scholarly journals Targeting autophagy to overcome drug resistance: further developments

2020 ◽  
Vol 13 (1) ◽  
Author(s):  
Haocai Chang ◽  
Zhengzhi Zou

AbstractInhibiting cell survival and inducing cell death are the main approaches of tumor therapy. Autophagy plays an important role on intracellular metabolic homeostasis by eliminating dysfunctional or unnecessary proteins and damaged or aged cellular organelles to recycle their constituent metabolites that enable the maintenance of cell survival and genetic stability and even promotes the drug resistance, which severely limits the efficacy of chemotherapeutic drugs. Currently, targeting autophagy has a seemingly contradictory effect to suppress and promote tumor survival, which makes the effect of targeting autophagy on drug resistance more confusing and fuzzier. In the review, we summarize the regulation of autophagy by emerging ways, the action of targeting autophagy on drug resistance and some of the new therapeutic approaches to treat tumor drug resistance by interfering with autophagy-related pathways. The full-scale understanding of the tumor-associated signaling pathways and physiological functions of autophagy will hopefully open new possibilities for the treatment of tumor drug resistance and the improvement in clinical outcomes.

F1000Research ◽  
2016 ◽  
Vol 5 ◽  
pp. 2786 ◽  
Author(s):  
Daniel A. Vorobiof

Over the past few decades, the systemic therapy of breast cancer (early and advanced) has changed considerably. For the past 40–50 years, and since the discovery and further therapeutic use of tamoxifen, a selective estrogen receptor modulator, breast cancer treatment has become the model for the development and success of tailored medical treatment. Much still needs to be done in improving outcomes for all patients with breast cancer, and especially for those who have advanced breast cancer, a challenging area for medical oncologists. Ongoing international clinical trials are currently evaluating new therapeutic approaches and identifying specific biological subsets that could determine a patient’s ability to respond to particular chemotherapeutic drugs.


2009 ◽  
Vol 147 (4) ◽  
pp. 421-423
Author(s):  
T. A. Rajewskaya ◽  
S. A. Goncharova ◽  
N. P. Konovalova ◽  
A. A. Terent’ev ◽  
M. A. Lapshina

2017 ◽  
Vol 127 (6) ◽  
pp. 1219-1230 ◽  
Author(s):  
Khadijeh Bijangi-Vishehsaraei ◽  
M. Reza Saadatzadeh ◽  
Haiyan Wang ◽  
Angie Nguyen ◽  
Malgorzata M. Kamocka ◽  
...  

OBJECTIVEDefects in the apoptotic machinery and augmented survival signals contribute to drug resistance in glioblastoma (GBM). Moreover, another complexity related to GBM treatment is the concept that GBM development and recurrence may arise from the expression of GBM stem cells (GSCs). Therefore, the use of a multifaceted approach or multitargeted agents that affect specific tumor cell characteristics will likely be necessary to successfully eradicate GBM. The objective of this study was to investigate the usefulness of sulforaphane (SFN)—a constituent of cruciferous vegetables with a multitargeted effect—as a therapeutic agent for GBM.METHODSThe inhibitory effects of SFN on established cell lines, early primary cultures, CD133-positive GSCs, GSC-derived spheroids, and GBM xenografts were evaluated using various methods, including GSC isolation and the sphere-forming assay, analysis of reactive oxygen species (ROS) and apoptosis, cell growth inhibition assay, comet assays for assessing SFN-triggered DNA damage, confocal microscopy, Western blot analysis, and the determination of in vivo efficacy as assessed in human GBM xenograft models.RESULTSSFN triggered the significant inhibition of cell survival and induced apoptotic cell death, which was associated with caspase 3 and caspase 7 activation. Moreover, SFN triggered the formation of mitochondrial ROS, and SFN-triggered cell death was ROS dependent. Comet assays revealed that SFN increased single- and double-strand DNA breaks in GBM. Compared with the vehicle control cells, a significantly higher amount of γ-H2AX foci correlated with an increase in DNA double-strand breaks in the SFN-treated samples. Furthermore, SFN robustly inhibited the growth of GBM cell–induced cell death in established cell cultures and early-passage primary cultures and, most importantly, was effective in eliminating GSCs, which play a major role in drug resistance and disease recurrence. In vivo studies revealed that SFN administration at 100 mg/kg for 5-day cycles repeated for 3 weeks significantly decreased the growth of ectopic xenografts that were established from the early passage of primary cultures of GBM10.CONCLUSIONSThese results suggest that SFN is a potent anti-GBM agent that targets several apoptosis and cell survival pathways and further preclinical and clinical studies may prove that SFN alone or in combination with other therapies may be potentially useful for GBM therapy.


