Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.216
Filter
1.
J Med Chem ; 67(7): 5458-5472, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38556750

ABSTRACT

The success of arsenic in acute promyelocytic leukemia (APL) treatment is hardly transferred to non-APL cancers, mainly due to the low selectivity and weak binding affinity of traditional arsenicals to oncoproteins critical for cancer survival. We present herein the reinvention of aliphatic trivalent arsenicals (As) as reversible covalent warheads of As-based targeting inhibitors toward Bruton's tyrosine kinase (BTK). The effects of As warheads' valency, thiol protection, methylation, spacer length, and size on inhibitors' activity were studied. We found that, in contrast to the bulky and rigid aromatic As warhead, the flexible aliphatic As warheads were well compatible with the well-optimized guiding group to achieve nanomolar inhibition against BTK. The optimized As inhibitors effectively blocked the BTK-mediated oncogenic signaling pathway, leading to elevated antiproliferative activities toward lymphoma cells and xenograft tumor. Our study provides a promising strategy enabling rational design of new aliphatic arsenic-based reversible covalent inhibitors toward non-APL cancer treatment.


Subject(s)
Arsenic , Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Arsenicals/pharmacology , Arsenicals/therapeutic use , Arsenic/pharmacology , Agammaglobulinaemia Tyrosine Kinase , Signal Transduction , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
J Ethnopharmacol ; 326: 117778, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38310990

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: In China, the Chinese patent drug Realgar-Indigo naturalis Formula (RIF) is utilized for the therapy of acute promyelocytic leukemia (APL). Comprising four traditional Chinese herb-Realgar, Indigo naturalis, Salvia miltiorrhiza, and Pseudostellaria heterophylla-it notably includes tetra-arsenic tetra-sulfide, indirubin, tanshinone IIa, and total saponins of Radix Pseudostellariae as its primary active components. Due to its arsenic content, RIF distinctly contributes to the therapy for APL. However, the challenge of arsenic resistance in APL patients complicates the clinical use of arsenic agents. Interestingly, RIF demonstrates a high remission rate in APL patients, suggesting that its efficacy is not significantly compromised by arsenic resistance. Yet, the current state of research on RIF's ability to reverse arsenic resistance remains unclear. AIM OF THE STUDY: To investigate the mechanism of different combinations of the compound of RIF in reversing arsenic resistance in APL. MATERIALS AND METHODS: The present study utilized the arsenic-resistant HL60-PMLA216V-RARα cell line to investigate the effects of various RIF compounds, namely tetra-arsenic tetra-sulfide (A), indirubin (I), tanshinone IIa (T), and total saponins of Radix Pseudostellariae (S). The assessment of cell viability, observation of cell morphology, and evaluation of cell apoptosis were performed. Furthermore, the mitochondrial membrane potential, changes in the levels of PMLA216V-RARα, apoptosis-related factors, and the PI3K/AKT/mTOR pathway were examined, along with autophagy in all experimental groups. Meanwhile, we observed the changes about autophagy after blocking the PI3K or mTOR pathway. RESULTS: Tanshinone IIa, indirubin and total saponins of Radix Pseudostellariae could enhance the effect of tetra-arsenic tetra-sulfide down-regulating PMLA216V-RARα, and the mechanism was suggested to be related to inhibiting mTOR pathway to activate autophagy. CONCLUSIONS: We illustrated that the synergistic effect of different compound combinations of RIF can regulate autophagy through the mTOR pathway, enhance cell apoptosis, and degrade arsenic-resistant PMLA216V-RARα.


Subject(s)
Abietanes , Arsenic , Arsenicals , Drugs, Chinese Herbal , Leukemia, Promyelocytic, Acute , Saponins , Humans , Arsenic/adverse effects , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/chemically induced , Phosphatidylinositol 3-Kinases , Arsenicals/pharmacology , Arsenicals/therapeutic use , Sulfides/pharmacology , Sulfides/therapeutic use , Saponins/therapeutic use
3.
Acta Parasitol ; 69(1): 465-470, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38190015

ABSTRACT

PURPOSE: This study was carried out to assess the prevalence of Trypanosoma evansi infection in naturally diseased Dromedary camels in Dammam, Eastern region of Saudi Arabia. The detection of Trypanosoma evansi was performed using the parasitological, serological, and molecular diagnosis and a comparison between such methods were analyzed. In addition, evaluation of therapeutic efficacy of selected antitrypanosomal drugs, cymelarsan and quinapyrmine (aquin-1.5), was trialed for treatment of diagnosed infected cases. METHODS: A total 350 randomly selected camels were evaluated using thin blood smear (TBS), RoTat1.2 PCR and CATT/T. evansi techniques. RESULTS: The total prevalence was 6.9%, 7.7%, and 32.8% by TBS, RoTat1.2 PCR and CATT/T. evansi techniques, respectively. Although PCR detect T. evansi in more samples than TBS, the agreement was good (K = 0.9). Among the CATT/T. evansi results, PCR detect T. evansi in 12 and 15 CATT positive and negative camels, respectively, with low agreement (Kappa = 0.1). The use of cymelarsan and quinapyramine sulfate in the treatment of naturally infected cases demonstrated a very efficient therapeutic response. CONCLUSION: It was found that 1. Comparing the CATT/T. evansi and PCR results, the positivity of CATT was higher than PCR detection, while the agreement was poor (K = 0.1). 2. Cymelarsan and aquin-1.5 proved to be effective in the treatment of naturally infected camels, but cymelarsan presented with higher effectiveness (100%) than aquin-treated camels (83.3%). a 3. The use of cymelarsan and CATT is recommended for disease treatment and control.


