Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Immunol Immunother ; 70(3): 721-732, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32915319

ABSTRACT

Faithful tumor mouse models are fundamental research tools to advance the field of immuno-oncology (IO). This is particularly relevant in diseases with low incidence, as in the case of pediatric malignancies, that rely on pre-clinical therapeutic development. However, conventional syngeneic and genetically engineered mouse models fail to recapitulate the tumor heterogeneity and microenvironmental complexity of human pathology that are essential determinants of cancer-directed immunity. Here, we characterize a novel mouse model that supports human natural killer (NK) cell development and engraftment of neuroblastoma orthotopic patient-derived xenograft (O-PDX) for pre-clinical antibody and cytokine testing. Using cytotoxicity assays, single-cell RNA-sequencing, and multi-color flow cytometry, we demonstrate that NK cells that develop in the humanized mice are fully licensed to execute NK cell cytotoxicity, permit human tumor engraftment, but can be therapeutically redirected to induce antibody-dependent cell-mediated cytotoxicity (ADCC). Although these cells share phenotypic and molecular features with healthy controls, we noted that they lacked an NK cell subset, termed activated NK cells, that is characterized by differentially expressed genes that are induced by cytokine activation. Because this subset of genes is also downregulated in patients with neuroblastoma compared to healthy controls, we hypothesize that this finding could be due to tumor-mediated suppressive effects. Thus, despite its technical complexity, this humanized patient-derived xenograft mouse model could serve as a faithful system for future testing of IO applications and studies of underlying immunologic processes.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Neuroblastoma/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Bone Marrow Transplantation , Case-Control Studies , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Treatment Outcome , Xenograft Model Antitumor Assays
2.
Cancer Cell ; 34(3): 411-426.e19, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30146332

ABSTRACT

Personalized cancer therapy targeting somatic mutations in patient tumors is increasingly being incorporated into practice. Other therapeutic vulnerabilities resulting from changes in gene expression due to tumor specific epigenetic perturbations are progressively being recognized. These genomic and epigenomic changes are ultimately manifest in the tumor proteome and phosphoproteome. We integrated transcriptomic, epigenomic, and proteomic/phosphoproteomic data to elucidate the cellular origins and therapeutic vulnerabilities of rhabdomyosarcoma (RMS). We discovered that alveolar RMS occurs further along the developmental program than embryonal RMS. We also identified deregulation of the RAS/MEK/ERK/CDK4/6, G2/M, and unfolded protein response pathways through our integrated analysis. Comprehensive preclinical testing revealed that targeting the WEE1 kinase in the G2/M pathway is the most effective approach in vivo for high-risk RMS.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Muscle Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Rhabdomyosarcoma/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Child , Epigenomics , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genomics , Humans , Male , Mice , Molecular Targeted Therapy/methods , Muscle Neoplasms/genetics , Muscle Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Precision Medicine/methods , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proteomics , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Unfolded Protein Response/genetics , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 24(7): 1654-1666, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29301833

ABSTRACT

Purpose: Curing all children with brain tumors will require an understanding of how each subtype responds to conventional treatments and how best to combine existing and novel therapies. It is extremely challenging to acquire this knowledge in the clinic alone, especially among patients with rare tumors. Therefore, we developed a preclinical brain tumor platform to test combinations of conventional and novel therapies in a manner that closely recapitulates clinic trials.Experimental Design: A multidisciplinary team was established to design and conduct neurosurgical, fractionated radiotherapy and chemotherapy studies, alone or in combination, in accurate mouse models of supratentorial ependymoma (SEP) subtypes and choroid plexus carcinoma (CPC). Extensive drug repurposing screens, pharmacokinetic, pharmacodynamic, and efficacy studies were used to triage active compounds for combination preclinical trials with "standard-of-care" surgery and radiotherapy.Results: Mouse models displayed distinct patterns of response to surgery, irradiation, and chemotherapy that varied with tumor subtype. Repurposing screens identified 3-hour infusions of gemcitabine as a relatively nontoxic and efficacious treatment of SEP and CPC. Combination neurosurgery, fractionated irradiation, and gemcitabine proved significantly more effective than surgery and irradiation alone, curing one half of all animals with aggressive forms of SEP.Conclusions: We report a comprehensive preclinical trial platform to assess the therapeutic activity of conventional and novel treatments among rare brain tumor subtypes. It also enables the development of complex, combination treatment regimens that should deliver optimal trial designs for clinical testing. Postirradiation gemcitabine infusion should be tested as new treatments of SEP and CPC. Clin Cancer Res; 24(7); 1654-66. ©2018 AACR.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Evaluation, Preclinical/methods , Humans , Mice , Mice, Nude , Treatment Outcome , Gemcitabine
4.
Nature ; 549(7670): 96-100, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28854174

ABSTRACT

Paediatric solid tumours arise from endodermal, ectodermal, or mesodermal lineages. Although the overall survival of children with solid tumours is 75%, that of children with recurrent disease is below 30%. To capture the complexity and diversity of paediatric solid tumours and establish new models of recurrent disease, here we develop a protocol to produce orthotopic patient-derived xenografts at diagnosis, recurrence, and autopsy. Tumour specimens were received from 168 patients, and 67 orthotopic patient-derived xenografts were established for 12 types of cancer. The origins of the patient-derived xenograft tumours were reflected in their gene-expression profiles and epigenomes. Genomic profiling of the tumours, including detailed clonal analysis, was performed to determine whether the clonal population in the xenograft recapitulated the patient's tumour. We identified several drug vulnerabilities and showed that the combination of a WEE1 inhibitor (AZD1775), irinotecan, and vincristine can lead to complete response in multiple rhabdomyosarcoma orthotopic patient-derived xenografts tumours in vivo.


Subject(s)
Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Bortezomib/pharmacology , Bortezomib/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Child , Clone Cells , Drug Therapy, Combination , Epigenesis, Genetic , Female , Heterografts/drug effects , Heterografts/metabolism , Heterografts/pathology , Heterografts/transplantation , High-Throughput Screening Assays/methods , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Irinotecan , Mice , Neoplasms/genetics , Nuclear Proteins/antagonists & inhibitors , Panobinostat , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrimidinones , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Vincristine/pharmacology , Vincristine/therapeutic use
5.
Nat Genet ; 47(8): 878-87, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26075792

ABSTRACT

Cancers are characterized by non-random chromosome copy number alterations that presumably contain oncogenes and tumor-suppressor genes (TSGs). The affected loci are often large, making it difficult to pinpoint which genes are driving the cancer. Here we report a cross-species in vivo screen of 84 candidate oncogenes and 39 candidate TSGs, located within 28 recurrent chromosomal alterations in ependymoma. Through a series of mouse models, we validate eight new ependymoma oncogenes and ten new ependymoma TSGs that converge on a small number of cell functions, including vesicle trafficking, DNA modification and cholesterol biosynthesis, identifying these as potential new therapeutic targets.


Subject(s)
Ependymoma/genetics , Genes, Tumor Suppressor , Genetic Predisposition to Disease/genetics , Oncogenes/genetics , Animals , Cells, Cultured , Chromosome Aberrations , DNA Copy Number Variations , Ependymoma/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Kaplan-Meier Estimate , Male , Mice, Nude , Mice, Transgenic , Microscopy, Confocal , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transfection
6.
Cancer Chemother Pharmacol ; 75(5): 897-906, 2015 May.
Article in English | MEDLINE | ID: mdl-25724157

ABSTRACT

Clofarabine, a deoxyadenosine analog, was an active anticancer drug in our in vitro high-throughput screening against mouse ependymoma neurospheres. To characterize the clofarabine disposition in mice for further preclinical efficacy studies, we evaluated the plasma and central nervous system disposition in a mouse model of ependymoma. A plasma pharmacokinetic study of clofarabine (45 mg/kg, IP) was performed in CD1 nude mice bearing ependymoma to obtain initial plasma pharmacokinetic parameters. These estimates were used to derive D-optimal plasma sampling time points for cerebral microdialysis studies. A simulation of clofarabine pharmacokinetics in mice and pediatric patients suggested that a dosage of 30 mg/kg IP in mice would give exposures comparable to that in children at a dosage of 148 mg/m(2). Cerebral microdialysis was performed to study the tumor extracellular fluid (ECF) disposition of clofarabine (30 mg/kg, IP) in the ependymoma cortical allografts. Plasma and tumor ECF concentration-time data were analyzed using a nonlinear mixed effects modeling approach. The median unbound fraction of clofarabine in mouse plasma was 0.79. The unbound tumor to plasma partition coefficient (K pt,uu: ratio of tumor to plasma AUCu,0-inf) of clofarabine was 0.12 ± 0.05. The model-predicted mean tumor ECF clofarabine concentrations were below the in vitro 1-h IC50 (407 ng/mL) for ependymoma neurospheres. Thus, our results show the clofarabine exposure reached in the tumor ECF was below that associated with an antitumor effect in our in vitro washout study. Therefore, clofarabine was de-prioritized as an agent to treat ependymoma, and further preclinical studies were not pursued.


Subject(s)
Adenine Nucleotides/pharmacology , Adenine Nucleotides/pharmacokinetics , Arabinonucleosides/pharmacology , Arabinonucleosides/pharmacokinetics , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Ependymoma/drug therapy , Ependymoma/metabolism , Adenine Nucleotides/blood , Adolescent , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/pharmacology , Arabinonucleosides/blood , Blood Proteins/metabolism , Brain/metabolism , Brain Neoplasms/blood , Child , Child, Preschool , Clofarabine , Ependymoma/blood , Female , Humans , Mice , Mice, Nude , Models, Biological , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Neurobiol Dis ; 59: 26-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23859799

ABSTRACT

The p38 mitogen-activated protein kinase (MAPK) isoforms are phosphorylated by a variety of stress stimuli in neurodegenerative disease and act as upstream activators of myriad pathogenic processes. Thus, p38 MAPK inhibitors are of growing interest as possible therapeutic interventions. Axonal dysfunction is an early component of most neurodegenerative disorders, including the most prevalent optic neuropathy, glaucoma. Sensitivity to intraocular pressure at an early stage disrupts anterograde transport along retinal ganglion cell (RGC) axons to projection targets in the brain with subsequent degeneration of the axons themselves; RGC body loss is much later. Here we show that elevated ocular pressure in rats increases p38 MAPK activation in retina, especially in RGC bodies. Topical eye-drop application of a potent and selective inhibitor of the p38 MAPK catalytic domain (Ro3206145) prevented both the degradation of anterograde transport to the brain and degeneration of axons in the optic nerve. Ro3206145 reduced in the retina phosphorylation of tau and heat-shock protein 27, both down-stream targets of p38 MAPK activation implicated in glaucoma, as well as expression of two inflammatory responses. We also observed increased p38 MAPK activation in mouse models. Thus, inhibition of p38 MAPK signaling in the retina may represent a therapeutic target for preventing early pathogenesis in optic neuropathies.


Subject(s)
Axons/pathology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , Retinal Degeneration/prevention & control , Signal Transduction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , Activating Transcription Factor 2/metabolism , Animals , Chaperonin 60/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Imidazoles/therapeutic use , In Vitro Techniques , Intraocular Pressure/physiology , Intraocular Pressure/radiation effects , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Ocular Hypertension/complications , Ocular Hypertension/drug therapy , Ocular Hypertension/etiology , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Retina/drug effects , Retina/metabolism , Retina/radiation effects , Retinal Degeneration/etiology , Retinal Degeneration/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Signal Transduction/drug effects , Time Factors
8.
Mamm Genome ; 16(9): 672-82, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16245024

ABSTRACT

The mutant allelic series of the mouse quaking gene consists of the spontaneous quaking(viable) (qk(v)) allele, which is homozygous viable with a dysmyelination phenotype, and four ENU-induced alleles (qk(kt 1), qk(k2), qk(kt3/4), and qk(l-1)), which are homozygous embryonic lethal. Here we report the isolation of qk(e5), the first ENU-induced viable allele of quaking. Unlike qk(v)/qk(v), qk(e5)/qk(e5) animals have early-onset seizures, severe ataxia, and a dramatically reduced lifespan. Ultrastructural analysis of qk(e5)/qk(e5) brains reveals severe dysmyelination when compared with both wild-type and qk(v)/qk(v) brains. In addition, Calbindin detection in young adult qk(e5)/qk(e5) mice reveals Purkinje cell axonal swellings indicative of neurodegeneration , which is not seen in young adult qk(v)/qk(v) mice. Although the molecular defect in the qk(e5) allele is not evident by sequencing, protein expression studies show that qk(e5)/qk(e5) postnatal oligodendrocytes lack the QKI-6 and QKI-7 isoforms and have reduced QKI-5 levels. The oligodendrocyte developmental markers PDGF alpha R, NG 2, O4, CNP, and MBP are also present in the qk(e5)/qk(e5) postnatal brain although CNP and MBP levels are considerably reduced. Because the qk(v) allele is a large deletion that affects the expression of three genes, the new neurologic qk(e5) allele is an important addition to this allelic series.


Subject(s)
Alleles , Central Nervous System/ultrastructure , Demyelinating Diseases/genetics , Phenotype , RNA-Binding Proteins/genetics , Animals , Blotting, Northern , Calbindins , Crosses, Genetic , DNA Primers , Demyelinating Diseases/pathology , Ethylnitrosourea , Immunohistochemistry , Mice , Mice, Quaking , Microscopy, Electron, Transmission , Mutagenesis , S100 Calcium Binding Protein G , Sequence Analysis, DNA
9.
Mov Disord ; 20(10): 1369-74, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16001410

ABSTRACT

The quakingviable (qkv) mutant mouse shows a recessive neurological phenotype that includes central nervous system (CNS) dysmyelination, seizures, and tremor associated with voluntary movement. The molecular defect of qkv has been previously reported to be a spontaneous approximately 1 megabase (Mb) deletion in the proximal region of mouse chromosome 17 that occurred in the DBA mouse strain more than four decades ago. The mutation has recently been shown to affect three genes in the region: Quaking (qk), Parkin-coregulated gene (Pacrg), and Parkin. Here we determine the exact deletion breakpoints and demonstrate that the mutation is not just comprised of a approximately 1.1 Mb deletion, but also harbors a small 163 bp duplication fragment between the deletion breakpoints. Although the distal deletion breakpoint is within the fifth intron of the mouse Parkin gene, the duplicated sequence is derived from the sixth Parkin intron and shows positive transcriptional activity on a reporter gene in vitro. This complexity provides insight into a well-studied neurological mutant and may have a role in affecting the phenotype observed.


Subject(s)
Chromosomes, Mammalian/genetics , Gene Duplication , Inteins/genetics , Point Mutation/genetics , Ubiquitin-Protein Ligases/genetics , Animals , DNA Fragmentation/genetics , DNA Primers/genetics , Demyelinating Diseases/genetics , Gene Expression Regulation/genetics , Genes, Reporter/genetics , Humans , Mice , Mice, Quaking , Microfilament Proteins , Molecular Chaperones , Phenotype , Proteins/genetics , Transcriptional Activation/genetics
10.
Am J Hum Genet ; 72(5): 1101-16, 2003 May.
Article in English | MEDLINE | ID: mdl-12649807

ABSTRACT

To investigate the potential involvement of genome architecture in nonrecurrent chromosome rearrangements, we analyzed the breakpoints of eight translocations and 18 unusual-sized deletions involving human proximal 17p. Surprisingly, we found that many deletion breakpoints occurred in low-copy repeats (LCRs); 13 were associated with novel large LCR17p structures, and 2 mapped within an LCR sequence (middle SMS-REP) within the Smith-Magenis syndrome (SMS) common deletion. Three translocation breakpoints involving 17p11 were found to be located within the centromeric alpha-satellite sequence D17Z1, three within a pericentromeric segment, and one at the distal SMS-REP. Remarkably, our analysis reveals that LCRs constitute >23% of the analyzed genome sequence in proximal 17p--an experimental observation two- to fourfold higher than predictions based on virtual analysis of the genome. Our data demonstrate that higher-order genomic architecture involving LCRs plays a significant role not only in recurrent chromosome rearrangements but also in translocations and unusual-sized deletions involving 17p.


Subject(s)
Chromosome Breakage/genetics , Chromosomes, Human, Pair 17/genetics , Genome , Sequence Deletion/genetics , Translocation, Genetic/genetics , Centromere/genetics , Humans , In Situ Hybridization, Fluorescence/methods , Physical Chromosome Mapping , Repetitive Sequences, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...