Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 507
Filter
1.
Nat Commun ; 15(1): 5345, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937474

ABSTRACT

Drug-tolerance has emerged as one of the major non-genetic adaptive processes driving resistance to targeted therapy (TT) in non-small cell lung cancer (NSCLC). However, the kinetics and sequence of molecular events governing this adaptive response remain poorly understood. Here, we combine real-time monitoring of the cell-cycle dynamics and single-cell RNA sequencing in a broad panel of oncogenic addiction such as EGFR-, ALK-, BRAF- and KRAS-mutant NSCLC, treated with their corresponding TT. We identify a common path of drug adaptation, which invariably involves alveolar type 1 (AT1) differentiation and Rho-associated protein kinase (ROCK)-mediated cytoskeletal remodeling. We also isolate and characterize a rare population of early escapers, which represent the earliest resistance-initiating cells that emerge in the first hours of treatment from the AT1-like population. A phenotypic drug screen identify farnesyltransferase inhibitors (FTI) such as tipifarnib as the most effective drugs in preventing relapse to TT in vitro and in vivo in several models of oncogenic addiction, which is confirmed by genetic depletion of the farnesyltransferase. These findings pave the way for the development of treatments combining TT and FTI to effectively prevent tumor relapse in oncogene-addicted NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug Resistance, Neoplasm , Farnesyltranstransferase , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Farnesyltranstransferase/genetics , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Animals , Mice , Oncogene Addiction/genetics , Molecular Targeted Therapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Xenograft Model Antitumor Assays , Oncogenes/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Quinolones
2.
Signal Transduct Target Ther ; 9(1): 144, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38853183

ABSTRACT

Respiratory syncytial virus (RSV) is the major cause of bronchiolitis and pneumonia in young children and the elderly. There are currently no approved RSV-specific therapeutic small molecules available. Using high-throughput antiviral screening, we identified an oral drug, the prenylation inhibitor lonafarnib, which showed potent inhibition of the RSV fusion process. Lonafarnib exhibited antiviral activity against both the RSV A and B genotypes and showed low cytotoxicity in HEp-2 and human primary bronchial epithelial cells (HBEC). Time-of-addition and pseudovirus assays demonstrated that lonafarnib inhibits RSV entry, but has farnesyltransferase-independent antiviral efficacy. Cryo-electron microscopy revealed that lonafarnib binds to a triple-symmetric pocket within the central cavity of the RSV F metastable pre-fusion conformation. Mutants at the RSV F sites interacting with lonafarnib showed resistance to lonafarnib but remained fully sensitive to the neutralizing monoclonal antibody palivizumab. Furthermore, lonafarnib dose-dependently reduced the replication of RSV in BALB/c mice. Collectively, lonafarnib could be a potential fusion inhibitor for RSV infection.


Subject(s)
Pyridines , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Viral Fusion Proteins , Humans , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/genetics , Pyridines/pharmacology , Mice , Animals , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/genetics , Viral Fusion Proteins/genetics , Viral Fusion Proteins/antagonists & inhibitors , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Piperidines/pharmacology , Piperidines/chemistry , Mice, Inbred BALB C , Protein Conformation , Dibenzocycloheptenes
3.
Nat Commun ; 15(1): 3422, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653965

ABSTRACT

Targeting Anaplastic lymphoma kinase (ALK) is a promising therapeutic strategy for aberrant ALK-expressing malignancies including neuroblastoma, but resistance to ALK tyrosine kinase inhibitors (ALK TKI) is a distinct possibility necessitating drug combination therapeutic approaches. Using high-throughput, genome-wide CRISPR-Cas9 knockout screens, we identify miR-1304-5p loss as a desensitizer to ALK TKIs in aberrant ALK-expressing neuroblastoma; inhibition of miR-1304-5p decreases, while mimics of this miRNA increase the sensitivity of neuroblastoma cells to ALK TKIs. We show that miR-1304-5p targets NRAS, decreasing cell viability via induction of apoptosis. It follows that the farnesyltransferase inhibitor (FTI) lonafarnib in addition to ALK TKIs act synergistically in neuroblastoma, inducing apoptosis in vitro. In particular, on combined treatment of neuroblastoma patient derived xenografts with an FTI and an ALK TKI complete regression of tumour growth is observed although tumours rapidly regrow on cessation of therapy. Overall, our data suggests that combined use of ALK TKIs and FTIs, constitutes a therapeutic approach to treat high risk neuroblastoma although prolonged therapy is likely required to prevent relapse.


Subject(s)
Anaplastic Lymphoma Kinase , Dibenzocycloheptenes , Farnesyltranstransferase , GTP Phosphohydrolases , MicroRNAs , Neuroblastoma , Piperidines , Protein Kinase Inhibitors , Pyridines , Animals , Female , Humans , Mice , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/metabolism , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Gene Expression Regulation, Neoplastic/drug effects , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/metabolism , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Xenograft Model Antitumor Assays
4.
Aging Cell ; 23(5): e14105, 2024 May.
Article in English | MEDLINE | ID: mdl-38504487

ABSTRACT

Hutchinson-Gilford Progeria syndrome (HGPS) is a severe premature ageing disorder caused by a 50 amino acid truncated (Δ50AA) and permanently farnesylated lamin A (LA) mutant called progerin. On a cellular level, progerin expression leads to heterochromatin loss, impaired nucleocytoplasmic transport, telomeric DNA damage and a permanent growth arrest called cellular senescence. Although the genetic basis for HGPS has been elucidated 20 years ago, the question whether the Δ50AA or the permanent farnesylation causes cellular defects has not been addressed. Moreover, we currently lack mechanistic insight into how the only FDA-approved progeria drug Lonafarnib, a farnesyltransferase inhibitor (FTI), ameliorates HGPS phenotypes. By expressing a variety of LA mutants using a doxycycline-inducible system, and in conjunction with FTI, we demonstrate that the permanent farnesylation, and not the Δ50AA, is solely responsible for progerin-induced cellular defects, as well as its rapid accumulation and slow clearance. Importantly, FTI does not affect clearance of progerin post-farnesylation and we demonstrate that early, but not late FTI treatment prevents HGPS phenotypes. Collectively, our study unravels the precise contributions of progerin's permanent farnesylation to its turnover and HGPS cellular phenotypes, and how FTI treatment ameliorates these. These findings are applicable to other diseases associated with permanently farnesylated proteins, such as adult-onset autosomal dominant leukodystrophy.


Subject(s)
Lamin Type A , Progeria , Lamin Type A/metabolism , Lamin Type A/genetics , Humans , Progeria/metabolism , Progeria/genetics , Progeria/pathology , Progeria/drug therapy , Farnesyltranstransferase/metabolism , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Protein Prenylation , Dibenzocycloheptenes , Piperidines , Pyridines
5.
Mol Neurodegener ; 17(1): 54, 2022 08 20.
Article in English | MEDLINE | ID: mdl-35987691

ABSTRACT

BACKGROUND: Amyloid plaque deposition and axonal degeneration are early events in AD pathogenesis. Aß disrupts microtubules in presynaptic dystrophic neurites, resulting in the accumulation of impaired endolysosomal and autophagic organelles transporting ß-site amyloid precursor protein cleaving enzyme (BACE1). Consequently, dystrophic neurites generate Aß42 and significantly contribute to plaque deposition. Farnesyltransferase inhibitors (FTIs) have recently been investigated for repositioning toward the treatment of neurodegenerative disorders and block the action of farnesyltransferase (FTase) to catalyze farnesylation, a post-translational modification that regulates proteins involved in lysosome function and microtubule stability. In postmortem AD brains, FTase and its downstream signaling are upregulated. However, the impact of FTIs on amyloid pathology and dystrophic neurites is unknown. METHODS: We tested the effects of the FTIs LNK-754 and lonafarnib in the 5XFAD mouse model of amyloid pathology. RESULTS: In 2-month-old 5XFAD mice treated chronically for 3 months, LNK-754 reduced amyloid plaque burden, tau hyperphosphorylation, and attenuated the accumulation of BACE1 and LAMP1 in dystrophic neurites. In 5-month-old 5XFAD mice treated acutely for 3 weeks, LNK-754 reduced dystrophic neurite size and LysoTracker-Green accumulation in the absence of effects on Aß deposits. Acute treatment with LNK-754 improved memory and learning deficits in hAPP/PS1 amyloid mice. In contrast to LNK-754, lonafarnib treatment was less effective at reducing plaques, tau hyperphosphorylation and dystrophic neurites, which could have resulted from reduced potency against FTase compared to LNK-754. We investigated the effects of FTIs on axonal trafficking of endolysosomal organelles and found that lonafarnib and LNK-754 enhanced retrograde axonal transport in primary neurons, indicating FTIs could support the maturation of axonal late endosomes into lysosomes. Furthermore, FTI treatment increased levels of LAMP1 in mouse primary neurons and in the brains of 5XFAD mice, demonstrating that FTIs stimulated the biogenesis of endolysosomal organelles. CONCLUSIONS: We show new data to suggest that LNK-754 promoted the axonal trafficking and function of endolysosomal compartments, which we hypothesize decreased axonal dystrophy, reduced BACE1 accumulation and inhibited amyloid deposition in 5XFAD mice. Our results agree with previous work identifying FTase as a therapeutic target for treating proteinopathies and could have important therapeutic implications in treating AD.


Subject(s)
Amyloid , Amyloidosis , Farnesyltranstransferase , Alzheimer Disease/metabolism , Amyloid/drug effects , Amyloid/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidogenic Proteins/metabolism , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Aspartic Acid Endopeptidases/metabolism , Axons/drug effects , Axons/pathology , Disease Models, Animal , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Mice , Mice, Transgenic , Plaque, Amyloid/pathology
6.
Bioorg Med Chem ; 51: 116513, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34798379

ABSTRACT

A series of new quinazolinedione derivatives have been readily synthesized and evaluated for their in vitro antiplasmodial growth inhibition activity. Most of the compounds inhibited P. falciparum FcB1 strain in the low to medium micromolar concentration. The 2-ethoxy 8ag', 2-trifluoromethoxy 8ai' and 4-fluoro-2-methoxy 8ak' showed the best inhibitory activity with EC50 values around 5 µM and were non-toxic to the primary human fibroblast cell line AB943. However, these compounds were less potent than the original hit MMV665916, which showed remarkable growth inhibition with EC50 value of 0.4 µM and presented the highest selectivity index (SI > 250). In addition, a novel approach for determining the docking poses of these quinazolinedione derivatives with their potential protein target, the P. falciparum farnesyltransferase PfFT, was investigated.


Subject(s)
Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Structure-Activity Relationship
7.
Oncogene ; 40(48): 6601-6613, 2021 12.
Article in English | MEDLINE | ID: mdl-34621019

ABSTRACT

Cancer stem cells (CSCs) are responsible for tumor progression, recurrence, and drug resistance. To identify genetic vulnerabilities of colon cancer, we performed targeted CRISPR dropout screens comprising 657 Drugbank targets and 317 epigenetic regulators on two patient-derived colon CSC-enriched spheroids. Next-generation sequencing of pooled genomic DNAs isolated from surviving cells yielded therapeutic candidates. We unraveled 44 essential genes for colon CSC-enriched spheroids propagation, including key cholesterol biosynthetic genes (HMGCR, FDPS, and GGPS1). Cholesterol biosynthesis was induced in colon cancer tissues, especially CSC-enriched spheroids. The genetic and pharmacological inhibition of HMGCR/FDPS impaired self-renewal capacity and tumorigenic potential of the spheroid models in vitro and in vivo. Mechanistically, HMGCR or FDPS depletion impaired cancer stemness characteristics by activating TGF-ß signaling, which in turn downregulated expression of inhibitors of differentiation (ID) proteins, key regulators of cancer stemness. Cholesterol and geranylgeranyl diphosphate (GGPP) rescued the growth inhibitory and signaling effect of HMGCR/FDPS blockade, implying a direct role of these metabolites in modulating stemness. Finally, cholesterol biosynthesis inhibitors and 5-FU demonstrated antitumor synergy in colon CSC-enriched spheroids, tumor organoids, and xenografts. Taken together, our study unravels novel genetic vulnerabilities of colon CSC-enriched spheroids and suggests cholesterol biosynthesis as a potential target in conjunction with traditional chemotherapy for colon cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , CRISPR-Cas Systems , Cholesterol/biosynthesis , Colonic Neoplasms/drug therapy , Dimethylallyltranstransferase/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Farnesyltranstransferase/antagonists & inhibitors , Geranyltranstransferase/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Animals , Apoptosis , Cell Proliferation , Cholesterol/chemistry , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Fluorouracil/administration & dosage , Humans , Lovastatin/administration & dosage , Male , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zoledronic Acid/administration & dosage
8.
Bioorg Med Chem ; 44: 116307, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34298413

ABSTRACT

Agents that inhibit the enzyme geranylgeranyl diphosphate synthase (GGDPS) have anti-cancer activity and our prior studies have investigated the structure-function relationship for a family of isoprenoid triazole bisphosphonates as GGDPS inhibitors. To further explore this structure-function relationship, a series of novel α-modified triazole phosphonates was prepared and evaluated for activity as GGDPS inhibitors in enzyme and cell-based assays. These studies revealed flexibility at the α position of the bisphosphonate derivatives with respect to being able to accommodate a variety of substituents without significantly affecting potency compared to the parent unsubstituted inhibitor. However, the monophosphonate derivatives lacked activity. These studies further our understanding of the structure-function relationship of the triazole-based GGDPS inhibitors and lay the foundation for future studies evaluating the impact of α-modifications on in vivo activity.


Subject(s)
Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Triazoles/pharmacology , Diphosphonates/chemical synthesis , Diphosphonates/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Humans , Molecular Structure , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
9.
Drug Res (Stuttg) ; 71(6): 341-347, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33862663

ABSTRACT

Farnesyltransferase (FTase) is a zinc enzyme that has been the subject of attention in anti-cancer research over the past. In this study, phytochemicals from Curcuma longa L., Taraxacum officinale, and Spondias mombin plants were screened for their inhibitory potentials on the human farnesyltransferase. A three-dimensional quantitative structure-activity relationship (3D-QSAR) model for the inhibition of farnesyltransferase was generated and the inhibition of farnesyltransferase by the hit, ascorbic acid was validated in an animal model of breast cancer. The lead compound, ascorbic acid makes extensive hydrogen bond interactions with key residues, lys-353, tyr-300, gly-290, leu-290 within the active site of farnesyltransferase. It downregulated the expression of FNTA mRNA in an animal model of breast cancer. The 3D-QSAR generated herein is robust, thoroughly validated, and should be employed in the pipelining of novel farnesyltransferase inhibitors. Ascorbic acid demonstrates its anticancer potentials through the inhibition of farnesyltransferase.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Ascorbic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Mammary Neoplasms, Experimental/drug therapy , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Anacardiaceae/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Ascorbic Acid/chemistry , Ascorbic Acid/therapeutic use , Carcinogens/administration & dosage , Carcinogens/toxicity , Curcuma/chemistry , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/metabolism , Female , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Rats , Taraxacum/chemistry
10.
Biochem Biophys Res Commun ; 556: 93-98, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33845310

ABSTRACT

Sepsis remains a leading cause of mortality in critically ill patients and is characterized by multi-organ dysfunction. Mitochondrial damage has been proposed to be involved in the pathophysiology of sepsis. In addition to metabolic impairments resulting from mitochondrial dysfunction, mitochondrial DNA (mtDNA) causes systemic inflammation as a damage-associated molecular pattern when it is released to the circulation. Metabolic derangements in skeletal muscle are a major complication of sepsis and negatively affects clinical outcomes of septic patients. However, limited knowledge is available about sepsis-induced mitochondrial damage in skeletal muscle. Here, we show that sepsis induced profound abnormalities in cristae structure, rupture of the inner and outer membranes and enlargement of the mitochondria in mouse skeletal muscle in a time-dependent manner, which was associated with increased plasma mtDNA levels. Farnesyltransferase inhibitor, FTI-277, prevented sepsis-induced morphological aberrations of the mitochondria, and blocked the increased plasma mtDNA levels along with improved survival. These results indicate that protein farnesylation plays a role in sepsis-induced damage of the mitochondria in mouse skeletal muscle. Our findings suggest that mitochondrial disintegrity in skeletal muscle may contribute to elevated circulating mtDNA levels in sepsis.


Subject(s)
DNA, Mitochondrial/blood , Farnesyltranstransferase/antagonists & inhibitors , Mitochondria/drug effects , Muscle, Skeletal/drug effects , Protective Agents/pharmacology , Protective Agents/therapeutic use , Sepsis/drug therapy , Animals , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mitochondria/pathology , Muscle, Skeletal/pathology , Sepsis/blood , Sepsis/pathology , Time Factors
11.
Bioorg Med Chem Lett ; 41: 127978, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33766764

ABSTRACT

A series of novel thiazole-containing amides were synthesized. A structure-activity relationship study of these compounds led to the identification of potent and selective PfFPPS/GGPPS inhibitors with good in vitro ADME profiles. The most promising candidate molecules were progressed to mouse in vivo PK studies and demonstrated adequate free drug exposure to warrant further investigation.


Subject(s)
Antimalarials/pharmacology , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Geranyltranstransferase/antagonists & inhibitors , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Diphosphonates/chemical synthesis , Diphosphonates/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Geranyltranstransferase/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Structure-Activity Relationship
12.
Pharmacol Res ; 167: 105528, 2021 05.
Article in English | MEDLINE | ID: mdl-33667685

ABSTRACT

Geranylgeranyl diphosphate synthase (GGDPS), an enzyme in the isoprenoid biosynthetic pathway (IBP), produces the isoprenoid (geranylgeranyl pyrophosphate, GGPP) used in protein geranylgeranylation reactions. Our prior studies utilizing triazole bisphosphonate-based GGDPS inhibitors (GGSIs) have revealed that these agents represent a novel strategy by which to induce cancer cell death, including multiple myeloma and pancreatic cancer. Statins inhibit the rate-limiting enzyme in the IBP and potentiate the effects of GGSIs in vitro. The in vivo effects of combination therapy with statins and GGSIs have not been determined. Here we evaluated the effects of combining VSW1198, a novel GGSI, with a statin (lovastatin or pravastatin) in CD-1 mice. Twice-weekly dosing with VSW1198 at the previously established maximally tolerated dose in combination with a statin led to hepatotoxicity, while once-weekly VSW1198-based combinations were feasible. No abnormalities in kidney, spleen, brain or skeletal muscle were observed with combination therapy. Combination therapy disrupted protein geranylgeranylation in vivo. Evaluation of hepatic isoprenoid levels revealed decreased GGPP levels in the single drug groups and undetectable GGPP levels in the combination groups. Additional studies with combinations using 50% dose-reductions of either VSW1198 or lovastatin revealed minimal hepatotoxicity with expected on-target effects of diminished GGPP levels and disruption of protein geranylgeranylation. Combination statin/GGSI therapy significantly slowed tumor growth in a myeloma xenograft model. Collectively, these studies are the first to demonstrate that combination IBP inhibitor therapy alters isoprenoid levels and disrupts protein geranylgeranylation in vivo as well as slows tumor growth in a myeloma xenograft model, thus providing the framework for future clinical exploration.


Subject(s)
Biosynthetic Pathways/drug effects , Diterpenes/administration & dosage , Drug Delivery Systems/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Protein Prenylation/drug effects , Terpenes/metabolism , Triazoles/administration & dosage , Animals , Biosynthetic Pathways/physiology , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Diterpenes/toxicity , Drug Evaluation, Preclinical/methods , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Lovastatin/administration & dosage , Lovastatin/toxicity , Mice , Mice, Inbred NOD , Mice, SCID , Pravastatin/administration & dosage , Pravastatin/toxicity , Protein Prenylation/physiology , Terpenes/antagonists & inhibitors , Triazoles/toxicity , Xenograft Model Antitumor Assays/methods
13.
Drugs ; 81(2): 283-289, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33590450

ABSTRACT

Lonafarnib (Zokinvy™) is an orally active farnesyltransferase inhibitor developed by Eiger BioPharmaceuticals under license from Merck & Co. for the treatment of hepatitis D virus (HDV) infections, and progeria and progeroid laminopathies. The drug was originally discovered by Merck & Co as an investigational drug in oncology. In progeria, lonafarnib inhibits farnesyltransferase to prevent farnesylation and subsequent accumulation of progerin and progerin-like proteins in the nucleus and cellular cytoskeleton. In November 2020, lonafarnib received its first approval in the USA to reduce the risk of mortality in Hutchinson-Gilford Progeria Syndrome (HGPS) and for the treatment of processing-deficient progeroid laminopathies (with either heterozygous LMNA mutation with progerin-like protein accumulation, or homozygous or compound heterozygous ZMPSTE24 mutations) in patients ≥ 12 months of age with a body surface area (BSA) of ≥ 0.39 m2. Lonafarnib is under regulatory review in the European Union. Clinical development for the treatment of HDV infections is underway in multiple countries. This article summarizes the milestones in the development of lonafarnib leading to this first approval.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Hepatitis D/drug therapy , Piperidines/pharmacology , Progeria/drug therapy , Pyridines/pharmacology , Antiviral Agents/chemistry , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Hepatitis D/metabolism , Humans , Piperidines/chemistry , Progeria/metabolism , Pyridines/chemistry
14.
Cell ; 184(2): 293, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33482093

ABSTRACT

The ultra-rare, pediatric premature aging disorder Hutchinson-Gilford progeria syndrome (HGPS) is caused by mutation of LMNA, encoding the nuclear architectural protein lamin A. Patients develop atherosclerosis and typically die of heart failure in their teens. FDA-approved Zokinvy prevents farnesylation of lamin A, reduces vascular stiffness, and extends survival in HGPS patients. To view this Bench to Bedside, open or download the PDF.


Subject(s)
Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Progeria/drug therapy , Progeria/enzymology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/metabolism , Humans , Molecular Targeted Therapy
15.
Trends Cancer ; 7(6): 525-540, 2021 06.
Article in English | MEDLINE | ID: mdl-33358111

ABSTRACT

The mevalonate synthesis inhibitors, statins, are mainstay therapeutics for cholesterol management and cardiovascular health. Thirty years of research have uncovered supportive roles for the mevalonate pathway in numerous cellular processes that support oncogenesis, most recently macropinocytosis. Central to the diverse mechanisms of statin sensitivity is an acquired dependence on one mevalonate pathway output, protein geranylgeranylation. New chemical prenylation probes and the discovery of a novel geranylgeranyl transferase hold promise to deepen our understanding of statin mechanisms of action. Further, insights into statin selection and the counterproductive role of dietary geranylgeraniol highlight how we should assess statins in the clinic. Lastly, rational combination strategies preview how statins will enter the oncology toolbox.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/metabolism , Neoplasms/drug therapy , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Diterpenes/administration & dosage , Diterpenes/adverse effects , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Feeding Behavior , Food-Drug Interactions , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Metabolic Networks and Pathways/drug effects , Mice , Neoplasms/metabolism , Neoplasms/pathology , Pinocytosis/drug effects , Polyisoprenyl Phosphates/metabolism , Prenylation/drug effects
16.
Org Biomol Chem ; 19(10): 2213-2223, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33349821

ABSTRACT

Photoactivatable protecting groups (PPGs) are useful for a broad range of applications ranging from biology to materials science. In chemical biology, induction of biological processes via photoactivation is a powerful strategy for achieving spatiotemporal control. The importance of cysteine, glutathione, and other bioactive thiols in regulating protein structure/activity and cell redox homeostasis makes modulation of thiol activity particularly useful. One major objective for enhancing the utility of photoactivatable protecting groups (PPGs) in living systems is creating PPGs with longer wavelength absorption maxima and efficient two-photon (TP) absorption. Toward these objectives, we developed a carboxyl- and dimethylamine-functionalized nitrodibenzofuran PPG scaffold (cDMA-NDBF) for thiol photoactivation, which has a bathochromic shift in the one-photon absorption maximum from λmax = 315 nm with the unfunctionalized NDBF scaffold to λmax = 445 nm. While cDMA-NDBF-protected thiols are stable in the presence of UV irradiation, they undergo efficient broad-spectrum TP photolysis at wavelengths as long as 900 nm. To demonstrate the wavelength orthogonality of cDMA-NDBF and NDBF photolysis in a biological setting, caged farnesyltransferase enzyme inhibitors (FTI) were prepared and selectively photoactivated in live cells using 850-900 nm TP light for cDMA-NDBF-FTI and 300 nm UV light for NDBF-FTI. These experiments represent the first demonstration of thiol photoactivation at wavelengths above 800 nm. Consequently, cDMA-NDBF-caged thiols should have broad applicability in a wide range of experiments in chemical biology and materials science.


Subject(s)
Benzofurans/chemistry , Enzyme Inhibitors/pharmacology , Sulfhydryl Compounds/pharmacology , Animals , Benzofurans/chemical synthesis , Benzofurans/radiation effects , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/radiation effects , Farnesyltranstransferase/antagonists & inhibitors , Infrared Rays , Madin Darby Canine Kidney Cells , Photolysis/radiation effects , Photons , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/radiation effects
17.
Br J Cancer ; 123(12): 1749-1756, 2020 12.
Article in English | MEDLINE | ID: mdl-32968206

ABSTRACT

BACKGROUND: Multi-targeted tyrosine kinase inhibitors (TKIs) are the standard of care for patients with advanced clear cell renal cell carcinoma (ccRCC). However, a significant number of ccRCC patients are primarily refractory to targeted therapeutics, showing neither disease stabilisation nor clinical benefits. METHODS: We used CRISPR/Cas9-based high-throughput loss of function (LOF) screening to identify cellular factors involved in the resistance to sunitinib. Next, we validated druggable molecular factors that are synthetically lethal with sunitinib treatment using cell and animal models of ccRCC. RESULTS: Our screening identified farnesyltransferase among the top hits contributing to sunitinib resistance in ccRCC. Combined treatment with farnesyltransferase inhibitor lonafarnib potently augmented the anti-tumour efficacy of sunitinib both in vitro and in vivo. CONCLUSION: CRISPR/Cas9 LOF screening presents a promising approach to identify and target cellular factors involved in the resistance to anti-cancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Drug Resistance, Neoplasm/genetics , Farnesyltranstransferase/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Piperidines/pharmacology , Pyridines/pharmacology , Sunitinib/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis , CRISPR-Cas Systems , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , DNA Fragmentation , Drug Interactions , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lysosomes , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Molecular Targeted Therapy , Neoplasm Transplantation , Progression-Free Survival , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering , Random Allocation , Sunitinib/pharmacokinetics
18.
Bioorg Chem ; 103: 104184, 2020 10.
Article in English | MEDLINE | ID: mdl-32891861

ABSTRACT

In the incessant search for innovative cancer control strategies, this study was devoted to the design, synthesis and pharmacological evaluation of dual inhibitors of farnesyltransferase and tubulin polymerization (FTI/MTIs). A series of indolizine-phenothiazine hybrids 16 (amides) and 17 (ketones) has been obtained in a 4-step procedure. The combination of the two heterocycles provided potent tubulin polymerization inhibitors with similar efficiency as the reference phenstatin and (-)-desoxypodophyllotoxin. Ketones 17 were also able to inhibit human farnesyltransferase (FTase) in vitro. Interestingly, three molecules 17c, 17d and 17f were very effective against both considered biological targets. Next, nine indolizine-phenothiazine hybrids 16c, 16f, 17a-f and 22b were evaluated for their cell growth inhibition potential on the NCI-60 cancer cell lines panel. Ketones 17a-f were the most active and displayed promising cellular activities. Not only they arrested the cell growth of almost all tested cancer cells, but they displayed cytotoxicity potential with GI50 values in the low nanomolar range. The most sensitive cell lines upon treatment with indolizine-phenothiazine hybrids were NCI-H522 (lung cancer), COLO-205 and HT29 (colon cancer), SF-539 (human glioblastoma), OVCAR-3 (ovarian cancer), A498 (renal cancer) and especially MDA-MB-435 (melanoma). Demonstrating the preclinical effectiveness of these dual inhibitors can be crucial. A single dual molecule could induce a synergy of antitumor activity, while increasing the effectiveness and reducing the toxicity of the classical combo treatments currently used in chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Indolizines/pharmacology , Phenothiazines/pharmacology , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Farnesyltranstransferase/chemistry , Farnesyltranstransferase/metabolism , Humans , Indolizines/chemical synthesis , Indolizines/metabolism , Molecular Docking Simulation , Molecular Structure , Phenothiazines/chemical synthesis , Phenothiazines/metabolism , Protein Binding , Structure-Activity Relationship , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/metabolism
19.
Bioorg Med Chem ; 28(16): 115604, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32690260

ABSTRACT

Geranylgeranyl diphosphate synthase (GGDPS) inhibitors are of potential therapeutic interest as a consequence of their activity against the bone marrow cancer multiple myeloma. A series of bisphosphonates linked to an isoprenoid tail through an amide linkage has been prepared and tested for the ability to inhibit GGDPS in enzyme and cell-based assays. The amides were designed as analogues to triazole-based GGDPS inhibitors. Several of the new compounds show GGDPS inhibitory activity in both enzyme and cell assays, with potency dependent on chain length and olefin stereochemistry.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Triazoles/chemistry , Triazoles/pharmacology , Amides/chemistry , Amides/pharmacology , Cell Line , Diphosphonates/chemistry , Diphosphonates/pharmacology , Farnesyltranstransferase/metabolism , Humans , Models, Molecular , Structure-Activity Relationship , Terpenes/chemistry , Terpenes/pharmacology
20.
Development ; 147(12)2020 06 24.
Article in English | MEDLINE | ID: mdl-32467241

ABSTRACT

Tendons and ligaments are crucial components of the musculoskeletal system, yet the pathways specifying these fates remain poorly defined. Through a screen of known bioactive chemicals in zebrafish, we identified a new pathway regulating tendon cell induction. We established that statin, through inhibition of the mevalonate pathway, causes an expansion of the tendon progenitor population. Co-expression and live imaging studies indicate that the expansion does not involve an increase in cell proliferation, but rather results from re-specification of cells from the neural crest-derived sox9a+/sox10+ skeletal lineage. The effect on tendon cell expansion is specific to the geranylgeranylation branch of the mevalonate pathway and is mediated by inhibition of Rac activity. This work establishes a novel role for the mevalonate pathway and Rac activity in regulating specification of the tendon lineage.


Subject(s)
Mevalonic Acid/metabolism , Tendons/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Animals, Genetically Modified/metabolism , Atorvastatin/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Morpholinos/metabolism , Neural Crest/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Tendons/cytology , Tendons/pathology , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...