Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 222
Filter
1.
Mol Biol Rep ; 51(1): 975, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259442

ABSTRACT

BACKGROUND: Many methods are used for cancer treatment, especially chemotherapy. In addition to the their therapeutic effects, chemotherapeutic drugs also have serious disadvantages, such as not being cell and tissue-specific, causing toxicity in many tissues, and developing drug resistance. Many methods, especially nanocarriers, have been designed to overcome these disadvantages. METHODS AND RESULTS: In this study, we synthesized mesoporous silica iron oxide nanoparticles with different pore diameters and loaded idarubicin (6MFe3O4-NH2-IDA and 35MFe3O4-NH2-IDA). The synthesized molecules were characterized using FT-IR, XRD, and SEM methods. The cytotoxic effects of unbound idarubicin and idarubicin-loaded nanoparticles on MCF7 and HL-60 cell lines were examined by MTT test. Additionally, the expression of anti-apoptotic (Survivin and BCL-2) and apoptotic (BAX, PUMA, and NOXA) genes of the nanoparticles were measured by PCR method. As a result of the analyses, it was seen that nanoparticles with the desired properties and sizes were synthesized. In MTT analysis, it was observed that both nanoparticles dramatically decreased the IC50 value in cell lines. However, the 35MFe3O4-NH2-IDA molecule was found to have lower IC50 values. IC50 values ​​for pristine IDA, 6MFe3O4-NH2, and 35MFe3O4-NH2 at 24 h were found to be 3.56, 1.24 and 0.25 µM in the MCF7 cell line and 4.15, 1.16 and 0.34 µM in the HL-60 cell line, respectively. Additionally, apoptotic gene expression increased, and anti-apoptotic gene expression decreased. CONCLUSIONS: Our study demonstrates that the effectiveness of idarubicin can be significantly enhanced by its application with mesoporous nanocarriers. This enhancement is attributed to the controlled release of idarubicin from the nanocarrier, which circumvents drug resistance mechanisms, improves drug solubility, and increases the drug-carrying capacity per unit volume due to the porous structure of the carrier. These findings underscore the potential of the synthesized nanocarrier in cancer treatment and provide a clear direction for future research in this field.


Subject(s)
Apoptosis , Idarubicin , Magnetite Nanoparticles , Humans , Idarubicin/pharmacology , Apoptosis/drug effects , MCF-7 Cells , HL-60 Cells , Magnetite Nanoparticles/chemistry , Cell Line, Tumor , Drug Carriers/chemistry , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Porosity
2.
J Med Chem ; 67(16): 13802-13812, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39088428

ABSTRACT

Anthracyclines comprise one of the most effective anticancer drug classes. Doxorubicin, daunorubicin, epirubicin, and idarubicin have been in clinical use for decades, but their application remains complicated by treatment-related toxicities and drug resistance. We previously demonstrated that the combination of DNA damage and histone eviction exerted by doxorubicin drives its associated adverse effects. However, whether the same properties dictate drug resistance is unclear. In the present study, we evaluate a library of 40 anthracyclines on their cytotoxicity, intracellular uptake, and subcellular localization in K562 wildtype versus ABCB1-transporter-overexpressing, doxorubicin-resistant cells. We identify several highly potent cytotoxic anthracyclines. Among these, N,N-dimethyl-idarubicin and anthracycline (composed of the idarubicin aglycon and the aclarubicin trisaccharide) stand out, due to their histone eviction-mediated cytotoxicity toward doxorubicin-resistant cells. Our findings thus uncover understudied anthracycline variants warranting further investigation in the quest for safer and more effective anticancer agents that circumvent cellular export by ABCB1.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Doxorubicin , Drug Resistance, Neoplasm , Idarubicin , Humans , Drug Resistance, Neoplasm/drug effects , Doxorubicin/pharmacology , Doxorubicin/chemistry , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Idarubicin/pharmacology , Idarubicin/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , K562 Cells , Structure-Activity Relationship
3.
Ann Hematol ; 103(8): 2853-2863, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38842564

ABSTRACT

Acute Myeloid Leukemia (AML) is a life-threatening disease whose induction treatment consists of combination chemotherapy with Idarubicin and Cytarabine for fit patients. Treatment failures are frequent, urging the need for novel treatments for this disease. The DNA Damage Response Mechanism (DDR) comprises numerous molecules and pathways intended to arrest the cell cycle until DNA damage is repaired or else drive the cell to apoptosis. AML-derived cell lines after treatment with Idarubicin and Cytarabine were used for studying the expression profile of 84 DDR genes, through PCR arrays. Utilizing de novo AML patient and control samples we studied the expression of PPP1R15A, CDKN1A, GADD45A, GADD45G, and EXO1. Next, we performed PPP1R15A silencing in AML cell lines in two separate experiments using siRNA and CRISPR-cas9, respectively. Our findings highlight that DDR regulators demonstrate increased expression in patients with high cytogenetic risk possibly reflecting increased genotoxic stress. Especially, PPP1R15A is mainly involved in the recovery of the cells from stress and it was the only DDR gene upregulated in AML patients. The PPP1R15A silencing resulted in decreased viability of Idarubicin and Cytarabine-treated cell lines, in contrast to untreated cells. These findings shed light on new strategies to enhance chemotherapy efficacy and demonstrate that PPP1R15A is an important DDR regulator in AML and its downregulation might be a safe and effective way to increase sensitivity to chemotherapy in this disease.


Subject(s)
Cytarabine , DNA Damage , Gene Silencing , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , DNA Damage/drug effects , Cytarabine/pharmacology , Cell Line, Tumor , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Idarubicin/pharmacology , Idarubicin/administration & dosage , Male , Female , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA Repair/drug effects , Middle Aged , Adult , Aged , Gene Expression Regulation, Leukemic/drug effects
4.
Genes (Basel) ; 15(5)2024 05 20.
Article in English | MEDLINE | ID: mdl-38790277

ABSTRACT

Acute myeloid leukemia is the second most frequent type of leukemia in adults. Due to a high risk of development of chemoresistance to first-line chemotherapy, the survival rate of patients in a 5-year period is below 30%. One of the reasons is that the AML population is heterogeneous, with cell populations partly composed of very primitive CD34+CD38- hematopoietic stem/progenitor cells, which are often resistant to chemotherapy. First-line treatment with cytarabine and idarubicin fails to inhibit the proliferation of CD34+CD38- cells. In this study, we investigated Metformin's effect with or without first-line conventional chemotherapy, or with other drugs like venetoclax and S63845, on primitive and undifferentiated CD34+ AML cells in order to explore the potential of Metformin or S63845 to serve as adjuvant therapy for AML. We found that first-line conventional chemotherapy treatment inhibited the growth of cells and arrested the cells in the S phase of the cell cycle; however, metformin affected the accumulation of cells in the G2/M phase. We observed that CD34+ KG1a cells respond better to lower doses of cytarabine or idarubicin in combination with metformin. Also, we determined that treatment with cytarabine, venetoclax, and S63845 downregulated the strong tendency of CD34+ KG1a cells to form cell aggregates in culture due to the downregulation of leukemic stem cell markers like CD34 and CD44, as well as adhesion markers. Also, we found that idarubicin slightly upregulated myeloid differentiation markers, CD11b and CD14. Treatment with cytarabine, idarubicin, venetoclax, metformin, and S63845 upregulated some cell surface markers like HLA-DR expression, and metformin upregulated CD9, CD31, and CD105 cell surface marker expression. In conclusion, we believe that metformin has the potential to be used as an adjuvant in the treatment of resistant-to-first-line-chemotherapy AML cells. Also, we believe that the results of our study will stimulate further research and the potential use of changes in the expression of cell surface markers in the development of new therapeutic strategies.


Subject(s)
Antigens, CD34 , Cytarabine , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute , Metformin , Humans , Metformin/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Drug Resistance, Neoplasm/drug effects , Antigens, CD34/metabolism , Cell Line, Tumor , Cytarabine/pharmacology , Cell Proliferation/drug effects , Sulfonamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Idarubicin/pharmacology
5.
Acta Pharmacol Sin ; 44(11): 2296-2306, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37316630

ABSTRACT

Current therapy for acute myeloid leukemia (AML) is largely hindered by the development of drug resistance of commonly used chemotherapy drugs, including cytarabine, daunorubicin, and idarubicin. In this study, we investigated the molecular mechanisms underlying the chemotherapy drug resistance and potential strategy to improve the efficacy of these drugs against AML. By analyzing data from ex vivo drug-response and multi-omics profiling public data for AML, we identified autophagy activation as a potential target in chemotherapy-resistant patients. In THP-1 and MV-4-11 cell lines, knockdown of autophagy-regulated genes ATG5 or MAP1LC3B significantly enhanced AML cell sensitivity to the chemotherapy drugs cytarabine, daunorubicin, and idarubicin. In silico screening, we found that chloroquine phosphate mimicked autophagy inactivation. We showed that chloroquine phosphate dose-dependently down-regulated the autophagy pathway in MV-4-11 cells. Furthermore, chloroquine phosphate exerted a synergistic antitumor effect with the chemotherapy drugs in vitro and in vivo. These results highlight autophagy activation as a drug resistance mechanism and the combination therapy of chloroquine phosphate and chemotherapy drugs can enhance anti-AML efficacy.


Subject(s)
Idarubicin , Leukemia, Myeloid, Acute , Humans , Idarubicin/pharmacology , Idarubicin/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Daunorubicin/pharmacology , Daunorubicin/therapeutic use , Cytarabine/pharmacology , Cytarabine/therapeutic use , Autophagy , Chloroquine/pharmacology , Chloroquine/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
6.
Parasitol Int ; 96: 102774, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37380124

ABSTRACT

Babesia gibsoni is mainly transmitted by hard ticks of the genus Rhipicephalus (R. sanguineus) and Haemaphysalis (H. longicornis), and causes canine babesiosis. Clinical manifestations of B. gibsoni infection include fever, hemoglobinemia, hemoglobinuria, and progressive anemia. Traditional antibabesial therapy, such as imidocarb dipropionate or diminazene aceturate, can only alleviate severe clinical manifestations and cannot eliminate parasites in the host. Food and Drug Administration (FDA)-approved drugs are a solid starting point for researching novel therapy strategies for canine babesiosis. In this work, we screened 640 FDA-approved drugs against the growth of B. gibsoni in vitro. Among them, 13 compounds (at 10 µM) exhibited high growth inhibition (>60%), and two compounds, namely idarubicin hydrochloride (idamycin) and vorinostat, were chosen for further investigation. The half-maximal inhibitory concentration (IC50) values of idamycin and vorinostat were determined to be 0.044 ± 0.008 µM and 0.591 ± 0.107 µM, respectively. Viability results indicated that a concentration of 4 × IC50 of vorinostat prevented the regrowth of treated B. gibsoni, whereas parasites treated with 4 × IC50 concentration of idamycin remained viable. The B. gibsoni parasites treated with vorinostat exhibited degeneration within erythrocytes and merozoites, in contrast to the oval or signet-ring shape of normal B. gibsoni parasites. In conclusion, FDA-approved drugs offer a valuable platform for drug repositioning in antibabesiosis research. Particularly, vorinostat demonstrated promising inhibitory effects against B. gibsoni in vitro, and further studies on vorinostat are necessary to elucidate its mechanism as a novel treatment in infected animal models.


Subject(s)
Babesia , Babesiosis , Dog Diseases , Ixodidae , United States , Animals , Dogs , Babesiosis/parasitology , Vorinostat/pharmacology , Vorinostat/therapeutic use , Idarubicin/pharmacology , Idarubicin/therapeutic use , United States Food and Drug Administration , Dog Diseases/drug therapy , Dog Diseases/parasitology
7.
Acta Biomater ; 157: 337-351, 2023 02.
Article in English | MEDLINE | ID: mdl-36509402

ABSTRACT

Transarterial chemoembolization (TACE) is an image-guided locoregional therapy used for the treatment of patients with primary hepatocellular carcinoma (HCC). However, conventional TACE formulations such as epirubicin-lipiodol emulsion are rapidly dissociated due to the instability of the emulsion, resulting in insufficient local drug concentrations in the target tumor. To overcome these limitations, we used biodegradable Idarubicin loaded microspheres (BILMs), which were prepared from gelatin and carrageenan and could be loaded with Idarubicin (IDA-MS). The morphology and the ability to load and release IDA of BILMs were characterized in vitro. We evaluated tumor changes and side effects after TACE treatment with IDA-MS in VX2 rabbit and C57BL/6 mice HCC models. In addition, the effect of IDA-MS on the tumor immune microenvironment of HCC tumors was elucidated via mass spectrometry and immunohistochemistry. Result showed that IDA-MS was developed as a new TACE formulation to overcome the poor delivery of drugs due to rapid elimination of the anticancer drug into the systemic circulation. We demonstrated in rabbits and mice HCC models that TACE with IDA-MS resulted in significant tumor shrinkage and no more severe adverse events than those observed in the IDA group. TACE with IDA-MS could also significantly enhance the sensitivity of anti-PD1 immunotherapy, improve the expression of CD8+ T cells, and activate the tumor immune microenvironment in HCC. This study provides a new approach for TACE therapy and immunotherapy and illuminates the future of HCC treatment. STATEMENT OF SIGNIFICANCE: Conventional transarterial chemoembolization (TACE) formulations are rapidly dissociated due to the instability of the emulsion, resulting in insufficient local drug concentrations in hepatocellular carcinoma (HCC). To overcome these limitations, we used biodegradable microspheres called BILMs, which could be loaded with Idarubicin (IDA-MS). We demonstrated in rabbits and mice HCC models that TACE with IDA-MS resulted in significant tumor shrinkage and no more severe adverse events than those observed in the IDA group. TACE with IDA-MS could also significantly enhance the sensitivity of anti-PD1 immunotherapy, improve the expression of CD8+ T cells, and activate the tumor immune microenvironment in HCC. This study provides a new approach for TACE therapy and immunotherapy and illuminates the future of HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Liver Neoplasms , Rabbits , Animals , Mice , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Idarubicin/pharmacology , Liver Neoplasms/therapy , Liver Neoplasms/pathology , Microspheres , CD8-Positive T-Lymphocytes/pathology , Emulsions , Treatment Outcome , Chemoembolization, Therapeutic/methods , Mice, Inbred C57BL , Immunotherapy , Tumor Microenvironment
8.
Oxid Med Cell Longev ; 2022: 3659278, 2022.
Article in English | MEDLINE | ID: mdl-36187332

ABSTRACT

Cardiotoxicity is the major side effect of anthracyclines (doxorubicin, daunorubicin, epirubicin, and idarubicin), though being the most commonly used chemotherapy drugs and the mainstay of therapy in solid and hematological neoplasms. Advances in the field of cardio-oncology have expanded our understanding of the molecular mechanisms underlying anthracycline-induced cardiotoxicity (AIC). AIC has a complex pathogenesis that includes a variety of aspects such as oxidative stress, autophagy, and inflammation. Emerging evidence has strongly suggested that the loss of mitochondrial quality control (MQC) plays an important role in the progression of AIC. Mitochondria are vital organelles in the cardiomyocytes that serve as the key regulators of reactive oxygen species (ROS) production, energy metabolism, cell death, and calcium buffering. However, as mitochondria are susceptible to damage, the MQC system, including mitochondrial dynamics (fusion/fission), mitophagy, mitochondrial biogenesis, and mitochondrial protein quality control, appears to be crucial in maintaining mitochondrial homeostasis. In this review, we summarize current evidence on the role of MQC in the pathogenesis of AIC and highlight the therapeutic potential of restoring the cardiomyocyte MQC system in the prevention and intervention of AIC.


Subject(s)
Anthracyclines , Cardiotoxicity , Anthracyclines/toxicity , Antibiotics, Antineoplastic/pharmacology , Calcium/metabolism , Cardiotoxicity/metabolism , Daunorubicin/metabolism , Daunorubicin/pharmacology , Doxorubicin/pharmacology , Epirubicin/metabolism , Epirubicin/pharmacology , Humans , Idarubicin/metabolism , Idarubicin/pharmacology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism
9.
Acta Biochim Biophys Sin (Shanghai) ; 54(1): 91-98, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35130631

ABSTRACT

Idarubicin (IDA), an anthracycline antineoplastic drug, is commonly used in the treatment of acute myeloid leukemia (AML) with reasonable response rates and clinical benefits. However, some patients still relapse, or do not respond, and suffer high fatality rates. Recent studies have shown that overexpression of PARP-1 may represent an important risk factor in AML patients. The aim of the present study was to determine the underlying molecular mechanisms by which the PARP-1 inhibitor Olaparib enhances the chemosensitivity of the leukemia cell line K562 and THP1 to IDA. Our data demonstrated that PARP-1 is upregulated in AML patients as well as in K562 and THP1 cells, and that the suppression of PARP-1 activity by Olaparib enhances the inhibitory effect of IDA. A mechanistic study revealed that Olaparib decreases the expressions of p-ATM, p-IκBα, XIAP and p65, and upregulates Bax, cleaved-Caspase-3 and γ-H2AX. Olaparib can enhance the induction of DNA damage by IDA, probably mediated by the inhibition of the ATM-related DNA damage response. Moreover, we also found that the nuclear translocation of p65 and the nuclear export of NEMO are inhibited when IDA and Olaparib are combined. Our results suggest that Olaparib attenuates the activity of the NF-κB pathway and decreases the DNA damage response induced by IDA. Therefore, we conclude that Olaparib is a potentially valuable chemosensitizer for leukemia patients.


Subject(s)
Leukemia, Myeloid, Acute , NF-kappa B , Cell Line, Tumor , DNA Damage , Humans , Idarubicin/pharmacology , Idarubicin/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
10.
Molecules ; 26(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34641328

ABSTRACT

This study aims to enhance efficacy and reduce toxicity of the combination treatment of a drug and curcumin (Cur) on leukemic stem cell and leukemic cell lines, including KG-1a and KG-1 (FLT3+ LSCs), EoL-1 (FLT3+ LCs), and U937 (FLT3- LCs). The cytotoxicity of co-treatments of doxorubicin (Dox) or idarubicin (Ida) at concentrations of the IC10-IC80 values and each concentration of Cur at the IC20, IC30, IC40, and IC50 values (conditions 1, 2, 3, and 4) was determined by MTT assays. Dox-Cur increased cytotoxicity in leukemic cells. Dox-Cur co-treatment showed additive and synergistic effects in several conditions. The effect of this co-treatment on FLT3 expression in KG-1a, KG-1, and EoL-1 cells was examined by Western blotting. Dox-Cur decreased FLT3 protein levels and total cell numbers in all the cell lines in a dose-dependent manner. In summary, this study exhibits a novel report of Dox-Cur co-treatment in both enhancing cytotoxicity of Dox and inhibiting cell proliferation via FLT3 protein expression in leukemia stem cells and leukemic cells. This is the option of leukemia treatment with reducing side effects of chemotherapeutic drugs to leukemia patients.


Subject(s)
Curcumin/pharmacology , Doxorubicin/pharmacology , Idarubicin/pharmacology , Leukemia, Myeloid, Acute/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/metabolism , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcuma/chemistry , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Rhizome/chemistry
11.
Biomolecules ; 11(8)2021 07 30.
Article in English | MEDLINE | ID: mdl-34439794

ABSTRACT

Recent breakthroughs have reignited interest in RAS GEFs as direct therapeutic targets. To search for new inhibitors of SOS GEF activity, a repository of known/approved compounds (NIH-NACTS) and a library of new marine compounds (Biomar Microbial Technologies) were screened by means of in vitro RAS-GEF assays using purified, bacterially expressed SOS and RAS constructs. Interestingly, all inhibitors identified in our screenings (two per library) shared related chemical structures belonging to the anthraquinone family of compounds. All our anthraquinone SOS inhibitors were active against the three canonical RAS isoforms when tested in our SOS GEF assays, inhibited RAS activation in mouse embryonic fibroblasts, and were also able to inhibit the growth of different cancer cell lines harboring WT or mutant RAS genes. In contrast to the commercially available anthraquinone inhibitors, our new marine anthraquinone inhibitors did not show in vivo cardiotoxicity, thus providing a lead for future discovery of stronger, clinically useful anthraquinone SOS GEF blockers.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , GTP Phosphohydrolases/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Cardiotoxicity/prevention & control , Cell Line, Transformed , Cell Line, Tumor , Doxorubicin/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Idarubicin/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/genetics , SOS1 Protein/metabolism , Son of Sevenless Proteins/deficiency , Son of Sevenless Proteins/genetics
12.
Theranostics ; 11(12): 5700-5712, 2021.
Article in English | MEDLINE | ID: mdl-33897876

ABSTRACT

Rationale: Increasing the bioavailable drug level in a tumor is the key to enhance efficacy of chemotherapy. Thermosensitive smart drug delivery systems (SDDS) in combination with local hyperthermia facilitate high local drug levels, thus improving uptake in the tumor. However, inability to rapidly and efficiently absorb the locally released drug results in reduced efficacy, as well as undesired redistribution of the drug away from the tumor to the system. Methods: Based on this paradigm we propose a novel approach in which we replaced doxorubicin (DXR), one of the classic drugs for nanocarrier-based delivery, with idarubicin (IDA), a hydrophobic anthracycline used solely in the free form for treatment hematologic cancers. We established a series of in vitro and in vivo experiments to in depth study the kinetics of SDDS-based delivery, drug release, intratumor biodistribution and subsequent cell uptake. Results: We demonstrate that IDA is taken up over 10 times more rapidly by cancer cells than DXR in vitro. Similar trend is observed in in vivo online imaging and less drug redistribution is shown for IDA, together resulting in 4-times higher whole tumor drug uptake for IDA vs. DXR. Together his yielded an improved intratumoral drug distribution for IDA-SDDS, translating into superior tumor response compared to DXR-SDDS treatment at the same dose. Thus, IDA - a drug that is not used for treatment of solid cancers - shows superior therapeutic index and better outcome when administered in externally triggered SDDS. Conclusions: We show that a shift in selection of chemotherapeutics is urgently needed, away from the classic drugs towards selection based on properties of a chemotherapeutic in context of the nanoparticle and delivery mode, to maximize the therapeutic efficacy.


Subject(s)
Idarubicin/pharmacology , Idarubicin/pharmacokinetics , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Biological Transport/drug effects , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Drug Liberation/drug effects , Humans , Hydrogen-Ion Concentration , Hyperthermia, Induced/methods , Kinetics , Mice , Nanoparticles/chemistry , Neoplasms/metabolism , Tissue Distribution/drug effects
13.
Mol Biol Rep ; 48(1): 1017-1023, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33387196

ABSTRACT

Glioblastoma (GBM) is a lethal astrocyte-derived tumor that is currently treated with a multi-modal approach of surgical resection, radiotherapy, and temozolomide-based chemotherapy. Alternatives to current therapies are urgently needed as its prognosis remains poor. Anthracyclines are a class of compounds that show great potential as GBM chemotherapeutic agents and are widely used to treat solid tumors outside the central nervous system. Here we investigate the cytotoxic effects of doxorubicin and other anthracyclines on GL261 glioma tumor cells in anticipation of novel anthracycline-based CNS therapies. Three methods were used to quantify dose-dependent effects of anthracyclines on adherent GL261 tumor cells, a murine cell-based model of GBM. MTT assays quantified anthracycline effects on cell viability, comet assays examined doxorubicin genotoxicity, and flow cytometry with Annexin V/PI staining characterized doxorubicin-induced apoptosis and necrosis. Dose-dependent reductions in GL261 cell viability were found in cells treated with doxorubicin (EC50 = 4.9 µM), epirubicin (EC50 = 5.9 µM), and idarubicin (EC50 = 4.4 µM). Comet assays showed DNA damage following doxorubicin treatments, peaking at concentrations of 1.0 µM and declining after 25 µM. Lastly, flow cytometric analysis of doxorubicin-treated cells showed dose-dependent induction of apoptosis (EC50 = 5.2 µM). Together, these results characterized the cytotoxic effects of anthracyclines on GL261 glioma cells. We found dose-dependent apoptotic induction; however at high concentrations we find that cell death is likely necrotic. Our results support the continued exploration of anthracyclines as compounds with significant potential for improved GBM treatments.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cytotoxins/pharmacology , Doxorubicin/pharmacology , Epirubicin/pharmacology , Idarubicin/pharmacology , Neuroglia/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Comet Assay , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Glioma/drug therapy , Humans , Mice , Models, Biological , Neuroglia/metabolism , Neuroglia/pathology
14.
Stem Cell Rev Rep ; 17(2): 616-627, 2021 04.
Article in English | MEDLINE | ID: mdl-33078278

ABSTRACT

Leukemia stem cells (LSCs) are considered to be the root of relapse for acute myeloid leukemia (AML). Conventional chemotherapeutic drugs fail to eliminate LSCs. Therefore, new therapeutic strategies eliminating LSCs are urgently needed. Our results showed that low-dose Triptolide (TPL) enhanced the anti-AML activity of Idarubicin (IDA) in vitro against LSC-like cells (CD34 + CD38- KG1αand CD34 + CD38- kasumi-1 cells) and CD34+ primary AML cells, while sparing normal cells. Inspiringly, the combination treatment with low-dose TPL and IDA was also effective against CD34 + blasts from AML patients with FLT3-ITD mutation, which is an unfavorable risk factor for AML patients. Moreover, the combination of TPL and IDA induced a remarkable suppression of human leukemia growth in a xenograft mouse model. Mechanistically, the enhanced effect of low dose TPL on IDA against LSCs was attributed to inhibiting DNA damage repair response. Thus, our study may provide a theoretical basis to facilitate the development of a novel LSCs-targeting strategy for AML.Graphical abstract.


Subject(s)
DNA Damage , DNA Repair/drug effects , Diterpenes/pharmacology , Idarubicin , Leukemia, Myeloid, Acute , Neoplastic Stem Cells/drug effects , Phenanthrenes/pharmacology , Animals , Drug Synergism , Epoxy Compounds/pharmacology , Humans , Idarubicin/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice
15.
Biochem Biophys Res Commun ; 535: 73-79, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33341676

ABSTRACT

The Wnt/ß-catenin pathway is an attractive target for the treatment of acute myelogenous leukemia (AML), since aberrant activation of the Wnt/ß-catenin pathway contributes to carcinogenesis in various types of cancers including AML. Screening of an in-house compound library, constructed at Kyoto Pharmaceutical University, identified a novel compound designated "31" that was found to be an inhibitor of the Wnt/ß-catenin pathway. The compound inhibited T-cell factor (TCF) activity in a TCF firefly luciferase-reporter assay and suppressed the proliferation of several human AML cell lines in a dose-dependent manner. Compound 31 arrested the cell cycle of AML cells at the G1 stage and induced apoptosis. Decrease in protein and mRNA expression level of Wnt pathway-related molecules was confirmed by the analyses of western blotting and quantitative reverse transcription-polymerase chain reaction. In addition, compound 31 combined with idarubicin synergistically inhibited the proliferation of AML cells. In conclusion, these results strongly suggest that compound 31 has potential as a novel anti-AML agent targeting the Wnt/ß-catenin signaling pathway.


Subject(s)
Dipeptides/pharmacology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Wnt Signaling Pathway/drug effects , Antineoplastic Agents/analysis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dipeptides/chemistry , Drug Design , Drug Evaluation, Preclinical , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Idarubicin/pharmacology , Luciferases/metabolism
16.
BMC Cancer ; 20(1): 1193, 2020 Dec 04.
Article in English | MEDLINE | ID: mdl-33276759

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is a heterogeneous disease that frequently relapses after standard chemotherapy. Therefore, there is a need for the development of novel chemotherapeutic agents that could treat AML effectively. Radotinib, an oral BCR-ABL tyrosine kinase inhibitor, was developed as a drug for the treatment of chronic myeloid leukemia. Previously, we reported that radotinib exerts increased cytotoxic effects towards AML cells. However, little is known about the effects of combining radotinib with Ara-C, a conventional chemotherapeutic agent for AML, with respect to cell death in AML cells. Therefore, we investigated combination effects of radotinib and Ara-C on AML in this study. METHODS: Synergistic anti-cancer effects of radotinib and Ara-C in AML cells including HL60, HEL92.1.7, THP-1 and bone marrow cells from AML patients have been examined. Diverse cell biological assays such as cell viability assay, Annexin V-positive cells, caspase-3 activity, cell cycle distribution, and related signaling pathway have been performed. RESULTS: The combination of radotinib and Ara-C was found to induce AML cell apoptosis, which involved the mitochondrial pathway. In brief, combined radotinib and Ara-C significantly induced Annexin V-positive cells, cytosolic cytochrome C, and the pro-apoptotic protein Bax in AML cells including HL60, HEL92.1.7, and THP-1. In addition, mitochondrial membrane potential and Bcl-xl protein were markedly decreased by radotinib and Ara-C. Moreover, this combination induced caspase-3 activity. Cleaved caspase-3, 7, and 9 levels were also increased by combined radotinib and Ara-C. Additionally, radotinib and Ara-C co-treatment induced G0/G1 arrest via the induction of CDKIs such as p21 and p27 and the inhibition of CDK2 and cyclin E. Thus, radotinib/Ara-C induces mitochondrial-dependent apoptosis and G0/G1 arrest via the regulation of the CDKI-CDK-cyclin cascade in AML cells. In addition, our results showed that combined treatment with radotinib and Ara-C inhibits AML cell growth, including tumor volumes and weights in vivo. Also, the combination of radotinib and Ara-C can sensitize cells to chemotherapeutic agents such as daunorubicin or idarubicin in AML cells. CONCLUSIONS: Therefore, our results can be concluded that radotinib in combination with Ara-C possesses a strong anti-AML activity.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cytarabine/pharmacology , Leukemia, Myeloid, Acute/pathology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Daunorubicin/pharmacology , Drug Synergism , HL-60 Cells , Humans , Idarubicin/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Random Allocation , Single-Blind Method , Specific Pathogen-Free Organisms , Tumor Cells, Cultured
17.
Biochem J ; 477(21): 4167-4190, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33030198

ABSTRACT

Drug repurposing is an alternative avenue for identifying new drugs to treat tuberculosis (TB). Despite the broad-range of anti-tubercular drugs, the emergence of multi-drug-resistant and extensively drug-resistant strains of Mycobacterium tuberculosis (Mtb) H37Rv, as well as the significant death toll globally, necessitates the development of new and effective drugs to treat TB. In this study, we have employed a drug repurposing approach to address this drug resistance problem by screening the drugbank database to identify novel inhibitors of the Mtb target enzyme, DNA gyrase. The compounds were screened against the ATPase domain of the gyrase B subunit (MtbGyrB47), and the docking results showed that echinacoside, doxorubicin, epirubicin, and idarubicin possess high binding affinities against MtbGyrB47. Comprehensive assessment using fluorescence spectroscopy, surface plasmon resonance spectroscopy (SPR), and circular dichroism (CD) titration studies revealed echinacoside as a potent binder of MtbGyrB47. Furthermore, ATPase, and DNA supercoiling assays exhibited an IC50 values of 2.1-4.7 µM for echinacoside, doxorubicin, epirubicin, and idarubicin. Among these compounds, the least MIC90 of 6.3 and 12 µM were observed for epirubicin and echinacoside, respectively, against Mtb. Our findings indicate that echinacoside and epirubicin targets mycobacterial DNA gyrase, inhibit its catalytic cycle, and retard mycobacterium growth. Further, these compounds exhibit potential scaffolds for optimizing novel anti-mycobacterial agents that can act on drug-resistant strains.


Subject(s)
Antitubercular Agents/pharmacology , DNA Gyrase/metabolism , Mycobacterium tuberculosis/enzymology , Adenosine Triphosphatases/metabolism , Antitubercular Agents/chemistry , Circular Dichroism , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Design , Drug Repositioning/methods , Epirubicin/chemistry , Epirubicin/pharmacology , Glycosides/chemistry , Glycosides/pharmacology , Idarubicin/chemistry , Idarubicin/pharmacology , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Structure-Activity Relationship , Surface Plasmon Resonance , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology
18.
Anticancer Res ; 40(10): 5399-5404, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32988859

ABSTRACT

BACKGROUND/AIM: The aim of the present study was to investigate whether idarubicin (IDR) induces oxidative DNA damage in the presence of copper (II). MATERIALS AND METHODS: DNA damage was evaluated by pBR322 plasmid DNA cleavage. The formation of oxidative stress markers [O2 •- and 8-hydroxy-2'-deoxyguanosine (8-OHdG)] was analysed. RESULTS: IDR induced DNA damage and O2 •- and 8-OHdG generation in the presence of copper (II). CONCLUSION: IDR induced oxidative DNA damage in the presence of copper (II). Since it has been reported that the concentration of copper in the serum of cancer patients is higher than that in healthy groups, IDR-induced oxidative DNA damage in the presence of copper (II) may play an important role in anticancer therapeutic strategies.


Subject(s)
Anthracyclines/pharmacology , Idarubicin/pharmacology , Neoplasms/drug therapy , Oxidative Stress/drug effects , Anthracyclines/chemistry , Copper/chemistry , DNA Damage/drug effects , Humans , Idarubicin/chemistry , Neoplasms/genetics , Neoplasms/pathology , Reactive Oxygen Species/chemistry , Superoxide Dismutase/genetics
19.
Virulence ; 11(1): 636-651, 2020 01 01.
Article in English | MEDLINE | ID: mdl-32423280

ABSTRACT

BACKGROUND: MRSA is a major concern in community settings and in health care. The emergence of biofilms and persister cells substantially increases its antimicrobial resistance. It is very urgent to develop new antimicrobials to solve this problem. OBJECTIVE: Idarubicin was profiled to assess its antimicrobial effects in vitro and in vivo, and the underlying mechanisms. METHODS: We investigated the antimicrobial effects of idarubicin against MRSA by time-kill analysis. The antibiofilm efficacy of idarubicin was assessed by crystal violet and XTT staining, followed by laser confocal microscopy observation. The mechanisms underlying the antimicrobial effects were studied by transmission electron microscopy, all-atom molecular dynamic simulations, SYTOX staining, surface plasma resonance, and DNA gyrase inhibition assay. Further, we addressed the antimicrobial efficacy in wound and subcutaneous abscess infection in vivo. RESULTS: Idarubicin kills MRSA cells by disrupting the lipid bilayers and interrupting the DNA topoisomerase IIA subunits, and idarubicin shows synergistic antimicrobial effects with fosfomycin. Through synergy with a single dose treatment fosfomycin and the addition of the cell protector amifostine, the cytotoxicity and cardiotoxicity of idarubicin were significantly reduced without affecting its antimicrobial effects. Idarubicin alone or in combination with fosfomycin exhibited considerable efficacy in a subcutaneous abscess mouse model of MRSA infection. In addition, idarubicin also showed a low probability of causing resistance and good postantibiotic effects. CONCLUSIONS: Idarubicin and its analogs have the potential to become a new class of antimicrobials for the treatment of MRSA-related infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Idarubicin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Skin Infections/drug therapy , Animals , Biofilms/drug effects , Drug Repositioning , Drug Synergism , Female , Fosfomycin/pharmacology , Fosfomycin/therapeutic use , Idarubicin/analogs & derivatives , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Specific Pathogen-Free Organisms
20.
Clin Lymphoma Myeloma Leuk ; 20(7): e382-e391, 2020 07.
Article in English | MEDLINE | ID: mdl-32336675

ABSTRACT

INTRODUCTION: The purpose of this study was to explore the outcomes of all-trans-retinoic acid (ATRA) and arsenic trioxide (ATO) plus idarubicin (IDA) as a frontline treatment in adult patients with acute promyelocytic leukemia (APL). PATIENTS AND METHODS: We analyzed the outcomes of ATRA and intravenous ATO plus IDA as a frontline induction therapy in 118 patients with APL with high-risk (HR) and standard-risk (SR) disease from January 2008 to December 2017. The medical records of 118 patients with APL (HR, n = 45; SR, n = 73) who received the frontline triple combination regimen at Henan Provincial People's Hospital and Tongji Hospital were retrospectively reviewed. Consolidation therapy comprised 6 cycles of ATO and ATRA plus IDA, and IDA was administered in 1 to 4 cycles of consolidation therapy based on the comparable clinical efficacy compared with 6 cycles and fewer side effects to myocardium without subsequent maintenance therapy. RESULTS: Of 118 patients, there were 3 (2.5%) early deaths and 115 (97.5%) hematologic complete remissions; 102 (88.7%) of 115 patients achieved molecular complete remission following induction therapy, and all patients were polymerase chain reaction-negative for promyelocytic leukemia-retinoic acid receptor alpha after the first cycle of consolidation therapy. The 5-year overall survival (OS) and event-free survival (EFS) were 93.0% ± 2.9% and 92.4% ± 3.0%, respectively. Early death, hematologic complete remissions, molecular complete remissions, and toxicities were comparable between the HR and SR groups. The cumulative incidence of relapse in the HR group was 4.7% (n = 2), and the cumulative incidence of relapse in the SR group was 0. The OS and EFS of the SR and HR groups were comparable (92.3% ± 4.5% vs. 92.8% ± 4.0%; X2 = 0.263; P = .608; 92.3% ± 4.5% vs. 91.1% ± 4.2%, X2 = 0.917; P = .338). The total dosage of IDA was approximately 181 to 258 mg, and no patient experienced cardiotoxicity. OS and EFS were not influenced by fms-related tyrosine kinase 3 internal tandem duplication mutation status (P = .405 and P = .528, respectively). CONCLUSION: The triple combination of ATRA and ATO plus IDA as both an induction and consolidation therapy for the HR and SR groups attained excellent outcomes, and this regimen was effective, safe, and easy, without maintenance therapy. The triple combination treatment might be a preferred frontline therapy for patients with APL, especially for those with HR or the APL fms-related tyrosine kinase 3 internal tandem duplication mutation.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic Trioxide/therapeutic use , Idarubicin/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Adolescent , Adult , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arsenic Trioxide/pharmacology , Female , Humans , Idarubicin/pharmacology , Male , Middle Aged , Prognosis , Retrospective Studies , Risk Factors , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL