Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Sci Rep ; 13(1): 13375, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37591961

ABSTRACT

Sign-tracking is a conditioned response where animals interact with reward-predictive cues due to the cues having motivational value, or incentive salience. The nucleus accumbens core (NAc) has been implicated in mediating the sign-tracking response. Additionally, acetylcholine (ACh) transmission throughout the striatum has been attributed to both incentive motivation and behavioral flexibility. Here, we demonstrate a role for NAc ACh receptors in the flexibility of sign-tracking. Sign-tracking animals were exposed to an omission contingency, in which vigorous sign-tracking was punished by reward omission. Animals rapidly adjusted their behavior, but they maintained sign-tracking in a less vigorous manner that did not cancel reward. Within this context of sign-tracking being persistent yet flexible in structure, blockade of NAc nicotinic receptors (nAChRs) led to a persistence in the initial sign-tracking response during omission followed by a period of change in the makeup of sign-tracking, whereas blockade of muscarinic receptors (mAChRs) oppositely enhanced the omission-related development of the new sign-tracking behaviors. Later, once omission learning had occurred, nAChR blockade uniquely led to reduced sign-tracking and elevated reward-directed behaviors instead. These results indicate that NAc ACh receptors have opposing roles in maintaining learned patterns of sign-tracking, with nAChRs having a special involvement in regulating the structure of the sign-tracking response.


Subject(s)
Motivation , Receptors, Nicotinic , Animals , Cues , Nucleus Accumbens , Learning
2.
bioRxiv ; 2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36711550

ABSTRACT

The dorsolateral striatum (DLS) is linked to the learning and honing of action routines. However, the DLS is also important for performing behaviors that have been successful in the past. The learning function can be thought of as prospective, helping to plan ongoing actions to be efficient and often optimal. The performance function is more retrospective, helping the animal continue to behave in a way that had worked previously. How the DLS manages this all is curious. What happens when a learned behavior becomes sub-optimal due to environment changes. In this case, the prospective function of the DLS would cause animals to (adaptively) learn and plan more optimal actions. In contrast, the retrospective function would cause animals to (maladaptively) favor the old behavior. Here we find that, during a change in learned task rules, DLS inhibition causes animals to adjust less rapidly to the new task (and to behave less vigorously) in a 'maladaptive' way. Yet, when the task is changed back to the initially learned rules, DLS inhibition instead causes a rapid and vigorous adjustment of behavior in an 'adaptive' way. These results show that inhibiting the DLS biases behavior towards initially acquired strategies, implying a more retrospective outlook in action selection when the DLS is offline. Thus, an active DLS could encourage planning and learning action routines more prospectively. Moreover, the DLS control over behavior can appear to be either advantageous/flexible or disadvantageous/inflexible depending on task context, and its control over vigor can change depending on task context. Significant Statement: Basal ganglia networks aid behavioral learning (a prospective planning function) but also favor the use of old behaviors (a retrospective performance function), making it unclear what happens when learned behaviors become suboptimal. Here we inhibit the dorsolateral striatum (DLS) as animals encounter a change in task rules, and again when they shift back to those learned task rules. DLS inhibition reduces adjustment to new task rules (and reduces behavioral vigor), but it increases adjustment back to the initially learned task rules later (and increases vigor). Thus, in both cases, DLS inhibition favored the use of the initially learned behavioral strategy, which could appear either maladaptive or adaptive. We suggest that the DLS might promote a prospective orientation of action control.

3.
Nature ; 609(7929): 1012-1020, 2022 09.
Article in English | MEDLINE | ID: mdl-36131015

ABSTRACT

Medulloblastoma, a malignant childhood cerebellar tumour, segregates molecularly into biologically distinct subgroups, suggesting that a personalized approach to therapy would be beneficial1. Mouse modelling and cross-species genomics have provided increasing evidence of discrete, subgroup-specific developmental origins2. However, the anatomical and cellular complexity of developing human tissues3-particularly within the rhombic lip germinal zone, which produces all glutamatergic neuronal lineages before internalization into the cerebellar nodulus-makes it difficult to validate previous inferences that were derived from studies in mice. Here we use multi-omics to resolve the origins of medulloblastoma subgroups in the developing human cerebellum. Molecular signatures encoded within a human rhombic-lip-derived lineage trajectory aligned with photoreceptor and unipolar brush cell expression profiles that are maintained in group 3 and group 4 medulloblastoma, suggesting a convergent basis. A systematic diagnostic-imaging review of a prospective institutional cohort localized the putative anatomical origins of group 3 and group 4 tumours to the nodulus. Our results connect the molecular and phenotypic features of clinically challenging medulloblastoma subgroups to their unified beginnings in the rhombic lip in the early stages of human development.


Subject(s)
Cell Lineage , Cerebellar Neoplasms , Medulloblastoma , Metencephalon , Animals , Cerebellar Neoplasms/classification , Cerebellar Neoplasms/embryology , Cerebellar Neoplasms/pathology , Cerebellum/embryology , Humans , Medulloblastoma/classification , Medulloblastoma/embryology , Medulloblastoma/pathology , Metencephalon/embryology , Mice , Neurons/pathology , Prospective Studies
4.
Mol Cancer Ther ; 21(8): 1306-1317, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35709750

ABSTRACT

Group3 (G3) medulloblastoma (MB) is one of the deadliest forms of the disease for which novel treatment is desperately needed. Here we evaluate ribociclib, a highly selective CDK4/6 inhibitor, with gemcitabine in mouse and human G3MBs. Ribociclib central nervous system (CNS) penetration was assessed by in vivo microdialysis and by IHC and gene expression studies and found to be CNS-penetrant. Tumors from mice treated with short term oral ribociclib displayed inhibited RB phosphorylation, downregulated E2F target genes, and decreased proliferation. Survival studies to determine the efficacy of ribociclib and gemcitabine combination were performed on mice intracranially implanted with luciferase-labeled mouse and human G3MBs. Treatment of mice with the combination of ribociclib and gemcitabine was well tolerated, slowed tumor progression and metastatic spread, and increased survival. Expression-based gene activity and cell state analysis investigated the effects of the combination after short- and long-term treatments. Molecular analysis of treated versus untreated tumors showed a significant decrease in the activity and expression of genes involved in cell-cycle progression and DNA damage response, and an increase in the activity and expression of genes implicated in neuronal identity and neuronal differentiation. Our findings in both mouse and human patient-derived orthotopic xenograft models suggest that ribociclib and gemcitabine combination therapy warrants further investigation as a treatment strategy for children with G3MB.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Cerebellar Neoplasms/drug therapy , Child , Deoxycytidine/analogs & derivatives , Humans , Medulloblastoma/drug therapy , Mice , Purines , Gemcitabine
6.
STAR Protoc ; 3(2): 101292, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35463474

ABSTRACT

This protocol summarizes the pipeline for analysis of tumor-derived cell-free DNA (cfDNA) from cerebrospinal fluid (CSF) using low-coverage whole-genome sequencing (lcWGS). This approach enables resolution of chromosomal and focal copy-number variations (CNVs) as oncologic signatures, particularly for patients with central nervous system tumors. Our strategy tolerates sub-nanogram cfDNA input and is thus optimized for CSF samples where cfDNA yields are typically low. Overall, the detection of tumor-specific signatures in CSF-derived cfDNA is a promising biomarker for personalization of brain-tumor therapy. For complete details on the use and execution of this protocol, please refer to Liu et al. (2021).


Subject(s)
Brain Neoplasms , Cell-Free Nucleic Acids , Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/genetics , Cell-Free Nucleic Acids/cerebrospinal fluid , Cell-Free Nucleic Acids/genetics , DNA Copy Number Variations/genetics , Humans , Whole Genome Sequencing
7.
Brain Res ; 1784: 147886, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35314149

ABSTRACT

Current treatments for obesity do not reliably reduce body weight over time. New interventional strategies, including chemogenetics, carry promise based on preclinical animal studies. Here, we focused on the ventral pallidum (VP) due to its clearly established role in eating behavior. Chronic inhibitory or excitatory chemogenetic activation was used to modulate the activity of VP-targeted neurons in rats on an obesogenic diet. Based on studies using acute VP manipulations, we hypothesized that VP inhibition would decrease weight gain, while VP stimulation would increase weight. Instead, both manipulations caused weight gain over time, and in a manner not clearly linked to consumption levels. We theorize that the complex reciprocal feedback between ventral striatal structures and metabolic centers likely underpin our unexpected findings. Regardless, this study suggests that the result of strategies to prevent obesity with chronic neuromodulation could be difficult to predict from prior preclinical studies that have used acute interventions.


Subject(s)
Basal Forebrain , Animals , Basal Forebrain/physiology , Diet , Male , Neurons/physiology , Obesity , Rats , Reward , Weight Gain
8.
Neuro Oncol ; 24(7): 1178-1190, 2022 07 01.
Article in English | MEDLINE | ID: mdl-34935967

ABSTRACT

BACKGROUND: Embryonal tumors of the CNS are the most common malignant tumors occurring in the first years of life. This study evaluated the feasibility and safety of incorporating novel non-cytotoxic therapy with vorinostat and isotretinoin to an intensive cytotoxic chemotherapy backbone. METHODS: PBTC-026 was a prospective multi-institutional clinical trial for children <48 months of age with newly diagnosed embryonal tumors of the CNS. Treatment included three 21-day cycles of induction therapy with vorinostat and isotretinoin, cisplatin, vincristine, cyclophosphamide, and etoposide; three 28-day cycles of consolidation therapy with carboplatin and thiotepa followed by stem cell rescue; and twelve 28-day cycles of maintenance therapy with vorinostat and isotretinoin. Patients with M0 medulloblastoma (MB) received focal radiation following consolidation therapy. Molecular classification was by DNA methylation array. RESULTS: Thirty-one patients with median age of 26 months (range 6-46) received treatment on study; 19 (61%) were male. Diagnosis was MB in 20 and supratentorial CNS embryonal tumor in 11. 24/31 patients completed induction therapy within a pre-specified feasibility window of 98 days. Five-year progression-free survival (PFS) and overall survival (OS) for all 31 patients were 55 ± 15 and 61 ± 13, respectively. Five-year PFS was 42 ± 13 for group 3 MB (n = 12); 80 ± 25 for SHH MB (n = 5); 33 ± 19 for embryonal tumor with multilayered rosettes (ETMR, n = 6). CONCLUSION: It was safe and feasible to incorporate vorinostat and isotretinoin into an intensive chemotherapy regimen. Further study to define efficacy in this high-risk group of patients is warranted.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Medulloblastoma , Neoplasms, Germ Cell and Embryonal , Neuroectodermal Tumors, Primitive , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/pathology , Cerebellar Neoplasms/drug therapy , Child , Child, Preschool , Cyclophosphamide , Etoposide , Female , Humans , Infant , Isotretinoin/therapeutic use , Male , Medulloblastoma/pathology , Neoplasms, Germ Cell and Embryonal/drug therapy , Neuroectodermal Tumors, Primitive/drug therapy , Prospective Studies , Vincristine , Vorinostat
9.
Cancer Cell ; 39(11): 1519-1530.e4, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34678152

ABSTRACT

Nearly one-third of children with medulloblastoma, a malignant embryonal tumor of the cerebellum, succumb to their disease. Conventional response monitoring by imaging and cerebrospinal fluid (CSF) cytology remains challenging, and a marker for measurable residual disease (MRD) is lacking. Here, we show the clinical utility of CSF-derived cell-free DNA (cfDNA) as a biomarker of MRD in serial samples collected from children with medulloblastoma (123 patients, 476 samples) enrolled on a prospective trial. Using low-coverage whole-genome sequencing, tumor-associated copy-number variations in CSF-derived cfDNA are investigated as an MRD surrogate. MRD is detected at baseline in 85% and 54% of patients with metastatic and localized disease, respectively. The number of MRD-positive patients declines with therapy, yet those with persistent MRD have significantly higher risk of progression. Importantly, MRD detection precedes radiographic progression in half who relapse. Our findings advocate for the prospective assessment of CSF-derived liquid biopsies in future trials for medulloblastoma.


Subject(s)
Cell-Free Nucleic Acids/cerebrospinal fluid , Cerebellar Neoplasms/diagnosis , Medulloblastoma/diagnosis , Whole Genome Sequencing/methods , Biomarkers, Tumor/cerebrospinal fluid , Biomarkers, Tumor/genetics , Cerebellar Neoplasms/cerebrospinal fluid , Cerebellar Neoplasms/genetics , Child , Chromosomal Instability , DNA Copy Number Variations , Disease Progression , Female , Humans , Liquid Biopsy , Male , Medulloblastoma/cerebrospinal fluid , Medulloblastoma/genetics , Neoplasm, Residual , Prospective Studies
10.
Acta Neuropathol ; 142(5): 859-871, 2021 11.
Article in English | MEDLINE | ID: mdl-34409497

ABSTRACT

Medulloblastoma, a common pediatric malignant central nervous system tumour, represent a small proportion of brain tumours in adults. Previously it has been shown that in adults, Sonic Hedgehog (SHH)-activated tumours predominate, with Wingless-type (WNT) and Group 4 being less common, but molecular risk stratification remains a challenge. We performed an integrated analysis consisting of genome-wide methylation profiling, copy number profiling, somatic nucleotide variants and correlation of clinical variables across a cohort of 191 adult medulloblastoma cases identified through the Medulloblastoma Advanced Genomics International Consortium. We identified 30 WNT, 112 SHH, 6 Group 3, and 41 Group 4 tumours. Patients with SHH tumours were significantly older at diagnosis compared to other subgroups (p < 0.0001). Five-year progression-free survival (PFS) for WNT, SHH, Group 3, and Group 4 tumours was 64.4 (48.0-86.5), 61.9% (51.6-74.2), 80.0% (95% CI 51.6-100.0), and 44.9% (95% CI 28.6-70.7), respectively (p = 0.06). None of the clinical variables (age, sex, metastatic status, extent of resection, chemotherapy, radiotherapy) were associated with subgroup-specific PFS. Survival among patients with SHH tumours was significantly worse for cases with chromosome 3p loss (HR 2.9, 95% CI 1.1-7.6; p = 0.02), chromosome 10q loss (HR 4.6, 95% CI 2.3-9.4; p < 0.0001), chromosome 17p loss (HR 2.3, 95% CI 1.1-4.8; p = 0.02), and PTCH1 mutations (HR 2.6, 95% CI 1.1-6.2; p = 0.04). The prognostic significance of 3p loss and 10q loss persisted in multivariable regression models. For Group 4 tumours, chromosome 8 loss was strongly associated with improved survival, which was validated in a non-overlapping cohort (combined cohort HR 0.2, 95% CI 0.1-0.7; p = 0.007). Unlike in pediatric medulloblastoma, whole chromosome 11 loss in Group 4 and chromosome 14q loss in SHH was not associated with improved survival, where MYCN, GLI2 and MYC amplification were rare. In sum, we report unique subgroup-specific cytogenetic features of adult medulloblastoma, which are distinct from those in younger patients, and correlate with survival disparities. Our findings suggest that clinical trials that incorporate new strategies tailored to high-risk adult medulloblastoma patients are urgently needed.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Adolescent , Adult , Biomarkers, Tumor/genetics , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Cohort Studies , Female , Humans , Male , Medulloblastoma/mortality , Medulloblastoma/pathology , Progression-Free Survival , Risk Factors , Young Adult
11.
JAMA Oncol ; 7(9): 1313-1321, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34292305

ABSTRACT

IMPORTANCE: Brain tumors are the leading cause of disease-related death in children. Medulloblastoma is the most common malignant embryonal brain tumor, and strategies to increase survival are needed. OBJECTIVE: To evaluate therapy intensification with carboplatin as a radiosensitizer and isotretinoin as a proapoptotic agent in children with high-risk medulloblastoma in a randomized clinical trial and, with a correlative biology study, facilitate planned subgroup analysis according to World Health Organization consensus molecular subgroups of medulloblastoma. DESIGN, SETTING, AND PARTICIPANTS: A randomized clinical phase 3 trial was conducted from March 2007 to September 2018. Analysis was completed in September 2020. Patients aged 3 to 21 years with newly diagnosed high-risk medulloblastoma from Children's Oncology Group institutions within the US, Canada, Australia, and New Zealand were included. High-risk features included metastasis, residual disease, or diffuse anaplasia. INTERVENTIONS: Patients were randomized to receive 36-Gy craniospinal radiation therapy and weekly vincristine with or without daily carboplatin followed by 6 cycles of maintenance chemotherapy with cisplatin, cyclophosphamide, and vincristine with or without 12 cycles of isotretinoin during and following maintenance. MAIN OUTCOMES AND MEASURES: The primary clinical trial end point was event-free survival, using the log-rank test to compare arms. The primary biology study end point was molecular subgroup classification by DNA methylation array. RESULTS: Of 294 patients with medulloblastoma, 261 were evaluable after central radiologic and pathologic review; median age, 8.6 years (range, 3.3-21.2); 183 (70%) male; 189 (72%) with metastatic disease; 58 (22%) with diffuse anaplasia; and 14 (5%) with greater than 1.5-cm2 residual disease. For all participants, the 5-year event-free survival was 62.9% (95% CI, 55.6%-70.2%) and overall survival was 73.4% (95% CI, 66.7%-80.1%). Isotretinoin randomization was closed early owing to futility. Five-year event-free survival was 66.4% (95% CI, 56.4%-76.4%) with carboplatin vs 59.2% (95% CI, 48.8%-69.6%) without carboplatin (P = .11), with the effect exclusively observed in group 3 subgroup patients: 73.2% (95% CI, 56.9%-89.5%) with carboplatin vs 53.7% (95% CI, 35.3%-72.1%) without (P = .047). Five-year overall survival differed by molecular subgroup (P = .006): WNT pathway activated, 100% (95% CI, 100%-100%); SHH pathway activated, 53.6% (95% CI, 33.0%-74.2%); group 3, 73.7% (95% CI, 61.9%-85.5%); and group 4, 76.9% (95% CI, 67.3%-86.5%). CONCLUSIONS AND RELEVANCE: In this randomized clinical trial, therapy intensification with carboplatin improved event-free survival by 19% at 5 years for children with high-risk group 3 medulloblastoma. These findings further support the value of an integrated clinical and molecular risk stratification for medulloblastoma. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT00392327.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/adverse effects , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Disease-Free Survival , Humans , Isotretinoin/adverse effects , Male , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Young Adult
12.
J Clin Oncol ; 39(24): 2685-2697, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34110925

ABSTRACT

PURPOSE: Children with average-risk medulloblastoma (MB) experience survival rates of ≥ 80% at the expense of adverse consequences of treatment. Efforts to mitigate these effects include deintensification of craniospinal irradiation (CSI) dose and volume. METHODS: ACNS0331 (ClinicalTrials.gov identifier: NCT00085735) randomly assigned patients age 3-21 years with average-risk MB to receive posterior fossa radiation therapy (PFRT) or involved field radiation therapy (IFRT) following CSI. Young children (3-7 years) were also randomly assigned to receive standard-dose CSI (SDCSI; 23.4 Gy) or low-dose CSI (LDCSI; 18 Gy). Post hoc molecular classification and mutational analysis contextualized outcomes according to known biologic subgroups (Wingless, Sonic Hedgehog, group 3, and group 4) and genetic biomarkers. Neurocognitive changes and ototoxicity were monitored over time. RESULTS: Five hundred forty-nine patients were enrolled on study, of which 464 were eligible and evaluable to compare PFRT versus IFRT and 226 for SDCSI versus LDCSI. The five-year event-free survival (EFS) was 82.5% (95% CI, 77.2 to 87.8) and 80.5% (95% CI, 75.2 to 85.8) for the IFRT and PFRT regimens, respectively, and 71.4% (95% CI, 62.8 to 80) and 82.9% (95% CI, 75.6 to 90.2) for the LDCSI and SDCSI regimens, respectively. IFRT was not inferior to PFRT (hazard ratio, 0.97; 94% upper CI, 1.32). LDCSI was inferior to SDCSI (hazard ratio, 1.67%; 80% upper CI, 2.10). Improved EFS was observed in patients with Sonic Hedgehog MB who were randomly assigned to the IFRT arm (P = .018). Patients with group 4 MB receiving LDCSI exhibited inferior EFS (P = .047). Children receiving SDCSI exhibited greater late declines in IQ (estimate = 5.87; P = .021). CONCLUSION: Reducing the radiation boost volume in average-risk MB is safe and does not compromise survival. Reducing CSI dose in young children with average-risk MB results in inferior outcomes, possibly in a subgroup-dependent manner, but is associated with better neurocognitive outcome. Molecularly informed patient selection warrants further exploration for children with MB to be considered for late-effect sparing approaches.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Medulloblastoma/drug therapy , Medulloblastoma/radiotherapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Child , Child, Preschool , Female , Humans , Male , Young Adult
13.
Behav Neurosci ; 135(2): 89-107, 2021 Apr.
Article in English | MEDLINE | ID: mdl-34060867

ABSTRACT

Technological advances over the last decade are changing the face of behavioral neuroscience research. Here we review recent work on the use of one such transformative tool in behavioral neuroscience research, chemogenetics (or Designer Receptors Exclusively Activated by Designer Drugs, DREADDS). As transformative technologies such as DREADDs are introduced, applied, and refined, their utility in addressing complex questions about behavior and cognition becomes clear and exciting. In the behavioral neuroscience field, remarkable new findings now regularly appear as a result of the ability to monitor and intervene in neural processes with high anatomical precision as animals behave in complex task environments. As these new tools are applied to behavioral questions, individualized procedures for their use find their way into diverse labs. Thus, "tips of the trade" become important for wide dissemination not only for laboratories that are using the tools but also for those who are interested in incorporating them into their own work. Our aim is to provide an up-to-date perspective on how the DREADD technique is being used for research on learning and memory, decision making, and goal-directed behavior, as well as to provide suggestions and considerations for current and future users based on our collective experience. (PsycInfo Database Record (c) 2021 APA, all rights reserved).


