Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Biol ; 20(5): e3001624, 2022 05.
Article in English | MEDLINE | ID: mdl-35617197

ABSTRACT

Test compounds used on in vitro model systems are conventionally delivered to cell culture wells as fixed concentration bolus doses; however, this poorly replicates the pharmacokinetic (PK) concentration changes seen in vivo and reduces the predictive value of the data. Herein, proof-of-concept experiments were performed using a novel microfluidic device, the Microformulator, which allows in vivo like PK profiles to be applied to cells cultured in microtiter plates and facilitates the investigation of the impact of PK on biological responses. We demonstrate the utility of the device in its ability to reproduce in vivo PK profiles of different oncology compounds over multiweek experiments, both as monotherapy and drug combinations, comparing the effects on tumour cell efficacy in vitro with efficacy seen in in vivo xenograft models. In the first example, an ERK1/2 inhibitor was tested using fixed bolus dosing and Microformulator-replicated PK profiles, in 2 cell lines with different in vivo sensitivities. The Microformulator-replicated PK profiles were able to discriminate between cell line sensitivities, unlike the conventional fixed bolus dosing. In a second study, murine in vivo PK profiles of multiple Poly(ADP-Ribose) Polymerase 1/2 (PARP) and DNA-dependent protein kinase (DNA-PK) inhibitor combinations were replicated in a FaDu cell line resulting in a reduction in cell growth in vitro with similar rank ordering to the in vivo xenograft model. Additional PK/efficacy insight into theoretical changes to drug exposure profiles was gained by using the Microformulator to expose FaDu cells to the DNA-PK inhibitor for different target coverage levels and periods of time. We demonstrate that the Microformulator enables incorporating PK exposures into cellular assays to improve in vitro-in vivo translation understanding for early therapeutic insight.


Subject(s)
Cell Culture Techniques , Microfluidics , Animals , DNA , Humans , Mice , Models, Biological
2.
Mol Cancer Ther ; 20(9): 1614-1626, 2021 09.
Article in English | MEDLINE | ID: mdl-34158341

ABSTRACT

Radiotherapy is an effective anticancer treatment, but combinations with targeted agents that maximize efficacy while sparing normal tissue are needed. Here, we assess the radiopotentiation profiles of DNA damage response inhibitors (DDRi) olaparib (PARP1/2), ceralasertib (ATR), adavosertib (WEE1), AZD0156 (ATM), and KU-60648 (DNA-PK). We performed a radiotherapy combination screen and assessed how drug concentration and cellular DDR deficiencies influence the radiopotentiation ability of DDRi. We pre-selected six lung cancer cell lines with different genetic/signaling aberrations (including mutations in TP53 and ATM) and assessed multiple concentrations of DDRi in combination with a fixed radiotherapy dose by clonogenic assay. The effective concentration of DDRi in radiotherapy combinations is lower than that required for single-agent efficacy. This has the potential to be exploited further in the context of DDR deficiencies to increase therapeutic index and we demonstrate that low concentrations of AZD0156 preferentially sensitized p53-deficient cells. Moreover, testing multiple concentrations of DDRi in radiotherapy combinations indicated that olaparib, ceralasertib, and adavosertib have a desirable safety profile showing moderate increases in radiotherapy dose enhancement with increasing inhibitor concentration. Small increases in concentration of AZD0156 and particularly KU-60648, however, result in steep increases in dose enhancement. Radiopotentiation profiling can inform on effective drug doses required for radiosensitization in relation to biomarkers, providing an opportunity to increase therapeutic index. Moreover, multiple concentration testing demonstrates a relationship between drug concentration and radiotherapy effect that provides valuable insights that, with future in vivo validation, can guide dose-escalation strategies in clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , DNA Repair , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Apoptosis , Cell Proliferation , Humans , Lung Neoplasms/pathology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Tumor Cells, Cultured
3.
Nat Commun ; 10(1): 5065, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31699977

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is a critical player in the DNA damage response (DDR) and instrumental in the non-homologous end-joining pathway (NHEJ) used to detect and repair DNA double-strand breaks (DSBs). We demonstrate that the potent and highly selective DNA-PK inhibitor, AZD7648, is an efficient sensitizer of radiation- and doxorubicin-induced DNA damage, with combinations in xenograft and patient-derived xenograft (PDX) models inducing sustained regressions. Using ATM-deficient cells, we demonstrate that AZD7648, in combination with the PARP inhibitor olaparib, increases genomic instability, resulting in cell growth inhibition and apoptosis. AZD7648 enhanced olaparib efficacy across a range of doses and schedules in xenograft and PDX models, enabling sustained tumour regression and providing a clear rationale for its clinical investigation. Through its differentiated mechanism of action as an NHEJ inhibitor, AZD7648 complements the current armamentarium of DDR-targeted agents and has potential in combination with these agents to achieve deeper responses to current therapies.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Drug Synergism , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyrans/pharmacology , Radiation Tolerance/drug effects , Triazoles/pharmacology , A549 Cells , Animals , Antibiotics, Antineoplastic/pharmacology , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Genomic Instability/drug effects , Humans , Lung Neoplasms , Mice , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Polyethylene Glycols/pharmacology , Radiotherapy , Xenograft Model Antitumor Assays
4.
Sci Adv ; 4(6): eaat1719, 2018 06.
Article in English | MEDLINE | ID: mdl-29938225

ABSTRACT

Poor survival rates of patients with tumors arising from or disseminating into the brain are attributed to an inability to excise all tumor tissue (if operable), a lack of blood-brain barrier (BBB) penetration of chemotherapies/targeted agents, and an intrinsic tumor radio-/chemo-resistance. Ataxia-telangiectasia mutated (ATM) protein orchestrates the cellular DNA damage response (DDR) to cytotoxic DNA double-strand breaks induced by ionizing radiation (IR). ATM genetic ablation or pharmacological inhibition results in tumor cell hypersensitivity to IR. We report the primary pharmacology of the clinical-grade, exquisitely potent (cell IC50, 0.78 nM), highly selective [>10,000-fold over kinases within the same phosphatidylinositol 3-kinase-related kinase (PIKK) family], orally bioavailable ATM inhibitor AZD1390 specifically optimized for BBB penetration confirmed in cynomolgus monkey brain positron emission tomography (PET) imaging of microdosed 11C-labeled AZD1390 (Kp,uu, 0.33). AZD1390 blocks ATM-dependent DDR pathway activity and combines with radiation to induce G2 cell cycle phase accumulation, micronuclei, and apoptosis. AZD1390 radiosensitizes glioma and lung cancer cell lines, with p53 mutant glioma cells generally being more radiosensitized than wild type. In in vivo syngeneic and patient-derived glioma as well as orthotopic lung-brain metastatic models, AZD1390 dosed in combination with daily fractions of IR (whole-brain or stereotactic radiotherapy) significantly induced tumor regressions and increased animal survival compared to IR treatment alone. We established a pharmacokinetic-pharmacodynamic-efficacy relationship by correlating free brain concentrations, tumor phospho-ATM/phospho-Rad50 inhibition, apoptotic biomarker (cleaved caspase-3) induction, tumor regression, and survival. On the basis of the data presented here, AZD1390 is now in early clinical development for use as a radiosensitizer in central nervous system malignancies.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Protein Kinase Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Membrane Permeability , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Mice , Phosphorylation , Protein Kinase Inhibitors/chemistry , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/chemistry , Signal Transduction/drug effects , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , X-Rays , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 24(10): 2395-2407, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29391353

ABSTRACT

Purpose: Myeloma is a plasma cell malignancy characterized by the overproduction of immunoglobulin, and is therefore susceptible to therapies targeting protein homeostasis. We hypothesized that heat shock factor 1 (HSF1) was an attractive therapeutic target for myeloma due to its direct regulation of transcriptional programs implicated in both protein homeostasis and the oncogenic phenotype. Here, we interrogate HSF1 as a therapeutic target in myeloma using bioinformatic, genetic, and pharmacologic means.Experimental Design: To assess the clinical relevance of HSF1, we analyzed publicly available patient myeloma gene expression datasets. Validation of this novel target was conducted in in vitro experiments using shRNA or inhibitors of the HSF1 pathway in human myeloma cell lines and primary cells as well as in in vivo human myeloma xenograft models.Results: Expression of HSF1 and its target genes were associated with poorer myeloma patient survival. ShRNA-mediated knockdown or pharmacologic inhibition of the HSF1 pathway with a novel chemical probe, CCT251236, or with KRIBB11, led to caspase-mediated cell death that was associated with an increase in EIF2α phosphorylation, CHOP expression and a decrease in overall protein synthesis. Importantly, both CCT251236 and KRIBB11 induced cytotoxicity in human myeloma cell lines and patient-derived primary myeloma cells with a therapeutic window over normal cells. Pharmacologic inhibition induced tumor growth inhibition and was well-tolerated in a human myeloma xenograft murine model with evidence of pharmacodynamic biomarker modulation.Conclusions: Taken together, our studies demonstrate the dependence of myeloma cells on HSF1 for survival and support the clinical evaluation of pharmacologic inhibitors of the HSF1 pathway in myeloma. Clin Cancer Res; 24(10); 2395-407. ©2018 AACRSee related commentary by Parekh, p. 2237.


Subject(s)
Biomarkers, Tumor , Cell Survival/genetics , Heat Shock Transcription Factors/genetics , Multiple Myeloma/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Caspases/metabolism , Cell Line, Tumor , Computational Biology/methods , Disease Models, Animal , Gene Expression Profiling , Gene Knockdown Techniques , Heat Shock Transcription Factors/antagonists & inhibitors , Heat Shock Transcription Factors/metabolism , Humans , Kaplan-Meier Estimate , Mice , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/mortality , Signal Transduction , Xenograft Model Antitumor Assays
6.
PLoS One ; 9(6): e99493, 2014.
Article in English | MEDLINE | ID: mdl-24923560

ABSTRACT

The chromosomal translocation t(4;14) deregulates MMSET (WHSC1/NSD2) expression and is a poor prognostic factor in multiple myeloma (MM). MMSET encodes two major protein isoforms. We have characterized the role of the shorter isoform (REIIBP) in myeloma cells and identified a clear and novel interaction of REIIBP with members of the SMN (survival of motor neuron) complex that directly affects the assembly of the spliceosomal ribonucleic particles. Using RNA-seq we show that REIIBP influences the RNA splicing pattern of the cell. This new discovery provides novel insights into the understanding of MM pathology, and potential new leads for therapeutic targeting.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , RNA Processing, Post-Transcriptional , Repressor Proteins/metabolism , SMN Complex Proteins/metabolism , Cell Proliferation , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , HeLa Cells , Histones/metabolism , Humans , Introns/genetics , Mass Spectrometry , Methylation , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Phenotype , Protein Binding , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA Splicing/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/metabolism , Translocation, Genetic
7.
Oncotarget ; 5(5): 1132-48, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24675290

ABSTRACT

Heat shock proteins are molecular chaperones with a central role in protein folding and cellular protein homeostasis. They also play major roles in the development of cancer and in recent years have emerged as promising therapeutic targets. In this review, we discuss the known molecular mechanisms of various heat shock protein families and their involvement in cancer and in particular, multiple myeloma. In addition, we address the current progress and challenges in pharmacologically targeting these proteins as anti-cancer therapeutic strategies.


Subject(s)
Heat-Shock Proteins/physiology , Multiple Myeloma/drug therapy , Chaperonin 10/metabolism , Chaperonin 60/metabolism , DNA-Binding Proteins/antagonists & inhibitors , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Humans , Multiple Myeloma/metabolism , Protein Folding , Signal Transduction , Transcription Factors/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...