targeted therapy into late-stage clinical trials for newly diagnosed cases. Methods for the diagnostic assessment of alterations in the Ras/mitogen-activated protein kinase pathway, typical for these tumors, were also considered. While the overall tone was positive, with a consensus that progress is being and will continue to be made, the scale of the challenge presented by this complex group of tumors was also acknowledged. The conclusions and recommendations of the meeting panel are provided here as an outline of current thinking and a basis for further discussion.


Journal of Neuropathology and Experimental Neurology | 2016

Whole Chromosome 7 Gain Predicts Higher Risk of Recurrence in Pediatric Pilocytic Astrocytomas Independently From KIAA1549-BRAF Fusion Status.

Jacquelyn J. Roth; Tamara M. Fierst; Angela J. Waanders; Li Yimei; Jaclyn A. Biegel; Mariarita Santi

The most frequent genetic alteration identified in pediatric pilocytic astrocytomas and pilomyxoid variant is the KIAA1549-BRAF fusion, which typically results from a 2.0 Mb tandem duplication in chromosome band 7q34. Less frequent abnormalities include fusion genes, BRAF, FGFR, KRAS, and NF1 point mutations, and whole chromosome gains. To correlate genetic alterations with clinical course data, we retrospectively analyzed the tumors with pilocytic and pilomyxoid histology of a cohort of 116 pediatric patients, aged 5 months to 23 years. Gross total resection was associated with a decreased risk of recurrence (p = 0.001), supporting previous findings that complete tumor excision correlates with long-term and disease-free survival. We found no significant association between recurrence rate and the presence of the KIAA1549-BRAF fusion or BRAF mutation (p = 0.167). Interestingly, gain of whole chromosome 7 (WC7) was associated with a 4.7-fold increased risk of tumor recurrence, even after adjusting for surgical status (p = 0.025), and other genetic alterations. Using fluorescence in situ hybridization, we demonstrated that when WC7 gain accompanies the KIAA1549-BRAF fusion, the fusion likely arises first. This study highlights the utility of genetic studies for risk assessment of pilocytic and pilomyxoid astrocytomas, which may impact treatment selections.


Cancer Cell | 2018

Molecular, Pathological, Radiological, and Immune Profiling of Non-brainstem Pediatric High-Grade Glioma from the HERBY Phase II Randomized Trial

Alan Mackay; Anna Burford; Valeria Molinari; David T. W. Jones; Elisa Izquierdo; Jurriaan Brouwer-Visser; Felice Giangaspero; Christine Haberler; Torsten Pietsch; Ts Jacques; Dominique Figarella-Branger; Daniel Rodriguez; Paul S. Morgan; Pichai Raman; Angela J. Waanders; Adam C. Resnick; Maura Massimino; Maria Luisa Garrè; Helen Smith; David Capper; Stefan M. Pfister; Thomas Wurdinger; Rachel Tam; Josep Garcia; Meghna Das Thakur; Gilles Vassal; Jacques Grill; Tim Jaspan; Pascale Varlet; Chris Jones

Summary The HERBY trial was a phase II open-label, randomized, multicenter trial evaluating bevacizumab (BEV) in addition to temozolomide/radiotherapy in patients with newly diagnosed non-brainstem high-grade glioma (HGG) between the ages of 3 and 18 years. We carried out comprehensive molecular analysis integrated with pathology, radiology, and immune profiling. In post-hoc subgroup analysis, hypermutator tumors (mismatch repair deficiency and somatic POLE/POLD1 mutations) and those biologically resembling pleomorphic xanthoastrocytoma ([PXA]-like, driven by BRAF_V600E or NF1 mutation) had significantly more CD8+ tumor-infiltrating lymphocytes, and longer survival with the addition of BEV. Histone H3 subgroups (hemispheric G34R/V and midline K27M) had a worse outcome and were immune cold. Future clinical trials will need to take into account the diversity represented by the term “HGG” in the pediatric population.


Oncotarget | 2017

Overcoming resistance to single-agent therapy for oncogenic BRAF gene fusions via combinatorial targeting of MAPK and PI3K/mTOR signaling pathways

Payal Jain; Amanda Silva; Harry J. Han; Shih-Shan Lang; Yuankun Zhu; Katie Boucher; Tiffany Smith; Aesha Vakil; Patrick Diviney; Namrata Choudhari; Pichai Raman; Christine Busch; Tim Delaney; Xiaodong Yang; Aleksandra Olow; Sabine Mueller; Daphne A. Haas-Kogan; Elizabeth Fox; Phillip B. Storm; Adam C. Resnick; Angela J. Waanders

Pediatric low-grade gliomas (PLGGs) are frequently associated with activating BRAF gene fusions, such as KIAA1549-BRAF, that aberrantly drive the mitogen activated protein kinase (MAPK) pathway. Although RAF inhibitors (RAFi) have been proven effective in BRAF-V600E mutant tumors, we have previously shown how the KIAA1549-BRAF fusion can be paradoxically activated by RAFi. While newer classes of RAFi, such as PLX8394, have now been shown to inhibit MAPK activation by KIAA1549-BRAF, we sought to identify alternative MAPK pathway targeting strategies using clinically relevant MEK inhibitors (MEKi), along with potential escape mechanisms of acquired resistance to single-agent MAPK pathway therapies. We demonstrate effectiveness of multiple MEKi against diverse BRAF-fusions with novel N-terminal partners, with trametinib being the most potent. However, resistance to MEKi or PLX8394 develops via increased RTK expression causing activation of PI3K/mTOR pathway in BRAF-fusion expressing resistant clones. To circumvent acquired resistance, we show potency of combinatorial targeting with trametinib and everolimus, an mTOR inhibitor (mTORi) against multiple BRAF-fusions. While single-agent mTORi and MEKi PLGG clinical trials are underway, our study provides preclinical rationales for using MEKi and mTORi combinatorial therapy to stave off or prevent emergent drug-resistance in BRAF-fusion driven PLGGs.

Collaboration


Dive into the Angela J. Waanders's collaboration.

Top Co-Authors

Avatar

Adam C. Resnick

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Sabine Mueller

University of California

View shared research outputs
Top Co-Authors

Avatar

Phillip B. Storm

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Payal Jain

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Pichai Raman

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Javad Nazarian

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yuankun Zhu

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Amanda Silva

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mariarita Santi

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge