Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Commun Biol ; 4(1): 497, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888863

ABSTRACT

Cyclic dinucleotide (CDN) agonists of the STimulator of InterferoN Genes (STING) pathway have shown immune activation and tumor clearance in pre-clinical models. However, CDNs administered intratumorally also promote STING activation leading to direct cytotoxicity of many cell types in the tumor microenvironment (TME), systemic inflammation due to rapid tumor extravasation of the CDN, and immune ablation in the TME. These result in a failure to establish immunological memory. ExoSTING, an engineered extracellular vesicle (EV) exogenously loaded with CDN, enhances the potency of CDN and preferentially activates antigen presenting cells in the TME. Following intratumoral injection, exoSTING was retained within the tumor, enhanced local Th1 responses and recruitment of CD8+ T cells, and generated systemic anti-tumor immunity to the tumor. ExoSTING at therapeutically active doses did not induce systemic inflammatory cytokines, resulting in an enhanced therapeutic window. ExoSTING is a novel, differentiated therapeutic candidate that leverages the natural biology of EVs to enhance the activity of CDNs.


Subject(s)
Extracellular Vesicles/physiology , Immunologic Surveillance , Tumor Microenvironment/physiology , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
Clin Cancer Res ; 26(15): 3958-3968, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32299817

ABSTRACT

PURPOSE: A persistent issue in cancer drug development is the discordance between robust antitumor drug activity observed in laboratory models and the limited benefit frequently observed when patients are treated with the same agents in clinical trials. Difficulties in accurately modeling the complexities of human tumors may underlie this problem. To address this issue, we developed Comparative In Vivo Oncology (CIVO), which enables in situ investigation of multiple microdosed drugs simultaneously in a patient's tumor. This study was designed to test CIVO's safety and feasibility in patients with soft tissue sarcoma (STS). PATIENTS AND METHODS: We conducted a single arm, prospective, 13-patient pilot study. Patients scheduled for incisional biopsy or tumor resection were CIVO-injected 1 to 3 days prior to surgery. Saline or microdoses of anticancer agents were percutaneously injected into the tumor in a columnar fashion through each of eight needles. Following excision, drug responses were evaluated in the injected tissue. RESULTS: The primary objective was met, establishing CIVO's feasibility and safety. Device-related adverse events were limited to transient grade 1 nonserious events. In addition, biomarker evaluation of localized tumor response to CIVO microinjected drugs by IHC or with NanoString GeoMx Digital Spatial Profiler demonstrated consistency with known mechanisms of action of each drug, impact on the tumor microenvironment, and historic clinical activity. CONCLUSIONS: These results are an advance toward use of CIVO as a translational research tool for early evaluation of investigational agents and drug combinations in a novel approach to phase 0 trials.See related commentary by Sleijfer and Lolkema, p. 3897.


Subject(s)
Antineoplastic Agents , Sarcoma , Antineoplastic Agents/adverse effects , Humans , Pilot Projects , Prospective Studies , Sarcoma/drug therapy , Tumor Microenvironment
3.
Nat Med ; 24(11): 1752-1761, 2018 11.
Article in English | MEDLINE | ID: mdl-30349086

ABSTRACT

Brain tumors are the leading cause of cancer-related death in children. Genomic studies have provided insights into molecular subgroups and oncogenic drivers of pediatric brain tumors that may lead to novel therapeutic strategies. To evaluate new treatments, better preclinical models adequately reflecting the biological heterogeneity are needed. Through the Children's Oncology Group ACNS02B3 study, we have generated and comprehensively characterized 30 patient-derived orthotopic xenograft models and seven cell lines representing 14 molecular subgroups of pediatric brain tumors. Patient-derived orthotopic xenograft models were found to be representative of the human tumors they were derived from in terms of histology, immunohistochemistry, gene expression, DNA methylation, copy number, and mutational profiles. In vivo drug sensitivity of targeted therapeutics was associated with distinct molecular tumor subgroups and specific genetic alterations. These models and their molecular characterization provide an unprecedented resource for the cancer community to study key oncogenic drivers and to evaluate novel treatment strategies.


Subject(s)
Biological Specimen Banks , Brain Neoplasms/pathology , Immunohistochemistry , Xenograft Model Antitumor Assays/methods , Animals , Cell Line, Tumor , Child , Child, Preschool , DNA Methylation/genetics , Female , Genomics , Humans , Male , Mice , Mutation , Pediatrics
4.
Sci Rep ; 7(1): 18007, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269870

ABSTRACT

Aberrant regulation of BCL-2 family members enables evasion of apoptosis and tumor resistance to chemotherapy. BCL-2 and functionally redundant counterpart, MCL-1, are frequently over-expressed in high-risk diffuse large B-cell lymphoma (DLBCL). While clinical inhibition of BCL-2 has been achieved with the BH3 mimetic venetoclax, anti-tumor efficacy is limited by compensatory induction of MCL-1. Voruciclib, an orally bioavailable clinical stage CDK-selective inhibitor, potently blocks CDK9, the transcriptional regulator of MCL-1. Here, we demonstrate that voruciclib represses MCL-1 protein expression in preclinical models of DLBCL. When combined with venetoclax in vivo, voruciclib leads to model-dependent tumor cell apoptosis and tumor growth inhibition. Strongest responses were observed in two models representing high-risk activated B-cell (ABC) DLBCL, while no response was observed in a third ABC model, and intermediate responses were observed in two models of germinal center B-cell like (GCB) DLBCL. Given the range of responses, we show that CIVO, a multiplexed tumor micro-dosing technology, represents a viable functional precision medicine approach for differentiating responders from non-responders to BCL-2/MCL-1 targeted therapy. These findings suggest that the combination of voruciclib and venetoclax holds promise as a novel, exclusively oral combination therapy for a subset of high-risk DLBCL patients.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzopyrans/pharmacology , Gene Expression Regulation/drug effects , Imino Furanoses/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Antineoplastic Agents/therapeutic use , Benzopyrans/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Line, Tumor , Drug Synergism , Humans , Imino Furanoses/therapeutic use , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
5.
PLoS One ; 11(6): e0158617, 2016.
Article in English | MEDLINE | ID: mdl-27359113

ABSTRACT

While advances in high-throughput screening have resulted in increased ability to identify synergistic anti-cancer drug combinations, validation of drug synergy in the in vivo setting and prioritization of combinations for clinical development remain low-throughput and resource intensive. Furthermore, there is currently no viable method for prospectively assessing drug synergy directly in human patients in order to potentially tailor therapies. To address these issues we have employed the previously described CIVO platform and developed a quantitative approach for investigating multiple combination hypotheses simultaneously in single living tumors. This platform provides a rapid, quantitative and cost effective approach to compare and prioritize drug combinations based on evidence of synergistic tumor cell killing in the live tumor context. Using a gemcitabine resistant model of pancreatic cancer, we efficiently investigated nine rationally selected Abraxane-based combinations employing only 19 xenografted mice. Among the drugs tested, the BCL2/BCLxL inhibitor ABT-263 was identified as the one agent that synergized with Abraxane® to enhance acute induction of localized apoptosis in this model of human pancreatic cancer. Importantly, results obtained with CIVO accurately predicted the outcome of systemic dosing studies in the same model where superior tumor regression induced by the Abraxane/ABT-263 combination was observed compared to that induced by either single agent. This supports expanded use of CIVO as an in vivo platform for expedited in vivo drug combination validation and sets the stage for performing toxicity-sparing drug combination studies directly in cancer patients with solid malignancies.


Subject(s)
Albumin-Bound Paclitaxel/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Pancreatic Neoplasms/drug therapy , Sulfonamides/therapeutic use , Xenograft Model Antitumor Assays/methods , Albumin-Bound Paclitaxel/administration & dosage , Aniline Compounds/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Drug Synergism , Mice , Pancreatic Neoplasms/pathology , Sulfonamides/administration & dosage
6.
Cancer Chemother Pharmacol ; 76(4): 699-712, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26231955

ABSTRACT

PURPOSE: nab-paclitaxel demonstrates improved clinical efficacy compared with conventional Cremophor EL (CrEL)-paclitaxel in multiple tumor types. This study explored the distinctions in drug distribution between nab-paclitaxel and CrEL-paclitaxel and the underlying mechanisms. METHODS: Uptake and transcytosis of paclitaxel were analyzed by vascular permeability assay across human endothelial cell monolayers. The tissue penetration of paclitaxel within tumors was evaluated by local injections into tumor xenografts and quantitative image analysis. The distribution profile of paclitaxel in solid-tumor patients was assessed using pharmacokinetic modeling and simulation. RESULTS: Live imaging demonstrated that albumin and paclitaxel were present in punctae in endothelial cells and could be observed in very close proximity, suggesting cotransport. Uptake and transport of albumin, nab-paclitaxel and paclitaxel were inhibited by clinically relevant CrEL concentrations. Further, nab-paclitaxel causes greater mitotic arrest in wider area within xenografted tumors than CrEL- or dimethyl sulfoxide-paclitaxel following local microinjection, demonstrating enhanced paclitaxel penetration and uptake by albumin within tumors. Modeling of paclitaxel distribution in patients with solid tumors indicated that nab-paclitaxel is more dependent upon transporter-mediated pathways for drug distribution into tissues than CrEL-paclitaxel. The percent dose delivered to tissue via transporter-mediated pathways is predicted to be constant with nab-paclitaxel but decrease with increasing CrEL-paclitaxel dose. CONCLUSIONS: Compared with CrEL-paclitaxel, nab-paclitaxel demonstrated more efficient transport across endothelial cells, greater penetration and cytotoxic induction in xenograft tumors, and enhanced extravascular distribution in patients that are attributed to carrier-mediated transport. These observations are consistent with the distinct clinical efficacy and toxicity profile of nab-paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Drug Delivery Systems , Endothelium, Vascular/metabolism , Nanoparticles/chemistry , Paclitaxel/administration & dosage , Pancreatic Neoplasms/drug therapy , Serum Albumin/chemistry , Animals , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Biological Transport/drug effects , Capillary Permeability/drug effects , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cells, Cultured , Endosomes/drug effects , Endosomes/metabolism , Endosomes/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Infusions, Intravenous , Mice, Nude , Microinjections , Paclitaxel/metabolism , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Serum Albumin/metabolism , Serum Albumin, Human , Tissue Distribution , Tubulin Modulators/administration & dosage , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacokinetics , Tubulin Modulators/therapeutic use , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
7.
Sci Transl Med ; 7(284): 284ra58, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904742

ABSTRACT

A fundamental problem in cancer drug development is that antitumor efficacy in preclinical cancer models does not translate faithfully to patient outcomes. Much of early cancer drug discovery is performed under in vitro conditions in cell-based models that poorly represent actual malignancies. To address this inconsistency, we have developed a technology platform called CIVO, which enables simultaneous assessment of up to eight drugs or drug combinations within a single solid tumor in vivo. The platform is currently designed for use in animal models of cancer and patients with superficial tumors but can be modified for investigation of deeper-seated malignancies. In xenograft lymphoma models, CIVO microinjection of well-characterized anticancer agents (vincristine, doxorubicin, mafosfamide, and prednisolone) induced spatially defined cellular changes around sites of drug exposure, specific to the known mechanisms of action of each drug. The observed localized responses predicted responses to systemically delivered drugs in animals. In pair-matched lymphoma models, CIVO correctly demonstrated tumor resistance to doxorubicin and vincristine and an unexpected enhanced sensitivity to mafosfamide in multidrug-resistant lymphomas compared with chemotherapy-naïve lymphomas. A CIVO-enabled in vivo screen of 97 approved oncology agents revealed a novel mTOR (mammalian target of rapamycin) pathway inhibitor that exhibits significantly increased tumor-killing activity in the drug-resistant setting compared with chemotherapy-naïve tumors. Finally, feasibility studies to assess the use of CIVO in human and canine patients demonstrated that microinjection of drugs is toxicity-sparing while inducing robust, easily tracked, drug-specific responses in autochthonous tumors, setting the stage for further application of this technology in clinical trials.


Subject(s)
Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor/methods , Lymphoma/drug therapy , Neoplasms/drug therapy , Animals , Biomarkers , Cell Line, Tumor , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/chemistry , Dogs , Doxorubicin/chemistry , Drug Delivery Systems , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Prednisolone/chemistry , TOR Serine-Threonine Kinases/metabolism , Vincristine/chemistry
8.
Cancer Res ; 73(22): 6828-37, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24092238

ABSTRACT

While medulloblastoma, a pediatric tumor of the cerebellum, is characterized by aberrations in developmental pathways, the majority of genetic determinants remain unknown. An unbiased Sleeping Beauty transposon screen revealed MyoD as a putative medulloblastoma tumor suppressor. This was unexpected, as MyoD is a muscle differentiation factor and not previously known to be expressed in cerebellum or medulloblastoma. In response to deletion of one allele of MyoD, two other Sonic hedgehog-driven mouse medulloblastoma models showed accelerated tumor formation and death, confirming MyoD as a tumor suppressor in these models. In normal cerebellum, MyoD was expressed in the proliferating granule neuron progenitors that are thought to be precursors to medulloblastoma. Similar to some other tumor suppressors that are induced in cancer, MyoD was expressed in proliferating medulloblastoma cells in three mouse models and in human medulloblastoma cases. This suggests that although expression of MyoD in a proliferating tumor is insufficient to prevent tumor progression, its expression in the cerebellum hinders medulloblastoma genesis.


Subject(s)
Cerebellar Neoplasms/genetics , Genes, Tumor Suppressor/physiology , Medulloblastoma/genetics , MyoD Protein/physiology , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cerebellum/embryology , Cerebellum/metabolism , Disease Progression , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Transgenic , MyoD Protein/genetics
9.
Epigenetics ; 8(12): 1254-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24107773

ABSTRACT

Genetic and epigenetic alterations are essential for the initiation and progression of human cancer. We previously reported that primary human medulloblastomas showed extensive cancer-specific CpG island DNA hypermethylation in critical developmental pathways. To determine whether genetically engineered mouse models (GEMMs) of medulloblastoma have comparable epigenetic changes, we assessed genome-wide DNA methylation in three mouse models of medulloblastoma. In contrast to human samples, very few loci with cancer-specific DNA hypermethylation were detected, and in almost all cases the degree of methylation was relatively modest compared with the dense hypermethylation in the human cancers. To determine if this finding was common to other GEMMs, we examined a Burkitt lymphoma and breast cancer model and did not detect promoter CpG island DNA hypermethylation, suggesting that human cancers and at least some GEMMs are fundamentally different with respect to this epigenetic modification. These findings provide an opportunity to both better understand the mechanism of aberrant DNA methylation in human cancer and construct better GEMMs to serve as preclinical platforms for therapy development.


Subject(s)
CpG Islands , DNA Methylation/genetics , Neoplasms, Experimental/genetics , Neoplasms/genetics , Promoter Regions, Genetic , Animals , Breast Neoplasms/genetics , Burkitt Lymphoma/genetics , Cerebellar Neoplasms/genetics , Female , Humans , Medulloblastoma/genetics , Mice , Mice, Transgenic
10.
Mol Cell Biol ; 32(20): 4104-15, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22869526

ABSTRACT

Deregulated developmental processes in the cerebellum cause medulloblastoma, the most common pediatric brain malignancy. About 25 to 30% of cases are caused by mutations increasing the activity of the Sonic hedgehog (Shh) pathway, a critical mitogen in cerebellar development. The proto-oncogene Smoothened (Smo) is a key transducer of the Shh pathway. Activating mutations in Smo that lead to constitutive activity of the Shh pathway have been identified in human medulloblastoma. To understand the developmental and oncogenic effects of two closely positioned point mutations in Smo, we characterized NeuroD2-SmoA2 mice and compared them to NeuroD2-SmoA1 mice. While both SmoA1 and SmoA2 transgenes cause medulloblastoma with similar frequencies and timing, SmoA2 mice have severe aberrations in cerebellar development, whereas SmoA1 mice are largely normal during development. Intriguingly, neurologic function, as measured by specific tests, is normal in the SmoA2 mice despite extensive cerebellar dysplasia. We demonstrate how two nearly contiguous point mutations in the same domain of the encoded Smo protein can produce striking phenotypic differences in cerebellar development and organization in mice.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellum/abnormalities , Disease Models, Animal , Medulloblastoma/genetics , Mice , Receptors, G-Protein-Coupled/genetics , Animals , Humans , Mice, Transgenic , Point Mutation , Proto-Oncogene Mas , Smoothened Receptor
11.
J Neurosci ; 28(2): 465-72, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18184789

ABSTRACT

Alpha-synuclein aggregates are a common feature of sporadic Parkinson's disease (PD), and mutations that increase alpha-synuclein abundance confer rare heritable forms of PD. Although these findings suggest that alpha-synuclein plays a central role in the pathogenesis of this disorder, little is known of the mechanism by which alpha-synuclein promotes neuron loss or the factors that regulate alpha-synuclein toxicity. To address these matters, we tested candidate modifiers of alpha-synuclein toxicity using a Drosophila model of PD. In the current work, we focused on phase II detoxification enzymes involved in glutathione metabolism. We find that the neuronal death accompanying alpha-synuclein expression in Drosophila is enhanced by loss-of-function mutations in genes that promote glutathione synthesis and glutathione conjugation. This neuronal loss can be overcome by genetic or pharmacological interventions that increase glutathione synthesis or glutathione conjugation activity. Moreover, these same pharmacological agents suppress neuron loss in Drosophila parkin mutants, a loss-of-function model of PD. Our results suggest that oxidative stress is a feature of alpha-synuclein toxicity and that induction of the phase II detoxification pathway represents a potential preventative therapy for PD.


Subject(s)
Metabolic Detoxication, Phase II/physiology , Metabolic Networks and Pathways/physiology , Neurons/physiology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Age Factors , Allyl Compounds , Animals , Animals, Genetically Modified , Cell Death/genetics , Disease Models, Animal , Disulfides/pharmacology , Dose-Response Relationship, Drug , Drosophila , Drosophila Proteins/genetics , Glutathione/metabolism , Isothiocyanates , Mutation , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/genetics , Neurons/pathology , Parkinson Disease/genetics , Sulfoxides , Thiocyanates/pharmacology , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...