Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Nature ; 593(7859): 429-434, 2021 05.
Article in English | MEDLINE | ID: mdl-34012082

ABSTRACT

Gene-editing technologies, which include the CRISPR-Cas nucleases1-3 and CRISPR base editors4,5, have the potential to permanently modify disease-causing genes in patients6. The demonstration of durable editing in target organs of nonhuman primates is a key step before in vivo administration of gene editors to patients in clinical trials. Here we demonstrate that CRISPR base editors that are delivered in vivo using lipid nanoparticles can efficiently and precisely modify disease-related genes in living cynomolgus monkeys (Macaca fascicularis). We observed a near-complete knockdown of PCSK9 in the liver after a single infusion of lipid nanoparticles, with concomitant reductions in blood levels of PCSK9 and low-density lipoprotein cholesterol of approximately 90% and about 60%, respectively; all of these changes remained stable for at least 8 months after a single-dose treatment. In addition to supporting a 'once-and-done' approach to the reduction of low-density lipoprotein cholesterol and the treatment of atherosclerotic cardiovascular disease (the leading cause of death worldwide7), our results provide a proof-of-concept for how CRISPR base editors can be productively applied to make precise single-nucleotide changes in therapeutic target genes in the liver, and potentially in other organs.


Subject(s)
CRISPR-Cas Systems , Cholesterol, LDL/blood , Gene Editing , Models, Animal , Proprotein Convertase 9/genetics , Adenine/metabolism , Animals , Cells, Cultured , Female , Hepatocytes/metabolism , Humans , Liver/enzymology , Loss of Function Mutation , Macaca fascicularis/blood , Macaca fascicularis/genetics , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Proprotein Convertase 9/blood , Proprotein Convertase 9/metabolism , Time Factors
2.
Circulation ; 147(3): 242-253, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36314243

ABSTRACT

BACKGROUND: VERVE-101 is an investigational in vivo CRISPR base-editing medicine designed to alter a single DNA base in the PCSK9 gene, permanently turn off hepatic protein production, and thereby durably lower low-density lipoprotein cholesterol. We test the efficacy, durability, tolerability, and potential for germline editing of VERVE-101 in studies of nonhuman primates and a murine F1 progeny study. METHODS: Cynomolgus monkeys were given a single intravenous infusion of a vehicle control (n=10) or VERVE-101 at a dose of 0.75 mg/kg (n=4) or 1.5 mg/kg (n=22) with subsequent follow-up up to 476 days. Two studies assessed the potential for germline editing, including sequencing sperm samples from sexually mature male nonhuman primates treated with VERVE-101 and genotyping offspring from female mice treated with the murine surrogate of VERVE-101 (VERVE-101mu). RESULTS: Liver biopsies 14 days after dosing noted mean PCSK9 editing of 46% and 70% in monkeys treated with VERVE-101 at 0.75 and 1.5 mg/kg, respectively. This translated into mean reductions in blood PCSK9 (proprotein convertase subtilisin/kexin type 9) of 67% and 83% and reductions of low-density lipoprotein cholesterol of 49% and 69% at the 0.75 and 1.5 mg/kg doses, respectively, assessed as time-weighted average change from baseline between day 28 and up to 476 days after dosing. Liver safety monitoring noted a transient rise in alanine aminotransferase and aspartate aminotransferase concentrations after infusion that fully resolved by day 14 with no accompanying change in total bilirubin. In a subset of monkeys necropsied 1 year after dosing, no findings related to VERVE-101 were identified on macroscopic and histopathologic assessment of the liver and other organs. In the study to assess potential germline editing of male nonhuman primates, sperm samples collected after VERVE-101 dosing showed no evidence of PCSK9 editing. Among 436 offspring of female mice treated with a saturating dose of VERVE-101mu, the PCSK9 edit was transmitted in 0 of 436 animals. CONCLUSIONS: VERVE-101 was well tolerated in nonhuman primates and led to 83% lower blood PCSK9 protein and 69% lower low-density lipoprotein cholesterol with durable effects up to 476 days after dosing. These results have supported the initiation of a first-in-human clinical trial in patients with heterozygous familial hypercholesterolemia and atherosclerotic cardiovascular disease.