2003 ◽  
Vol 14 (5) ◽  
pp. 2071-2087 ◽  
Author(s):  
Karl Deacon ◽  
Pratibha Mistry ◽  
Jonathan Chernoff ◽  
Jonathan L. Blank ◽  
Rajnikant Patel

Activation of the mitotic checkpoint by chemotherapeutic drugs such as taxol causes mammalian cells to arrest in mitosis and then undergo apoptosis. However, the biochemical basis of chemotherapeutic drug-induced cell death is unclear. Herein, we provide new evidence that both cell survival and cell death-signaling pathways are concomitantly activated during mitotic arrest by microtubule-interfering drugs. Treatment of HeLa cells with chemotherapeutic drugs activated both p38 mitogen-activated protein kinase (MAPK) and p21-activated kinase (PAK). p38 MAPK was necessary for chemotherapeutic drug-induced cell death because the p38 MAPK inhibitors SB203580 or SB202190 suppressed cell death. Dominant-active MKK6, a direct activator of p38 MAPK, also induced cell death by stimulating translocation of Bax from the cytosol to the mitochondria in a p38 MAPK-dependent manner. Dominant active PAK suppressed this MKK6-induced cell death. PAK seems to mediate cell survival by phosphorylating Bad, and inhibition of PAK in mitotically arrested cells reduced Bad phosphorylation and increased apoptosis. Our results suggest that therapeutic strategies that suppress PAK-mediated survival signals may improve the efficacy of current cancer chemotherapies by enhancing p38 MAPK-mediated cell death.


F1000Research ◽  
2019 ◽  
Vol 8 ◽  
pp. 1793 ◽  
Author(s):  
Andrew Gilmore ◽  
Louise King

Apoptosis is a highly conserved programme for removing damaged and unwanted cells. Apoptosis in most cells is coordinated on mitochondria by the Bcl-2 family of proteins. The balance between pro- and anti-apoptotic Bcl-2 family proteins sets a threshold for mitochondrial apoptosis, a balance that is altered during cancer progression. Consequently, avoidance of cell death is an established cancer hallmark. Although there is a general perception that tumour cells are more resistant to apoptosis than their normal counterparts, the realities of cell death regulation in cancer are more nuanced. In this review we discuss how a profound understanding of this control has led to new therapeutic approaches, including the new class of BH3-mimetics, which directly target apoptosis as a vulnerability in cancer. We discuss recent findings that highlight the current limitations in our understanding of apoptosis and how these novel therapeutics work.


Blood ◽  
1993 ◽  
Vol 81 (1) ◽  
pp. 151-157 ◽  
Author(s):  
T Miyashita ◽  
JC Reed

Previous studies have shown that the bcl-2 gene encodes a mitochondrial protein that contributes to neoplastic cell expansion primarily by promoting cell survival through interference with “programmed cell death” (PCD), also termed “apoptosis.” Because many chemotherapeutic drugs are capable of initiating pathways leading to apoptosis, we determined whether deregulated bcl-2 expression could render cells resistant to several drugs commonly used in the treatment of non- Hodgkin's lymphomas, including dexamethasone (DEX), methotrexate (MTX), 1-beta-D-arabinofuranosyl-cytosine (Ara-C), etoposide (VP-16), vincristine (VC), cisplatin (CP), and hydroperoxycyclophosphamide (4- HC). For these experiments, we achieved high levels of p26-Bcl-2 protein production in a human pre-B-cell leukemia line 697 by stable infection with a recombinant bcl-2-containing retrovirus and then compared these cells with control virus-infected 697 cells. Control 697 cells were induced to undergo apoptosis by all drugs tested as defined by DNA degradation into oligonucleosomal-length fragments, cell shrinkage, and subsequent cell death. In contrast, 697 cells with elevated Bcl-2 protein levels exhibited strikingly prolonged cell survival and markedly reduced DNA fragmentation when cultured in the presence of these antineoplastic agents. Although high levels of Bcl-2 protein protected 697 cells from the acute cytotoxic effects of DEX and the other drugs tested, Bcl-2 did not prevent these drugs from suppressing the proliferation of 697 cells. However, when 697 cells were treated with DEX or MTX for 3 days, then washed and cultured in semisolid media without drugs, bcl-2-virus-infected cells gave rise to colonies at much higher frequencies than 697 cells stably infected with control virus. These results indicate that by protecting 697 leukemic cells from the acute cytotoxicity of DEX and some other chemotherapeutic drugs, high levels of p26-Bcl-2 can create the opportunity for re-initiation of cell growth when drugs are withdrawn. The findings may be relevant to clinical correlative studies of non- Hodgkin's lymphoma patients that have found an association between worse prognosis and bcl-2 gene rearrangements or t[14;18] translocations.


