Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Nature ; 568(7753): 551-556, 2019 04.
Article in English | MEDLINE | ID: mdl-30971823

ABSTRACT

Synthetic lethality-an interaction between two genetic events through which the co-occurrence of these two genetic events leads to cell death, but each event alone does not-can be exploited for cancer therapeutics1. DNA repair processes represent attractive synthetic lethal targets, because many cancers exhibit an impairment of a DNA repair pathway, which can lead to dependence on specific repair proteins2. The success of poly(ADP-ribose) polymerase 1 (PARP-1) inhibitors in cancers with deficiencies in homologous recombination highlights the potential of this approach3. Hypothesizing that other DNA repair defects would give rise to synthetic lethal relationships, we queried dependencies in cancers with microsatellite instability (MSI), which results from deficient DNA mismatch repair. Here we analysed data from large-scale silencing screens using CRISPR-Cas9-mediated knockout and RNA interference, and found that the RecQ DNA helicase WRN was selectively essential in MSI models in vitro and in vivo, yet dispensable in models of cancers that are microsatellite stable. Depletion of WRN induced double-stranded DNA breaks and promoted apoptosis and cell cycle arrest selectively in MSI models. MSI cancer models required the helicase activity of WRN, but not its exonuclease activity. These findings show that WRN is a synthetic lethal vulnerability and promising drug target for MSI cancers.


Subject(s)
Microsatellite Instability , Microsatellite Repeats/genetics , Neoplasms/genetics , Synthetic Lethal Mutations/genetics , Werner Syndrome Helicase/genetics , Apoptosis/genetics , CRISPR-Cas Systems/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , Humans , Models, Genetic , Neoplasms/pathology , RNA Interference , Tumor Suppressor Protein p53/metabolism , Werner Syndrome Helicase/deficiency
2.
Nano Lett ; 17(3): 1733-1740, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28145723

ABSTRACT

Selective killing of cancer cells while minimizing damage to healthy tissues is the goal of clinical radiation therapy. This therapeutic ratio can be improved by image-guided radiation delivery and selective radiosensitization of cancer cells. Here, we have designed and tested a novel trimodal theranostic nanoparticle made of bismuth and gadolinium for on-site radiosensitization and image contrast enhancement to improve the efficacy and accuracy of radiation therapy. We demonstrate in vivo magnetic resonance (MR), computed tomography (CT) contrast enhancement, and tumor suppression with prolonged survival in a non-small cell lung carcinoma model during clinical radiation therapy. Histological studies show minimal off-target toxicities due to the nanoparticles or radiation. By mimicking existing clinical workflows, we show that the bismuth-gadolinium nanoparticles are highly compatible with current CT-guided radiation therapy and emerging MR-guided approaches. This study reports the first in vivo proof-of-principle for image-guided radiation therapy with a new class of theranostic nanoparticles.


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenocarcinoma/radiotherapy , Bismuth/therapeutic use , Contrast Media/therapeutic use , Gadolinium/therapeutic use , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/radiotherapy , Nanoparticles/therapeutic use , Silicon Dioxide/therapeutic use , A549 Cells , Adenocarcinoma of Lung , Animals , Bismuth/chemistry , Contrast Media/chemistry , Gadolinium/chemistry , Humans , Magnetic Resonance Imaging , Mice , Nanoparticles/chemistry , Radiotherapy, Image-Guided , Silicon Dioxide/chemistry , Theranostic Nanomedicine , Tomography, X-Ray Computed
4.
Clin Cancer Res ; 25(20): 6127-6140, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31409614

ABSTRACT

PURPOSE: PARP inhibitors are approved for the treatment of high-grade serous ovarian cancers (HGSOC). Therapeutic resistance, resulting from restoration of homologous recombination (HR) repair or replication fork stabilization, is a pressing clinical problem. We assessed the activity of prexasertib, a checkpoint kinase 1 (CHK1) inhibitor known to cause replication catastrophe, as monotherapy and in combination with the PARP inhibitor olaparib in preclinical models of HGSOC, including those with acquired PARP inhibitor resistance. EXPERIMENTAL DESIGN: Prexasertib was tested as a single agent or in combination with olaparib in 14 clinically annotated and molecularly characterized luciferized HGSOC patient-derived xenograft (PDX) models and in a panel of ovarian cancer cell lines. The ability of prexasertib to impair HR repair and replication fork stability was also assessed. RESULTS: Prexasertib monotherapy demonstrated antitumor activity across the 14 PDX models. Thirteen models were resistant to olaparib monotherapy, including 4 carrying BRCA1 mutation. The combination of olaparib with prexasertib was synergistic and produced significant tumor growth inhibition in an olaparib-resistant model and further augmented the degree and durability of response in the olaparib-sensitive model. HGSOC cell lines, including those with acquired PARP inhibitor resistance, were also sensitive to prexasertib, associated with induction of DNA damage and replication stress. Prexasertib also sensitized these cell lines to PARP inhibition and compromised both HR repair and replication fork stability. CONCLUSIONS: Prexasertib exhibits monotherapy activity in PARP inhibitor-resistant HGSOC PDX and cell line models, reverses restored HR and replication fork stability, and synergizes with PARP inhibition.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Cystadenocarcinoma, Serous/drug therapy , Ovarian Neoplasms/drug therapy , Pyrazines/pharmacology , Pyrazoles/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , BRCA1 Protein/genetics , Cell Line, Tumor , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , DNA Damage/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Humans , Neoplasm Grading , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrazines/therapeutic use , Pyrazoles/therapeutic use , Recombinational DNA Repair/drug effects , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 25(3): 977-988, 2019 02 01.
Article in English | MEDLINE | ID: mdl-29980530

ABSTRACT

PURPOSE: DNA-damaging agents comprise the backbone of systemic treatment for many tumor types; however, few reliable predictive biomarkers are available to guide use of these agents. In muscle-invasive bladder cancer (MIBC), cisplatin-based chemotherapy improves survival, yet response varies widely among patients. Here, we sought to define the role of the nucleotide excision repair (NER) gene ERCC2 as a biomarker predictive of response to cisplatin in MIBC. EXPERIMENTAL DESIGN: Somatic missense mutations in ERCC2 are associated with improved response to cisplatin-based chemotherapy; however, clinically identified ERCC2 mutations are distributed throughout the gene, and the impact of individual ERCC2 variants on NER capacity and cisplatin sensitivity is unknown. We developed a microscopy-based NER assay to profile ERCC2 mutations observed retrospectively in prior studies and prospectively within the context of an institution-wide tumor profiling initiative. In addition, we created the first ERCC2-deficient bladder cancer preclinical model for studying the impact of ERCC2 loss of function. RESULTS: We used our functional assay to test the NER capacity of clinically observed ERCC2 mutations and found that most ERCC2 helicase domain mutations cannot support NER. Furthermore, we show that introducing an ERCC2 mutation into a bladder cancer cell line abrogates NER activity and is sufficient to drive cisplatin sensitivity in an orthotopic xenograft model. CONCLUSIONS: Our data support a direct role for ERCC2 mutations in driving cisplatin response, define the functional landscape of ERCC2 mutations in bladder cancer, and provide an opportunity to apply combined genomic and functional approaches to prospectively guide therapy decisions in bladder cancer.See related commentary by Grivas, p. 907.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA Repair/genetics , Mutation, Missense , Urinary Bladder Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Xeroderma Pigmentosum Group D Protein/genetics , Animals , Cell Line , Cell Line, Tumor , Cisplatin/administration & dosage , Cohort Studies , Female , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Muscles/pathology , Neoplasm Invasiveness , Survival Analysis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL