Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(3): 101471, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38508142

ABSTRACT

Drug-tolerant persisters (DTPs) are a rare subpopulation of cells within a tumor that can survive therapy through nongenetic adaptive mechanisms to develop relapse and repopulate the tumor following drug withdrawal. Using a cancer cell line with an engineered suicide switch to kill proliferating cells, we perform both genetic screens and compound screens to identify the inhibition of bromodomain and extraterminal domain (BET) proteins as a selective vulnerability of DTPs. BET inhibitors are especially detrimental to DTPs that have reentered the cell cycle (DTEPs) in a broad spectrum of cancer types. Mechanistically, BET inhibition induces lethal levels of ROS through the suppression of redox-regulating genes highly expressed in DTPs, including GPX2, ALDH3A1, and MGST1. In vivo BET inhibitor treatment delays tumor relapse in both melanoma and lung cancer. Our study suggests that combining standard of care therapy with BET inhibitors to eliminate residual persister cells is a promising therapeutic strategy.


Subject(s)
Lung Neoplasms , Neoplasm Recurrence, Local , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics
2.
Nat Cancer ; 3(11): 1284-1299, 2022 11.
Article in English | MEDLINE | ID: mdl-36414711

ABSTRACT

Senolytics, drugs that kill senescent cells, have been proposed to improve the response to pro-senescence cancer therapies; however, this remains challenging due to a lack of broadly acting senolytic drugs. Using CRISPR/Cas9-based genetic screens in different senescent cancer cell models, we identify loss of the death receptor inhibitor cFLIP as a common vulnerability of senescent cancer cells. Senescent cells are primed for apoptotic death by NF-κB-mediated upregulation of death receptor 5 (DR5) and its ligand TRAIL, but are protected from death by increased cFLIP expression. Activation of DR5 signaling by agonistic antibody, which can be enhanced further by suppression of cFLIP by BRD2 inhibition, leads to efficient killing of a variety of senescent cancer cells. Moreover, senescent cells sensitize adjacent non-senescent cells to killing by DR5 agonist through a bystander effect mediated by secretion of cytokines. We validate this 'one-two punch' cancer therapy by combining pro-senescence therapy with DR5 activation in different animal models.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein , Neoplasms , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Apoptosis , NF-kappa B/metabolism , Signal Transduction , Neoplasms/drug therapy
3.
PLoS One ; 17(9): e0273182, 2022.
Article in English | MEDLINE | ID: mdl-36067171

ABSTRACT

Inducing senescence in cancer cells is emerging as a new therapeutic strategy. In order to find ways to enhance senescence induction by palbociclib, a CDK4/6 inhibitor approved for treatment of metastatic breast cancer, we performed functional genetic screens in palbociclib-resistant cells. Using this approach, we found that loss of CDK2 results in strong senescence induction in palbociclib-treated cells. Treatment with the CDK2 inhibitor indisulam, which phenocopies genetic CDK2 inactivation, led to sustained senescence induction when combined with palbociclib in various cell lines and lung cancer xenografts. Treating cells with indisulam led to downregulation of cyclin H, which prevented CDK2 activation. Combined treatment with palbociclib and indisulam induced a senescence program and sensitized cells to senolytic therapy. Our data indicate that inhibition of CDK2 through indisulam treatment can enhance senescence induction by CDK4/6 inhibition.


Subject(s)
Cyclin-Dependent Kinase 6 , Protein Kinase Inhibitors , Cell Line, Tumor , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Humans , Piperazines , Protein Kinase Inhibitors/pharmacology , Pyridines , Sulfonamides
4.
Life Sci Alliance ; 5(9)2022 09.
Article in English | MEDLINE | ID: mdl-35534224

ABSTRACT

Discovering biomarkers of drug response and finding powerful drug combinations can support the reuse of previously abandoned cancer drugs in the clinic. Indisulam is an abandoned drug that acts as a molecular glue, inducing degradation of splicing factor RBM39 through interaction with CRL4DCAF15 Here, we performed genetic and compound screens to uncover factors mediating indisulam sensitivity and resistance. First, a dropout CRISPR screen identified SRPK1 loss as a synthetic lethal interaction with indisulam that can be exploited therapeutically by the SRPK1 inhibitor SPHINX31. Moreover, a CRISPR resistance screen identified components of the degradation complex that mediate resistance to indisulam: DCAF15, DDA1, and CAND1. Last, we show that cancer cells readily acquire spontaneous resistance to indisulam. Upon acquiring indisulam resistance, pancreatic cancer (Panc10.05) cells still degrade RBM39 and are vulnerable to BCL-xL inhibition. The better understanding of the factors that influence the response to indisulam can assist rational reuse of this drug in the clinic.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Intracellular Signaling Peptides and Proteins , Neoplasms/drug therapy , Neoplasms/genetics , RNA Splicing Factors , Sulfonamides/pharmacology
5.
Nature ; 595(7869): 730-734, 2021 07.
Article in English | MEDLINE | ID: mdl-34290403

ABSTRACT

Hepatocellular carcinoma (HCC)-the most common form of liver cancer-is an aggressive malignancy with few effective treatment options1. Lenvatinib is a small-molecule inhibitor of multiple receptor tyrosine kinases that is used for the treatment of patients with advanced HCC, but this drug has only limited clinical benefit2. Here, using a kinome-centred CRISPR-Cas9 genetic screen, we show that inhibition of epidermal growth factor receptor (EGFR) is synthetic lethal with lenvatinib in liver cancer. The combination of the EGFR inhibitor gefitinib and lenvatinib displays potent anti-proliferative effects in vitro in liver cancer cell lines that express EGFR and in vivo in xenografted liver cancer cell lines, immunocompetent mouse models and patient-derived HCC tumours in mice. Mechanistically, inhibition of fibroblast growth factor receptor (FGFR)  by lenvatinib treatment leads to feedback activation of the EGFR-PAK2-ERK5 signalling axis, which is blocked by EGFR inhibition. Treatment of 12 patients with advanced HCC who were unresponsive to lenvatinib treatment with the combination of lenvatinib plus gefitinib (trial identifier NCT04642547) resulted in meaningful clinical responses. The combination therapy identified here may represent a promising strategy for the approximately 50% of patients with advanced HCC who have high levels of EGFR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gefitinib/pharmacology , Humans , Liver Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Fibroblast Growth Factor , Signal Transduction , Xenograft Model Antitumor Assays
6.
Cell Rep ; 36(4): 109441, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34320349

ABSTRACT

Cellular senescence is characterized as a stable proliferation arrest that can be triggered by multiple stresses. Most knowledge about senescent cells is obtained from studies in primary cells. However, senescence features may be different in cancer cells, since the pathways that are involved in senescence induction are often deregulated in cancer. We report here a comprehensive analysis of the transcriptome and senolytic responses in a panel of 13 cancer cell lines rendered senescent by two distinct compounds. We show that in cancer cells, the response to senolytic agents and the composition of the senescence-associated secretory phenotype are more influenced by the cell of origin than by the senescence trigger. Using machine learning, we establish the SENCAN gene expression classifier for the detection of senescence in cancer cell samples. The expression profiles and senescence classifier are available as an interactive online Cancer SENESCopedia.


Subject(s)
Cellular Senescence , Neoplasms/pathology , Aniline Compounds/pharmacology , Azepines/pharmacology , Cell Line, Tumor , Cellular Senescence/drug effects , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/genetics , Pyrimidines/pharmacology , Reproducibility of Results , Senescence-Associated Secretory Phenotype/drug effects , Senescence-Associated Secretory Phenotype/genetics , Senotherapeutics/pharmacology , Sulfonamides/pharmacology
7.
Mol Cancer Res ; 19(10): 1613-1621, 2021 10.
Article in English | MEDLINE | ID: mdl-34158393

ABSTRACT

Pro-senescence therapies are increasingly being considered for the treatment of cancer. Identifying additional targets to induce senescence in cancer cells could further enable such therapies. However, screening for targets whose suppression induces senescence on a genome-wide scale is challenging, as senescent cells become growth arrested, and senescence-associated features can take 1 to 2 weeks to develop. For a screen with a whole-genome CRISPR library, this would result in billions of undesirable proliferating cells by the time the senescent features emerge in the growth arrested cells. Here, we present a suicide switch system that allows genome-wide CRISPR screening in growth-arrested subpopulations by eliminating the proliferating cells during the screen through activation of a suicide switch in proliferating cells. Using this system, we identify in a genome-scale CRISPR screen several autophagy-related proteins as targets for senescence induction. We show that inhibiting macroautophagy with a small molecule ULK1 inhibitor can induce senescence in cancer cell lines of different origin. Finally, we show that combining ULK1 inhibition with the senolytic drug ABT-263 leads to apoptosis in a panel of cancer cell lines. IMPLICATIONS: Our suicide switch approach allows for genome-scale identification of pro-senescence targets, and can be adapted to simplify other screens depending on the nature of the promoter used to drive the switch.