Subject(s)
Designer Drugs , Neurosciences , Animals , Learning
14.
J Clin Oncol ; 39(7): 807-821, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33502920

ABSTRACT

PURPOSE: We sought to investigate clinical outcomes of relapsed medulloblastoma and to compare molecular features between patient-matched diagnostic and relapsed tumors. METHODS: Children and infants enrolled on either SJMB03 (NCT00085202) or SJYC07 (NCT00602667) trials who experienced medulloblastoma relapse were analyzed for clinical outcomes, including anatomic and temporal patterns of relapse and postrelapse survival. A largely independent, paired molecular cohort was analyzed by DNA methylation array and next-generation sequencing. RESULTS: A total of 72 of 329 (22%) SJMB03 and 52 of 79 (66%) SJYC07 patients experienced relapse with significant representation of Group 3 and wingless tumors. Although most patients exhibited some distal disease (79%), 38% of patients with sonic hedgehog tumors experienced isolated local relapse. Time to relapse and postrelapse survival varied by molecular subgroup with longer latencies for patients with Group 4 tumors. Postrelapse radiation therapy among previously nonirradiated SJYC07 patients was associated with long-term survival. Reirradiation was only temporizing for SJMB03 patients. Among 127 patients with patient-matched tumor pairs, 9 (7%) experienced subsequent nonmedulloblastoma CNS malignancies. Subgroup (96%) and subtype (80%) stabilities were largely maintained among the remainder. Rare subgroup divergence was observed from Group 4 to Group 3 tumors, which is coincident with genetic alterations involving MYC, MYCN, and FBXW7. Subgroup-specific patterns of alteration were identified for driver genes and chromosome arms. CONCLUSION: Clinical behavior of relapsed medulloblastoma must be contextualized in terms of up-front therapies and molecular classifications. Group 4 tumors exhibit slower biological progression. Utility of radiation at relapse is dependent on patient age and prior treatments. Degree and patterns of molecular conservation at relapse vary by subgroup. Relapse tissue enables verification of molecular targets and identification of occult secondary malignancies.


Subject(s)
Biomarkers, Tumor/genetics , Cerebellar Neoplasms/genetics , DNA Methylation , Medulloblastoma/genetics , Neoplasm Recurrence, Local , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/therapy , Child , Child, Preschool , Clinical Trials as Topic , Disease Progression , Epigenome , Epigenomics , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Medulloblastoma/mortality , Medulloblastoma/secondary , Medulloblastoma/therapy , Retreatment , Time Factors , Treatment Outcome
15.
Eur J Neurosci ; 53(8): 2567-2579, 2021 04.
Article in English | MEDLINE | ID: mdl-33462844

ABSTRACT

When pursuing desirable outcomes, one must make the decision between exploring possible actions to obtain those outcomes and exploiting known strategies to maximize efficiency. The dorsolateral striatum (DLS) has been extensively studied with respect to how actions can develop into habits and has also been implicated as an area involved in governing exploitative behavior. Surprisingly, prior work has shown that DLS cholinergic interneurons (ChIs) are not involved in the canonical habit formation function ascribed to the DLS but are instead modulators of behavioral flexibility after initial learning. To further probe this, we evaluated the role of DLS ChIs in behavioral exploration during a brief instrumental training experiment. Through designer receptors exclusively activated by designer drugs (DREADDs) in ChAT-Cre rats, ChIs in the DLS were inhibited during specific phases of the experiment: instrumental training, free-reward delivery, at both times, or never. Without ChI activity during instrumental training, animals biased their responding toward an "optimal" strategy while continuing to work efficiently. This effect was observed again when contingencies were removed as animals with ChIs offline during that phase, regardless of ChI inhibition previously, decreased responding more than animals with ChIs intact. These findings build upon a growing body of literature implicating ChIs in the striatum as gate-keepers of behavioral flexibility and exploration.


Subject(s)
Corpus Striatum , Neostriatum , Animals , Cholinergic Agents , Habits , Interneurons , Rats
16.
J Clin Oncol ; 39(7): 822-835, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33405951

ABSTRACT

PURPOSE: SJMB03 (ClinicalTrials.gov identifier: NCT00085202) was a phase III risk-adapted trial that aimed to determine the frequency and clinical significance of biological variants and genetic alterations in medulloblastoma. PATIENTS AND METHODS: Patients 3-21 years old were stratified into average-risk and high-risk treatment groups based on metastatic status and extent of resection. Medulloblastomas were molecularly classified into subgroups (Wingless [WNT], Sonic Hedgehog [SHH], group 3, and group 4) and subtypes based on DNA methylation profiles and overlaid with gene mutations from next-generation sequencing. Coprimary study end points were (1) to assess the relationship between ERBB2 protein expression in tumors and progression-free survival (PFS), and (2) to estimate the frequency of mutations associated with WNT and SHH tumors. Clinical and molecular risk factors were evaluated, and the most robust were used to model new risk-classification categories. RESULTS: Three hundred thirty eligible patients with medulloblastoma were enrolled. Five-year PFS was 83.2% (95% CI, 78.4 to 88.2) for average-risk patients (n = 227) and 58.7% (95% CI, 49.8 to 69.1) for high-risk patients (n = 103). No association was found between ERBB2 status and PFS in the overall cohort (P = .74) or when patients were stratified by clinical risk (P = .71). Mutations in CTNNB1 (96%), DDX3X (37%), and SMARCA4 (24%) were most common in WNT tumors and PTCH1 (38%), TP53 (21%), and DDX3X (19%) in SHH tumors. Methylome profiling classified 53 WNT (17.4%), 48 SHH (15.7%), 65 group 3 (21.3%), and 139 group 4 (45.6%) tumors. A comprehensive clinicomolecular risk factor analysis identified three low-risk groups (WNT, low-risk SHH, and low-risk combined groups 3 and 4) with excellent (5-year PFS > 90%) and two very high-risk groups (high-risk SHH and high-risk combined groups 3 and 4) with poor survival (5-year PFS < 60%). CONCLUSION: These results establish a new risk stratification for future medulloblastoma trials.


Subject(s)
Biomarkers, Tumor/genetics , Cerebellar Neoplasms/therapy , DNA Methylation , Medulloblastoma/therapy , Mutation , Adolescent , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Child , Child, Preschool , DNA Mutational Analysis , Epigenome , Epigenomics , Female , High-Throughput Nucleotide Sequencing , Humans , Magnetic Resonance Imaging , Male , Medulloblastoma/genetics , Medulloblastoma/mortality , Medulloblastoma/secondary , Predictive Value of Tests , Progression-Free Survival , Risk Assessment , Risk Factors , Time Factors , Young Adult
17.
Trends Genet ; 37(3): 235-250, 2021 03.
Article in English | MEDLINE | ID: mdl-33272592

ABSTRACT

Medulloblastoma (MB) is a highly malignant cerebellar tumor predominantly diagnosed during childhood. Driven by pathogenic activation of sonic hedgehog (SHH) signaling, SHH subgroup MB (SHH-MB) accounts for nearly one-third of diagnoses. Extensive molecular analyses have identified biologically and clinically relevant intertumoral heterogeneity among SHH-MB tumors, prompting the recognition of novel subtypes. Beyond germline and somatic mutations promoting constitutive SHH signaling, driver alterations affect a multitude of pathways and molecular processes, including TP53 signaling, chromatin modulation, and post-transcriptional gene regulation. Here, we review recent advances in the underpinnings of SHH-MB in the context of molecular subtypes, clarify novel somatic and germline drivers, highlight cellular origins and developmental hierarchies, and describe the composition of the tumor microenvironment and its putative role in tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Chromatin/genetics , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Genetic Heterogeneity , Germ-Line Mutation/genetics , Humans , Medulloblastoma/pathology , Signal Transduction/genetics , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics
18.
Genes Dev ; 34(17-18): 1161-1176, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32820036

ABSTRACT

Medulloblastoma is a malignant childhood brain tumor arising from the developing cerebellum. In Sonic Hedgehog (SHH) subgroup medulloblastoma, aberrant activation of SHH signaling causes increased proliferation of granule neuron progenitors (GNPs), and predisposes these cells to tumorigenesis. A second, cooperating genetic hit is often required to push these hyperplastic cells to malignancy and confer mutation-specific characteristics associated with oncogenic signaling. Somatic loss-of-function mutations of the transcriptional corepressor BCOR are recurrent and enriched in SHH medulloblastoma. To investigate BCOR as a putative tumor suppressor, we used a genetically engineered mouse model to delete exons 9/10 of Bcor (BcorΔE9-10 ) in GNPs during development. This mutation leads to reduced expression of C-terminally truncated BCOR (BCORΔE9-10). While BcorΔE9-10 alone did not promote tumorigenesis or affect GNP differentiation, BcorΔE9-10 combined with loss of the SHH receptor gene Ptch1 resulted in fully penetrant medulloblastomas. In Ptch1+/- ;BcorΔE9-10 tumors, the growth factor gene Igf2 was aberrantly up-regulated, and ectopic Igf2 overexpression was sufficient to drive tumorigenesis in Ptch1+/- GNPs. BCOR directly regulates Igf2, likely through the PRC1.1 complex; the repressive histone mark H2AK119Ub is decreased at the Igf2 promoter in Ptch1+/- ;BcorΔE9-10 tumors. Overall, our data suggests that BCOR-PRC1.1 disruption leads to Igf2 overexpression, which transforms preneoplastic cells to malignant tumors.


Subject(s)
Cerebellar Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Hedgehog Proteins/metabolism , Medulloblastoma/genetics , Polycomb-Group Proteins/metabolism , Repressor Proteins/genetics , Animals , Carcinogenesis/genetics , Disease Models, Animal , Hedgehog Proteins/genetics , Humans , Mice , Mutation , Patched-1 Receptor/genetics , Polycomb-Group Proteins/genetics , Repressor Proteins/metabolism , Sequence Deletion
19.
Acta Neuropathol ; 140(2): 209-225, 2020 08.
Article in English | MEDLINE | ID: mdl-32519082

ABSTRACT

Pediatric brain tumors are the leading cause of cancer-related death in children. Patient-derived orthotopic xenografts (PDOX) of childhood brain tumors have recently emerged as a biologically faithful vehicle for testing novel and more effective therapies. Herein, we provide the histopathological and molecular analysis of 37 novel PDOX models generated from pediatric brain tumor patients treated at St. Jude Children's Research Hospital. Using a combination of histopathology, whole-genome and whole-exome sequencing, RNA-sequencing, and DNA methylation arrays, we demonstrate the overall fidelity and inter-tumoral molecular heterogeneity of pediatric brain tumor PDOX models. These models represent frequent as well as rare childhood brain tumor entities, including medulloblastoma, ependymoma, atypical teratoid rhabdoid tumor, and embryonal tumor with multi-layer rosettes. PDOX models will be valuable platforms for evaluating novel therapies and conducting pre-clinical trials to accelerate progress in the treatment of brain tumors in children. All described PDOX models and associated datasets can be explored using an interactive web-based portal and will be made freely available to the research community upon request.


Subject(s)
Brain Neoplasms , Disease Models, Animal , Heterografts , Animals , Child , Humans , Mice
20.
Dev Cell ; 54(4): 455-470.e5, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32553121

ABSTRACT

DEAD-Box Helicase 3 X-Linked (DDX3X) is frequently mutated in the Wingless (WNT) and Sonic hedghog (SHH) subtypes of medulloblastoma-the commonest malignant childhood brain tumor, but whether DDX3X functions as a medulloblastoma oncogene or tumor suppressor gene is not known. Here, we show that Ddx3x regulates hindbrain patterning and development by controlling Hox gene expression and cell stress signaling. In mice predisposed to Wnt- or Shh medulloblastoma, Ddx3x sensed oncogenic stress and suppressed tumor formation. WNT and SHH medulloblastomas normally arise only in the lower and upper rhombic lips, respectively. Deletion of Ddx3x removed this lineage restriction, enabling both medulloblastoma subtypes to arise in either germinal zone. Thus, DDX3X is a medulloblastoma tumor suppressor that regulates hindbrain development and restricts the competence of cell lineages to form medulloblastoma subtypes.


Subject(s)
Brain Neoplasms/genetics , DEAD-box RNA Helicases/genetics , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Animals , Brain Neoplasms/pathology , Cell Lineage/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Homeobox , Humans , Medulloblastoma/pathology , Mice , Mutation/genetics , Rhombencephalon/metabolism , Rhombencephalon/pathology , Wnt Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...