Subject(s)
Gene Editing , Proprotein Convertase 9 , Animals , Female , Humans , Male , Mice , Cholesterol, LDL/genetics , Cholesterol, LDL/metabolism , Primates/genetics , Primates/metabolism , Proprotein Convertase 9/genetics , Proprotein Convertase 9/therapeutic use , Semen/metabolism , Gene Editing/methods , CRISPR-Cas Systems , Genetic Therapy/methods , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/therapy , Atherosclerosis/genetics , Atherosclerosis/therapy
3.
Nat Commun ; 14(1): 2776, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37188660

ABSTRACT

Lipid nanoparticles have demonstrated utility in hepatic delivery of a range of therapeutic modalities and typically deliver their cargo via low-density lipoprotein receptor-mediated endocytosis. For patients lacking sufficient low-density lipoprotein receptor activity, such as those with homozygous familial hypercholesterolemia, an alternate strategy is needed. Here we show the use of structure-guided rational design in a series of mouse and non-human primate studies to optimize a GalNAc-Lipid nanoparticle that allows for low-density lipoprotein receptor independent delivery. In low-density lipoprotein receptor-deficient non-human primates administered a CRISPR base editing therapy targeting the ANGPTL3 gene, the introduction of an optimized GalNAc-based asialoglycoprotein receptor ligand to the nanoparticle surface increased liver editing from 5% to 61% with minimal editing in nontargeted tissues. Similar editing was noted in wild-type monkeys, with durable blood ANGPTL3 protein reduction up to 89% six months post dosing. These results suggest that GalNAc-Lipid nanoparticles may effectively deliver to both patients with intact low-density lipoprotein receptor activity as well as those afflicted by homozygous familial hypercholesterolemia.


Subject(s)
Homozygous Familial Hypercholesterolemia , Nanoparticles , Animals , Gene Editing/methods , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Liver/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Lipoproteins, LDL/metabolism
4.
Cancer Discov ; 7(3): 277-287, 2017 03.
Article in English | MEDLINE | ID: mdl-27986707

ABSTRACT

Recent studies have identified somatic ESR1 mutations in patients with metastatic breast cancer and found some of them to promote estrogen-independent activation of the receptor. The degree to which all recurrent mutants can drive estrogen-independent activities and reduced sensitivity to ER antagonists like fulvestrant is not established. In this report, we characterize the spectrum of ESR1 mutations from more than 900 patients. ESR1 mutations were detected in 10%, with D538G being the most frequent (36%), followed by Y537S (14%). Several novel, activating mutations were also detected (e.g., L469V, V422del, and Y537D). Although many mutations lead to constitutive activity and reduced sensitivity to ER antagonists, only select mutants such as Y537S caused a magnitude of change associated with fulvestrant resistance in vivo Correspondingly, tumors driven by Y537S, but not D5358G, E380Q, or S463P, were less effectively inhibited by fulvestrant than more potent and bioavailable antagonists, including AZD9496. These data point to a need for antagonists with optimal pharmacokinetic properties to realize clinical efficacy against certain ESR1 mutants.Significance: A diversity of activating ESR1 mutations exist, only some of which confer resistance to existing ER antagonists that might be overcome by next-generation inhibitors such as AZD9496. Cancer Discov; 7(3); 277-87. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 235.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Cinnamates/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fulvestrant , Humans , Indoles/pharmacology , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Mutation , Protein Domains , Xenograft Model Antitumor Assays
5.
Oncotarget ; 7(15): 20773-87, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26980748