Subject(s)
Autophagy-Related Proteins/genetics , Autophagy/genetics , CRISPR-Cas Systems/genetics , Cellular Senescence/genetics , A549 Cells , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/genetics , CRISPR-Cas Systems/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cellular Senescence/drug effects , HEK293 Cells , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Small Molecule Libraries/pharmacology
8.
Elife ; 92020 10 05.
Article in English | MEDLINE | ID: mdl-33016874

ABSTRACT

The dependency of cancer cells on glutamine may be exploited therapeutically as a new strategy for treating cancers that lack druggable driver genes. Here we found that human liver cancer was dependent on extracellular glutamine. However, targeting glutamine addiction using the glutaminase inhibitor CB-839 as monotherapy had a very limited anticancer effect, even against the most glutamine addicted human liver cancer cells. Using a chemical library, we identified V-9302, a novel inhibitor of glutamine transporter ASCT2, as sensitizing glutamine dependent (GD) cells to CB-839 treatment. Mechanically, a combination of CB-839 and V-9302 depleted glutathione and induced reactive oxygen species (ROS), resulting in apoptosis of GD cells. Moreover, this combination also showed tumor inhibition in HCC xenograft mouse models in vivo. Our findings indicate that dual inhibition of glutamine metabolism by targeting both glutaminase and glutamine transporter ASCT2 represents a potential novel treatment strategy for glutamine addicted liver cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Glutamine/metabolism , Liver Neoplasms/metabolism , Amino Acid Transport System ASC/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzeneacetamides/pharmacology , Carrier Proteins/antagonists & inhibitors , Cell Line, Tumor , Drug Synergism , Glutaminase/antagonists & inhibitors , Humans , Mice , Minor Histocompatibility Antigens , Reactive Oxygen Species/metabolism , Thiadiazoles/pharmacology , Xenograft Model Antitumor Assays
9.
Nature ; 574(7777): 268-272, 2019 10.
Article in English | MEDLINE | ID: mdl-31578521

ABSTRACT

Liver cancer remains difficult to treat, owing to a paucity of drugs that target critical dependencies1,2; broad-spectrum kinase inhibitors such as sorafenib provide only a modest benefit to patients with hepatocellular carcinoma3. The induction of senescence may represent a strategy for the treatment of cancer, especially when combined with a second drug that selectively eliminates senescent cancer cells (senolysis)4,5. Here, using a kinome-focused genetic screen, we show that pharmacological inhibition of the DNA-replication kinase CDC7 induces senescence selectively in liver cancer cells with mutations in TP53. A follow-up chemical screen identified the antidepressant sertraline as an agent that kills hepatocellular carcinoma cells that have been rendered senescent by inhibition of CDC7. Sertraline suppressed mTOR signalling, and selective drugs that target this pathway were highly effective in causing the apoptotic cell death of hepatocellular carcinoma cells treated with a CDC7 inhibitor. The feedback reactivation of mTOR signalling after its inhibition6 is blocked in cells that have been treated with a CDC7 inhibitor, which leads to the sustained inhibition of mTOR and cell death. Using multiple in vivo mouse models of liver cancer, we show that treatment with combined inhibition of of CDC7 and mTOR results in a marked reduction of tumour growth. Our data indicate that exploiting an induced vulnerability could be an effective treatment for liver cancer.


Subject(s)
Apoptosis/drug effects , Cellular Senescence/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Molecular Targeted Therapy , Sertraline/pharmacology , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Female , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sertraline/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
10.
Cell Rep ; 21(3): 773-783, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29045843

ABSTRACT

Senescence is a proliferation arrest that can result from a variety of stresses. Cancer cells can also undergo senescence, but the stresses that provoke cancer cells to undergo senescence are unclear. Here, we use both functional genetic and compound screens in cancer cells harboring a reporter that is activated during senescence to find targets that induce senescence. We show that suppression of the SWI/SNF component SMARCB1 induces senescence in melanoma through strong activation of the MAP kinase pathway. From the compound screen, we identified multiple aurora kinase inhibitors as potent inducers of senescence in RAS mutant lung cancer. Senescent melanoma and lung cancer cells acquire sensitivity to the BCL2 family inhibitor ABT263. We propose a one-two punch approach for the treatment of cancer in which a drug is first used to induce senescence in cancer cells and a second drug is then used to kill senescent cancer cells.


Subject(s)
Cellular Senescence/genetics , Genetic Testing , High-Throughput Screening Assays , Neoplasms/genetics , Neoplasms/pathology , Aurora Kinases/antagonists & inhibitors , Aurora Kinases/metabolism , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cellular Senescence/drug effects , Down-Regulation/genetics , ErbB Receptors/metabolism , Gene Knockout Techniques , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , Melanoma/genetics , Melanoma/pathology , Oncogenes , Protein Kinase Inhibitors/pharmacology , SMARCB1 Protein/genetics , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...