2021 ◽  
Vol 22 (22) ◽  
pp. 12586
Author(s):  
Viviana De Martino ◽  
Michela Rossi ◽  
Giulia Battafarano ◽  
Jessica Pepe ◽  
Salvatore Minisola ◽  
...  

Osteosarcoma (OS) is a skeletal tumor affecting mainly children and adolescents. The presence of distance metastasis is frequent and it is localized preferentially to the lung, representing the main reason for death among patients. The therapeutic approaches are based on surgery and chemotherapeutics. However, the drug resistance and the side effects associated with the chemotherapy require the identification of new therapeutic approaches. The understanding of the complex biological scenario of the osteosarcoma will open the way for the identification of new targets for its treatment. Recently, a great interest of scientific community is for extracellular vesicles (EVs), that are released in the tumor microenvironment and are important regulators of tumor proliferation and the metastatic process. At the same time, circulating extracellular vesicles can be exploited as diagnostic and prognostic biomarkers, and they can be loaded with drugs as a new therapeutic approach for osteosarcoma patients. Thus, the characterization of OS-related EVs could represent a way to convert these vesicles from antagonists for human health into therapeutic and/or diagnostic agents.


2019 ◽  
Vol 20 (1) ◽  
Author(s):  
Yu-chi Shen ◽  
Caroline Arellano-Garcia ◽  
Rosa E. Menjivar ◽  
Ethan M. Jewett ◽  
Wolfgang Dohle ◽  
...  

Abstract Background Neurofibromatosis 1 and 2, although involving two different tumour suppressor genes (neurofibromin and merlin, respectively), are both cancer predisposition syndromes that disproportionately affect cells of neural crest origin. New therapeutic approaches for both NF1 and NF2 are badly needed. In promising previous work we demonstrated that two non-steroidal analogues of 2-methoxy-oestradiol (2ME2), STX3451(2-(3-bromo-4,5-dimethoxybenzyl)-7-methoxy-6-sulfamoyloxy-1,2,3,4-tetrahydroisoquinoline), and STX2895 (7-Ethyl-6-sulfamoyloxy-2-(3,4,5-trimethoxybenzyl)-1,2,3,4-tetrahydroisoquinoline) reduced tumour cell growth and induced apoptosis in malignant and benign human Neurofibromatosis 1 (NF1) tumour cells. In earlier NF1 mechanism of action studies we found that in addition to their effects on non-classical hormone-sensitive pathways, STX agents acted on the actin- and myosin-cytoskeleton, as well as PI3Kinase and MTOR signaling pathways. Tumour growth in NF2 cells is affected by different inhibitors from those affecting NF1 growth pathways: specifically, NF2 cells are affected by merlin-downstream pathway inhibitors. Because Merlin, the affected tumour suppressor gene in NF2, is also known to be involved in stabilizing membrane-cytoskeletal complexes, as well as in cell proliferation, and apoptosis, we looked for potentially common mechanisms of action in the agents’ effects on NF1 and NF2. We set out to determine whether STX agents could therefore also provide a prospective avenue for treatment of NF2. Methods STX3451 and STX2895 were tested in dose-dependent studies for their effects on growth parameters of malignant and benign NF2 human tumour cell lines in vitro. The mechanisms of action of STX3451 and STX2895 were also analysed. Results Although neither of the agents tested affected cell growth or apoptosis in the NF2 tumour cell lines tested through the same mechanisms by which they affect these parameters in NF1 tumour cell lines, both agents disrupted actin- and myosin-based cytoskeletal structures in NF2 cell lines, with subsequent effects on growth and cell death. Conclusions Both STX3451 and STX2895 provide new approaches for inducing cell death and lowering tumour burden in NF2 as well as in NF1, which both have limited treatment options.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 691-691
Author(s):  
Emilia Mahoney ◽  
Amy J. Johnson ◽  
Amy J. Wagner ◽  
Joshua Hessler ◽  
Sneha V. Gupta ◽  
...  

Abstract Abstract 691 Autophagy is an active process of intracellular degradation that occurs via sequestration of intracytoplasmic components (proteins, organelles) inside double membrane vesicles termed “autophagosomes”, followed by enzymatic degradation upon fusion with lysosomes. Autophagy is an adaptive process induced by cellular starvation but also contributes to development, aging, and pathogenesis of infections, neurodegenerative disease, atherosclerosis and cancer. In cancer, autophagy can function either as a cell survival signal or death response (i.e. type II cell death). To date, autophagy has not been characterized in chronic lymphocytic leukemia (CLL). The aim of our study is to investigate the presence and the role of autophagy in CLL and the potential contribution it may have to cell death or drug resistance with common therapeutics used in CLL. Our initial studies demonstrated that the essential cellular machinery for autophagy was present. CLL cells have a basal low level of autophagy when cultured in standard media that is actively promoted by cellular starvation or treatment with rapamycin. Given the potential contribution of autophagy in the death process or mechanism of drug resistance, we examined the influence of different therapeutics currently approved for CLL including chlorambucil, fludarabine, rituximab and dexamethasone. Cells were exposed to the drugs for four hours and examined by immunofluorescent staining of endogenous LC3 protein, an established marker of autophagy that becomes conjugated with phosphatydylethanolamine and recruited to the autophagosome membrane. Similar to rapamycin, fludarabine promotes autophagosome accumulation in all CLL samples tested, whereas dexamethasone caused this effect only in a subset. In contrast, no autophagosome accumulation was observed with chlorambucil or rituximab plus cross-linking.To determine the influence of this autophagic response on CLL cell survival, we next examined if chloroquine, which inhibits autophagy via preventing fusion of autophagosomes with lysosomes, affected fludarabine-mediated cell death at concentrations attainable in patients. Although chloroquine inhibited autophagy under these conditions as noted by confocal microscopy showing lack of co-localization between LC3 (autophagosome marker) and LAMP-2 (lysosome marker), it had no effect on fludarabine-mediated cell death. Subsequent studies showed that investigational agents including inhibitors of PI3-kinase, HSP-90, and cyclin-dependent kinases (CDK) each induced autophagy, while inhibitors of histone deacetylases (HDACs) did not. Notably, chloroquine enhanced cytotoxicity mediated by the CDK inhibitor flavopiridol in all patient samples examined, while it produced no effect on the cytotoxicity of the remaining agents. This sensitization to flavopiridol was most striking in CLL samples with the greatest viability after four hours of in vitro treatment with flavopiridol, as assessed by annexin/PI flow cytometry. Given that endoplasmic reticulum (ER) stress induces autophagy in normal cells, we examined this pathway in flavopiridol-treated CLL patient cells. These studies demonstrated that in a subset of samples, flavopiridol, but not fludarabine, promotes early intracellular ER-derived calcium flux, concomitant with the appearance of ER stress evidenced by quantitative real-time PCR showing increased gene expression of specific markers (XBP1, IRE1, Grp78) and standard PCR showing XBP1 splicing. To determine if this finding was relevant to the in vivo setting, we subsequently examined samples obtained serially from CLL patients during treatment with flavopiridol as part of two completed clinical trials at our institution. Similar to our in vitro studies, we found that flavopiridol actively induces autophagy in vivo through an ER stress-directed pathway. Collectively, our data demonstrate that autophagy is relevant to CLL biology and may serve as a pharmacodynamic marker of targeted therapy in CLL. Furthermore, induction of autophagy appears to contribute to flavopiridol resistance in CLL, whereas its role with other therapeutics is unclear. This work is supported by the Leukemia and Lymphoma Society, the D. Warren Brown Foundation, and the National Cancer Institute (CLL Research Consortium and OSU Leukemia SPORE). Disclosures: No relevant conflicts of interest to declare.


Cancers ◽  
2021 ◽  
Vol 13 (10) ◽  
pp. 2411
Author(s):  
Mélody Caillot ◽  
Hassan Dakik ◽  
Frédéric Mazurier ◽  
Brigitte Sola

Multiple myeloma (MM) is a common hematological disease characterized by the accumulation of clonal malignant plasma cells in the bone marrow. Over the past two decades, new therapeutic strategies have significantly improved the treatment outcome and patients survival. Nevertheless, most MM patients relapse underlying the need of new therapeutic approaches. Plasma cells are prone to produce large amounts of immunoglobulins causing the production of intracellular ROS. Although adapted to high level of ROS, MM cells die when exposed to drugs increasing ROS production either directly or by inhibiting antioxidant enzymes. In this review, we discuss the efficacy of ROS-generating drugs for inducing MM cell death and counteracting acquired drug resistance specifically toward proteasome inhibitors.


Sign in / Sign up

Export Citation Format

Share Document