ABSTRACT

The tumor microenvironment is emerging as a key regulator of cancer growth and progression, however the exact mechanisms of interaction with the tumor are poorly understood. Whilst the majority of genomic profiling efforts thus far have focused on the tumor, here we investigate RNA-Seq as a hypothesis-free tool to generate independent tumor and stromal biomarkers, and explore tumor-stroma interactions by exploiting the human-murine compartment specificity of patient-derived xenografts (PDX).Across a pan-cancer cohort of 79 PDX models, we determine that mouse stroma can be separated into distinct clusters, each corresponding to a specific stromal cell type. This implies heterogeneous recruitment of mouse stroma to the xenograft independent of tumor type. We then generate cross-species expression networks to recapitulate a known association between tumor epithelial cells and fibroblast activation, and propose a potentially novel relationship between two hypoxia-associated genes, human MIF and mouse Ddx6. Assessment of disease subtype also reveals MMP12 as a putative stromal marker of triple-negative breast cancer. Finally, we establish that our ability to dissect recruited stroma from trans-differentiated tumor cells is crucial to identifying stem-like poor-prognosis signatures in the tumor compartment.In conclusion, RNA-Seq is a powerful, cost-effective solution to global analysis of human tumor and mouse stroma simultaneously, providing new insights into mouse stromal heterogeneity and compartment-specific disease markers that are otherwise overlooked by alternative technologies. The study represents the first comprehensive analysis of its kind across multiple PDX models, and supports adoption of the approach in pre-clinical drug efficacy studies, and compartment-specific biomarker discovery.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Epithelial Cells/pathology , Gene Expression Profiling/methods , Stromal Cells/pathology , Transcriptome , Tumor Microenvironment/genetics , Animals , Breast Neoplasms/genetics , Epithelial Cells/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Stromal Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Oncotarget ; 7(34): 54120-54136, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27472462

ABSTRACT

Although endocrine therapy is successfully used to treat patients with estrogen receptor (ER) positive breast cancer, a substantial proportion of this population will relapse. Several mechanisms of acquired resistance have been described including activation of the mTOR pathway, increased activity of CDK4 and activating mutations in ER. Using a patient derived xenograft model harboring a common activating ER ligand binding domain mutation (D538G), we evaluated several combinatorial strategies using the selective estrogen receptor degrader (SERD) fulvestrant in combination with chromatin modifying agents, and CDK4/6 and mTOR inhibitors. In this model, fulvestrant binds WT and MT ER, reduces ER protein levels, and downregulated ER target gene expression. Addition of JQ1 or vorinostat to fulvestrant resulted in tumor regression (41% and 22% regression, respectively) though no efficacy was seen when either agent was given alone. Interestingly, although the CDK4/6 inhibitor palbociclib and mTOR inhibitor everolimus were efficacious as monotherapies, long-term delayed tumor growth was only observed when co-administered with fulvestrant. This observation was consistent with a greater inhibition of compensatory signaling when palbociclib and everolimus were co-dosed with fulvestrant. The addition of fulvestrant to JQ1, vorinostat, everolimus and palbociclib also significantly reduced lung metastatic burden as compared to monotherapy. The combination potential of fulvestrant with palbociclib or everolimus were confirmed in an MCF7 CRISPR model harboring the Y537S ER activating mutation. Taken together, these data suggest that fulvestrant may have an important role in the treatment of ER positive breast cancer with acquired ER mutations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Mutation , Receptors, Estrogen/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Everolimus/administration & dosage , Female , Fulvestrant , Humans , MCF-7 Cells , Mice , Piperazines/administration & dosage , Pyridines/administration & dosage , Receptors, Estrogen/analysis , Xenograft Model Antitumor Assays
7.
Cancer Res ; 76(11): 3307-18, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27020862

ABSTRACT

Fulvestrant is an estrogen receptor (ER) antagonist administered to breast cancer patients by monthly intramuscular injection. Given its present limitations of dosing and route of administration, a more flexible orally available compound has been sought to pursue the potential benefits of this drug in patients with advanced metastatic disease. Here we report the identification and characterization of AZD9496, a nonsteroidal small-molecule inhibitor of ERα, which is a potent and selective antagonist and downregulator of ERα in vitro and in vivo in ER-positive models of breast cancer. Significant tumor growth inhibition was observed as low as 0.5 mg/kg dose in the estrogen-dependent MCF-7 xenograft model, where this effect was accompanied by a dose-dependent decrease in PR protein levels, demonstrating potent antagonist activity. Combining AZD9496 with PI3K pathway and CDK4/6 inhibitors led to further growth-inhibitory effects compared with monotherapy alone. Tumor regressions were also seen in a long-term estrogen-deprived breast model, where significant downregulation of ERα protein was observed. AZD9496 bound and downregulated clinically relevant ESR1 mutants in vitro and inhibited tumor growth in an ESR1-mutant patient-derived xenograft model that included a D538G mutation. Collectively, the pharmacologic evidence showed that AZD9496 is an oral, nonsteroidal, selective estrogen receptor antagonist and downregulator in ER(+) breast cells that could provide meaningful benefit to ER(+) breast cancer patients. AZD9496 is currently being evaluated in a phase I clinical trial. Cancer Res; 76(11); 3307-18. ©2016 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Cinnamates/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Indoles/pharmacology , Mutation/genetics , Administration, Oral , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cinnamates/administration & dosage , Drug Evaluation, Preclinical , Estrogen Receptor Modulators/administration & dosage , Estrogen Receptor alpha/chemistry , Female , Humans , Indoles/administration & dosage , Mice , Mice, Inbred NOD , Mice, SCID , Protein Conformation , Rats , Tumor Cells, Cultured , Uterus/metabolism , Uterus/pathology , Xenograft Model Antitumor Assays
8.
Oncotarget ; 6(4): 2407-20, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25537515

ABSTRACT

Acquired resistance to PI3K/mTOR/Akt pathway inhibitors is often associated with compensatory feedback loops involving the activation of oncogenes. Here, we have generated everolimus resistance in ER+ breast cancer cells and in long-term estrogen deprived (LTED) models that mimic progression on anti-estrogens. This allowed us to uncover MYC as a driver of mTOR inhibitor resistance. We demonstrate that both everolimus resistance and acute treatment of everolimus can lead to the upregulation of MYC mRNA, protein expression and, consequently, the enrichment of MYC signatures as revealed by RNA sequencing data. Depletion of MYC resulted in resensitization to everolimus, confirming its functional importance in this setting. Furthermore, ChIP assays demonstrate that MYC upregulation in the everolimus resistant lines is mediated by increased association of the BRD4 transcription factor with the MYC gene. Finally, JQ1, a BRD4 inhibitor combined with everolimus exhibited increased tumor growth inhibition in 3D Matrigel models and an in vivo xenograft model. These data suggest that MYC plays an important role in mediating resistance to everolimus in ER+ and ER+/LTED models. Furthermore, given the regulation ofMYCby BRD4 in this setting, these data have implications for increased therapeutic potential of combining epigenetic agents with mTOR inhibitors to effectively downregulate otherwise difficult to target transcription factors such as MYC.


Subject(s)
Breast Neoplasms/drug therapy , Epigenesis, Genetic/drug effects , Everolimus/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , MCF-7 Cells , Mice, Nude , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Triazoles/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 13(2): 386-98, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344235

ABSTRACT

The hedgehog pathway has been implicated in the tumorigenesis, tumor progression, and metastasis of numerous human cancers. We generated the first fully human hedgehog antibody MEDI-5304 and characterized its antitumor activity and preclinical toxicology. MEDI-5304 bound sonic hedgehog (SHH) and Indian hedgehog (IHH) with low picomolar affinity and neutralized SHH and IHH activity in cellular mGLI1 reporter assays. The antibody inhibited transcription of hedgehog target genes and osteoblast differentiation of C3H10T1/2 cells. We evaluated the activity of MEDI-5304 in vivo in model systems that allowed us to evaluate two primary hypotheses of hedgehog function in human cancer, paracrine signaling between tumor and stromal cells and cancer stem cell (CSC) self-renewal. MEDI-5304 displayed robust pharmacodynamic effects in stromal cells that translated to antitumor efficacy as a single agent in an HT-29/MEF coimplantation model of paracrine hedgehog signaling. MEDI-5304 also improved responses to carboplatin in the HT-29/MEF model. The antibody, however, had no effect as a single agent or in combination with gemcitabine on the CSC frequency or growth of several primary pancreatic cancer explant models. These findings support the conclusion that hedgehog contributes to tumor biology via paracrine tumor-stromal signaling but not via CSC maintenance or propagation. Finally, the only safety study finding associated with MEDI-5304 was ondontodysplasia in rats. Thus, MEDI-5304 represents a potent dual hedgehog inhibitor suitable for continued development to evaluate efficacy and safety in human patients with tumors harboring elevated levels of SHH or IHH.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Paracrine Communication/drug effects , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , HT29 Cells , Hedgehog Proteins/immunology , Humans , Kinetics , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , NIH 3T3 Cells , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Paracrine Communication/immunology , Protein Binding/immunology , Rats, Wistar , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL