Network


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

Hotspot


Dive into the research topics where Andrea Griesinger is active.

Publication


Featured researches published by Andrea Griesinger.


Cancer Discovery | 2014

Autophagy inhibition improves chemosensitivity in BRAF(V600E) brain tumors.

Jean M. Mulcahy Levy; Joshua C. Thompson; Andrea Griesinger; Vladimir Amani; Andrew M. Donson; Diane K. Birks; Michael J. Morgan; David M. Mirsky; Michael H. Handler; Nicholas K. Foreman; Andrew Thorburn

UNLABELLED Autophagy inhibition is a potential therapeutic strategy in cancer, but it is unknown which tumors will benefit. The BRAF(V600E) mutation has been identified as important in pediatric central nervous system (CNS) tumors and is known to affect autophagy in other tumor types. We evaluated CNS tumor cells with BRAF(V600E) and found that mutant (but not wild-type) cells display high rates of induced autophagy, are sensitive to pharmacologic and genetic autophagy inhibition, and display synergy when the clinically used autophagy inhibitor chloroquine was combined with the RAF inhibitor vemurafenib or standard chemotherapeutics. Importantly, we also demonstrate that chloroquine can improve vemurafenib sensitivity in a resistant ex vivo primary culture and provide the first demonstration in a patient harboring the V600E mutation treated with vemurafenib that the addition of chloroquine can improve clinical outcomes. These findings suggest that CNS tumors with BRAF(V600E) are autophagy-dependent and should be targeted with autophagy inhibition in combination with other therapeutic strategies. SIGNIFICANCE Autophagy inhibition may improve cancer therapy, but it is unclear which tumors will benefit. We found that BRAF mutations cause brain tumor cells to depend on autophagy and display selective chemosensitization with autophagy inhibition. We present a pediatric case in which deliberate autophagy inhibition halted tumor growth and overcame acquired BRAF-inhibition resistance.


Journal of Immunology | 2013

Characterization of Distinct Immunophenotypes across Pediatric Brain Tumor Types

Andrea Griesinger; Diane K. Birks; Andrew M. Donson; Vladimir Amani; Lindsey M. Hoffman; Allen Waziri; Michael Wang; Michael H. Handler; Nicholas K. Foreman

Despite increasing evidence that antitumor immune control exists in the pediatric brain, these findings have yet to be exploited successfully in the clinic. A barrier to development of immunotherapeutic strategies in pediatric brain tumors is that the immunophenotype of these tumors’ microenvironment has not been defined. To address this, the current study used multicolor FACS of disaggregated tumor to systematically characterize the frequency and phenotype of infiltrating immune cells in the most common pediatric brain tumor types. The initial study cohort consisted of 7 pilocytic astrocytoma (PA), 19 ependymoma (EPN), 5 glioblastoma (GBM), 6 medulloblastoma (MED), and 5 nontumor brain (NT) control samples obtained from epilepsy surgery. Immune cell types analyzed included both myeloid and T cell lineages and respective markers of activated or suppressed functional phenotypes. Immune parameters that distinguished each of the tumor types were identified. PA and EPN demonstrated significantly higher infiltrating myeloid and lymphoid cells compared with GBM, MED, or NT. Additionally, PA and EPN conveyed a comparatively activated/classically activated myeloid cell–skewed functional phenotype denoted in particular by HLA-DR and CD64 expression. In contrast, GBM and MED contained progressively fewer infiltrating leukocytes and more muted functional phenotypes similar to that of NT. These findings were recapitulated using whole tumor expression of corresponding immune marker genes in a large gene expression microarray cohort of pediatric brain tumors. The results of this cross-tumor comparative analysis demonstrate that different pediatric brain tumor types exhibit distinct immunophenotypes, implying that specific immunotherapeutic approaches may be most effective for each tumor type.


Acta Neuropathologica | 2014

Molecular sub-group-specific immunophenotypic changes are associated with outcome in recurrent posterior fossa ependymoma.

Lindsey M. Hoffman; Andrew M. Donson; Ichiro Nakachi; Andrea Griesinger; Diane K. Birks; Vladimir Amani; Molly S. Hemenway; Arthur K. Liu; Michael Wang; Todd C. Hankinson; Michael H. Handler; Nicholas K. Foreman

Better understanding of ependymoma (EPN) biology at relapse is needed to improve therapy at this critical event. Convincing data exist defining transcriptionally distinct posterior fossa (PF) sub-groups A and B at diagnosis. The clinical and biological consequence of these sub-groups at recurrence has not yet been defined. Genome and transcriptome microarray profiles and clinical variables of matched primary and first recurrent PF EPN pairs were used to identify biologically distinct patterns of progression between EPN sub-groups at recurrence. Key findings were validated by histology and immune function assays. Transcriptomic profiles were partially conserved at recurrence. However, 4 of 14 paired samples changed sub-groups at recurrence, and significant sub-group-specific transcriptomic changes between primary and recurrent tumors were identified, which were predominantly immune-related. Further examination revealed that Group A primary tumors harbor an immune gene signature and cellular functionality consistent with an immunosuppressive phenotype associated with tissue remodeling and wound healing. Conversely, Group B tumors develop an adaptive, antigen-specific immune response signature and increased T-cell infiltration at recurrence. Clinical distinctions between sub-groups become more apparent after first recurrence. Group A tumors were more often sub-totally resected and had a significantly shorter time to subsequent progression and worse overall survival. Minimal tumor-specific genomic changes were observed for either PF Groups A or B at recurrence. Molecular sub-groups of PF EPN convey distinct immunobiologic signatures at diagnosis and recurrence, providing potential biologic rationale to their disparate clinical outcomes. Immunotherapeutic approaches may be warranted, particularly in Group A PF EPN.


Cancer immunology research | 2015

Interleukin-6/STAT3 pathway signaling drives an inflammatory phenotype in Group A ependymoma

Andrea Griesinger; Rebecca Josephson; Andrew M. Donson; Jean M. Mulcahy Levy; Vladimir Amani; Diane K. Birks; Lindsey M. Hoffman; Steffanie L. Furtek; Phillip Reigan; Michael H. Handler; Rajeev Vibhakar; Nicholas K. Foreman

Subgroup A ependymoma are brain tumors with a poor prognosis. Tumors were found to be IL6/STAT3-dependent and infiltrated with polarized myeloid cells. Targeting this pathway to relieve immunosuppression could be an important approach for this tumor type. Ependymoma (EPN) in childhood is a brain tumor with substantial mortality. Inflammatory response has been identified as a molecular signature of high-risk Group A EPN. To better understand the biology of this phenotype and aid therapeutic development, transcriptomic data from Group A and B EPN patient tumor samples, and additional malignant and normal brain data, were analyzed to identify the mechanism underlying EPN Group A inflammation. Enrichment of IL6 and STAT3 pathway genes were found to distinguish Group A EPN from Group B EPN and other brain tumors, implicating an IL6 activation of STAT3 mechanism. EPN tumor cell growth was shown to be dependent on STAT3 activity, as demonstrated using shRNA knockdown and pharmacologic inhibition of STAT3 that blocked proliferation and induced apoptosis. The inflammatory factors secreted by EPN tumor cells were shown to reprogram myeloid cells, and this paracrine effect was characterized by a significant increase in pSTAT3 and IL8 secretion. Myeloid polarization was shown to be dependent on tumor secretion of IL6, and these effects could be reversed using IL6-neutralizing antibody or IL6 receptor–targeted therapeutic antibody tocilizumab. Polarized myeloid cell production of IL8 drove unpolarized myeloid cells to upregulate CD163 and to produce a number of proinflammatory cytokines. Collectively, these findings indicate that constitutive IL6/STAT3 pathway activation is important in driving tumor growth and inflammatory cross-talk with myeloid cells within the Group A EPN microenvironment. Effective design of Group A–targeted therapy for children with EPN may require reversal of this potentially immunosuppressive and protumor pathway. Cancer Immunol Res; 3(10); 1165–74. ©2015 AACR.


eLife | 2017

Autophagy inhibition overcomes multiple mechanisms of resistance to BRAF inhibition in brain tumors

Jean M. Mulcahy Levy; Shadi Zahedi; Andrea Griesinger; Andrew Morin; Kurtis D. Davies; Dara L. Aisner; B. K. Kleinschmidt-DeMasters; Brent E. Fitzwalter; Megan L. Goodall; Jacqueline Thorburn; Vladimir Amani; Andrew M. Donson; Diane K. Birks; David M. Mirsky; Todd C. Hankinson; Michael H. Handler; Adam Green; Rajeev Vibhakar; Nicholas K. Foreman; Andrew Thorburn

Kinase inhibitors are effective cancer therapies, but tumors frequently develop resistance. Current strategies to circumvent resistance target the same or parallel pathways. We report here that targeting a completely different process, autophagy, can overcome multiple BRAF inhibitor resistance mechanisms in brain tumors. BRAFV600Emutations occur in many pediatric brain tumors. We previously reported that these tumors are autophagy-dependent and a patient was successfully treated with the autophagy inhibitor chloroquine after failure of the BRAFV600E inhibitor vemurafenib, suggesting autophagy inhibition overcame the kinase inhibitor resistance. We tested this hypothesis in vemurafenib-resistant brain tumors. Genetic and pharmacological autophagy inhibition overcame molecularly distinct resistance mechanisms, inhibited tumor cell growth, and increased cell death. Patients with resistance had favorable clinical responses when chloroquine was added to vemurafenib. This provides a fundamentally different strategy to circumvent multiple mechanisms of kinase inhibitor resistance that could be rapidly tested in clinical trials in patients with BRAFV600E brain tumors. DOI: http://dx.doi.org/10.7554/eLife.19671.001


Journal of Neuropathology and Experimental Neurology | 2017

Molecular Analyses Reveal Inflammatory Mediators in the Solid Component and Cyst Fluid of Human Adamantinomatous Craniopharyngioma.

Andrew M. Donson; John R. Apps; Andrea Griesinger; Vladimir Amani; Davis Witt; Richard C. E. Anderson; Toba Niazi; Gerald A. Grant; Mark Souweidane; James M. Johnston; Eric M. Jackson; B. K. Kleinschmidt-DeMasters; Michael H. Handler; Aik Choon Tan; Lia Gore; Alex Virasami; Jose Mario Gonzalez-Meljem; Ts Jacques; Juan Pedro Martinez-Barbera; Nicholas K. Foreman; Todd C. Hankinson

Pediatric adamantinomatous craniopharyngioma (ACP) is a highly solid and cystic tumor, often causing substantial damage to critical neuroendocrine structures such as the hypothalamus, pituitary gland, and optic apparatus. Paracrine signaling mechanisms driving tumor behavior have been hypothesized, with IL-6R overexpression identified as a potential therapeutic target. To identify potential novel therapies, we characterized inflammatory and immunomodulatory factors in ACP cyst fluid and solid tumor components. Cytometric bead analysis revealed a highly pro-inflammatory cytokine pattern in fluid from ACP compared to fluids from another cystic pediatric brain tumor, pilocytic astrocytoma. Cytokines and chemokines with particularly elevated concentrations in ACPs were IL-6, CXCL1 (GRO), CXCL8 (IL-8) and the immunosuppressive cytokine IL-10. These data were concordant with solid tumor compartment transcriptomic data from a larger cohort of ACPs, other pediatric brain tumors and normal brain. The majority of receptors for these cytokines and chemokines were also over-expressed in ACPs. In addition to IL-10, the established immunosuppressive factor IDO-1 was overexpressed by ACPs at the mRNA and protein levels. These data indicate that ACP cyst fluids and solid tumor components are characterized by an inflammatory cytokine and chemokine expression pattern. Further study regarding selective cytokine blockade may inform novel therapeutic interventions.


Oncotarget | 2016

Checkpoint kinase 1 expression is an adverse prognostic marker and therapeutic target in MYC-driven medulloblastoma

Eric Prince; Ilango Balakrishnan; Monil Shah; Jean M. Mulcahy Levy; Andrea Griesinger; Irina Alimova; Peter Harris; Diane K. Birks; Andrew M. Donson; Nathan Davidson; Marc Remke; Michael D. Taylor; Michael H. Handler; Nicholas K. Foreman; Sujatha Venkataraman; Rajeev Vibhakar

Checkpoint kinase 1 (CHK1) is an integral component of the cell cycle as well as the DNA Damage Response (DDR) pathway. Previous work has demonstrated the effectiveness of inhibiting CHK1 with small-molecule inhibitors, but the role of CHK1 mediated DDR in medulloblastoma is unknown. CHK1, both at the mRNA and protein level, is highly expressed in medulloblastoma and elevated CHK1 expression in Group3 medulloblastoma is an adverse prognostic marker. CHK1 inhibition with the small-molecule drug AZD7762, results in decreased cell growth, increased DNA damage and cell apoptosis. Furthermore, AZD7762 acts in synergy with cisplatin in reducing cell proliferation in medulloblastoma. Similar phenotypic changes were observed with another CHK1 inhibitor, PF477736, as well as genetic knockdown using siRNA against CHK1. Treatments with small-molecule inhibitors of CHK1 profoundly modulated the expression of both upstream and downstream target proteins within the CHK1 signaling pathways. This suggests the presence of a feedback loop in activating CHK1. Overall, our results demonstrate that small-molecule inhibition of CHK1 in combination with, cisplatin, is more advantageous than either treatment alone, especially for Group 3 medulloblastoma, and therefore this combined therapeutic approach serves as an avenue for further investigation.


OncoImmunology | 2014

Immunotherapeutic implications of the immunophenotype of pediatric brain tumors

Andrea Griesinger; Andrew M. Donson; Nicholas K. Foreman

The cytofluorometric analysis of dissociated tumor samples identified distinct immunophenotypes among the most common variants of pediatric brain tumor. These findings suggest that immunotherapeutic regimens against pediatric brain malignancies should be tailored to individual tumor types.


Journal for ImmunoTherapy of Cancer | 2018

Durable regression of Medulloblastoma after regional and intravenous delivery of anti-HER2 chimeric antigen receptor T cells

Anandani Nellan; Christopher Rota; Robbie G. Majzner; Cynthia M. Lester-McCully; Andrea Griesinger; Jean M. Mulcahy Levy; Nicholas K. Foreman; Katherine E. Warren; Daniel W. Lee

BackgroundStandard-of-care therapies for treating pediatric medulloblastoma have long-term side effects, even in children who are cured. One emerging modality of cancer therapy that could be equally effective without such side effects would be chimeric antigen receptor (CAR) T cells. Knowing that human epidermal growth factor receptor 2 (HER2) is overexpressed in many medulloblastomas and has been used as a CAR T target before, we sought to evaluate the efficacy of more sophisticated anti-HER2 CAR T cells, as well as the feasibility and efficacy of different routes of delivering these cells, for the treatment of pediatric medulloblastoma.MethodsDaoy, D283 and D425 medulloblastoma cell lines were characterized by flow cytometry to evaluate HER2 expression. Anti-tumor efficacy of HER2-BBz-CAR T cells in vitro was performed using cytokine release and immune cytotoxicity assays compared to control CD19 CAR T cells. In vivo, Daoy and D283 tumor cells were orthotopically implanted in the posterior fossa of NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice and treated with regional or intravenous HER2-BBz-CAR T cells or control CD19 CAR T cells. Non-human primates (NHPs) bearing ventricular and lumbar reservoirs were treated with target autologous cells bearing extracellular HER2 followed by autologous HER2-CAR T cells intraventricularly. Cerebrospinal fluid and blood were collected serially to measure the persistence of delivered cells and cytokines.ResultsHER2-BBz-CAR T cells effectively clear medulloblastoma orthotopically implanted in the posterior fossa of NSG mice via both regional and intravenous delivery in xenograft models. Intravenous delivery requires a log higher dose compared to regional delivery. NHPs tolerated intraventricular delivery of autologous cells bearing extracellular HER2 followed by HER2-BBz-CAR T cells without experiencing any systemic toxicity.ConclusionsHER2-BBz-CAR T cells show excellent pre-clinical efficacy in vitro and in mouse medulloblastoma models, and their intraventricular delivery is feasible and safe in NHPs. A clinical trial of HER2-BBz-CAR T cells directly delivered into cerebrospinal fluid should be designed for patients with relapsed medulloblastoma.


Pediatric Blood & Cancer | 2018

Specific expression of PD‐L1 in RELA‐fusion supratentorial ependymoma: Implications for PD‐1‐targeted therapy

Davis Witt; Andrew M. Donson; Vladimir Amani; Daniel C. Moreira; Bridget Sanford; Lindsey M. Hoffman; Michael H. Handler; Jean M. Mulcahy Levy; Kenneth L. Jones; Anandani Nellan; Nicholas K. Foreman; Andrea Griesinger

A desperate need for novel therapies in pediatric ependymoma (EPN) exists, as chemotherapy remains ineffective and radiotherapy often fails. EPN have significant infiltration of immune cells, which correlates with outcome. Immune checkpoint inhibitors provide an avenue for new treatments. This study characterizes tumor‐infiltrating immune cells in EPN and aims at predicting candidates for clinical trials using checkpoint inhibitors targeting PD‐L1/PD‐1 (programmed death ligand 1/programmed death 1).

Collaboration


Dive into the Andrea Griesinger's collaboration.

Top Co-Authors

Avatar

Nicholas K. Foreman

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Andrew M. Donson

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Michael H. Handler

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Vladimir Amani

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Rajeev Vibhakar

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Diane K. Birks

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Todd C. Hankinson

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lindsey M. Hoffman

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Davis Witt

Anschutz Medical Campus

View shared research outputs
Researchain Logo
Decentralizing Knowledge