Subject(s)
Camelus , Quinolinium Compounds , Triazines , Trypanocidal Agents , Trypanosoma , Trypanosomiasis , Animals , Camelus/parasitology , Trypanosoma/drug effects , Trypanosoma/genetics , Trypanosomiasis/veterinary , Trypanosomiasis/epidemiology , Trypanosomiasis/drug therapy , Trypanosomiasis/parasitology , Saudi Arabia/epidemiology , Trypanocidal Agents/therapeutic use , Trypanocidal Agents/pharmacology , Prevalence , Polymerase Chain Reaction/veterinary , Arsenicals/therapeutic use , Male
5.
FEBS J ; 291(7): 1422-1438, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38129745

ABSTRACT

Acute promyelocytic leukemia (APL) is characterized by the fusion gene promyelocytic leukemia-retinoic acid receptor-alpha (PML-RARA) and is conventionally treated with arsenic trioxide (ATO). ATO binds directly to the RING finger, B-box, coiled-coil (RBCC) domain of PML and initiates degradation of the fusion oncoprotein PML-RARA. However, the mutational hotspot at C212-S220 disrupts ATO binding, leading to drug resistance in APL. Therefore, structural consequences of these point mutations in PML that remain uncertain require comprehensive analysis. In this study, we investigated the structure-based ensemble properties of the promyelocytic leukemia-RING-B-box-coiled-coil (PML-RBCC) domains and ATO-resistant mutations. Oligomeric studies reveal that PML-RBCC wild-type and mutants C212R, S214L, A216T, L217F, and S220G predominantly form tetramers, whereas mutants C213R, A216V, L218P, and D219H tend to form dimers. The stability of the dimeric mutants was lower, exhibiting a melting temperature (Tm) reduction of 30 °C compared with the tetrameric mutants and wild-type PML protein. Furthermore, the exposed surface of the C213R mutation rendered it more prone to protease digestion than that of the C212R mutation. The spectroscopic analysis highlighted ATO-induced structural alterations in S214L, A216V, and D219H mutants, in contrast to C213R, L217F, and L218P mutations. Moreover, the computational analysis revealed that the ATO-resistant mutations C213R, A216V, L217F, and L218P caused changes in the size, shape, and flexibility of the PML-RBCC wild-type protein. The mutations C213R, A216V, L217F, and L218P destabilize the wild-type protein structure due to the adaptation of distinct conformational changes. In addition, these mutations disrupt several hydrogen bonds, including interactions involving C212, C213, and C215, which are essential for ATO binding. The local and global structural features induced by these mutations provide mechanistic insight into ATO resistance and APL pathogenesis.


Subject(s)
Antineoplastic Agents , Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Antineoplastic Agents/pharmacology , Arsenic Trioxide/therapeutic use , Arsenicals/pharmacology , Arsenicals/therapeutic use , Cysteine/genetics , Drug Resistance, Neoplasm/genetics , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Mutation , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oxides/pharmacology , Oxides/therapeutic use
6.
J Med Chem ; 66(24): 16579-16596, 2023 12 28.
Article in English | MEDLINE | ID: mdl-38069817

ABSTRACT

Arsenic trioxide (ATO) targets PML/RARα and leads to miraculous success in treating acute promyelocytic leukemia. Notably, ATO also targets p53, the most frequently mutated protein in cancers, through a similar binding mechanism. However, p53-targeting ATO trials are challenging due to the poor cellular uptake and cancer selectivity of ATO. Here, we analyzed the structure-activity relationship of arsenicals and rationally developed a novel arsenical (designated AcGlcAs) by conjugating arsenic to sulfur atoms and tetraacetyl-ß-d-thioglucose. AcGlcAs exhibited remarkable cellular uptake through a thiol-mediated pathway (maximally 127-fold higher than ATO), thereby potently targeting PML/RARα and mutant p53. Among the 55 tested cell lines, AcGlcAs preferentially killed cancer lines rather than normal lines. In preclinical studies, AcGlcAs significantly extended the survival of mice bearing a xenograft tumor with p53 mutation while showing high plasma stability and oral bioavailability. Thus, AcGlcAs is a potential clinical candidate for precisely treating numerous p53-mutated cancers.


Subject(s)
Antineoplastic Agents , Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Mice , Animals , Tumor Suppressor Protein p53/metabolism , Oxides/pharmacology , Oxides/metabolism , Apoptosis , Arsenic Trioxide/pharmacology , Arsenic Trioxide/metabolism , Arsenic Trioxide/therapeutic use , Arsenicals/pharmacology , Arsenicals/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
7.
Cancer Discov ; 13(12): 2505-2506, 2023 12 12.
Article in English | MEDLINE | ID: mdl-38084094

ABSTRACT

SUMMARY: The study by Bercier and colleagues investigates the mechanisms of action of arsenic trioxide (ATO). The authors find that ATO promotes transition of PML nuclear bodies to a gel-like state via the PML trimerization domain and a critical cysteine residue. Overall, this work sheds new light onto how PML-RARα, the oncogene of APL, is targeted by ATO for disease eradication. See related article by Bercier et al., p. 2548 (6).


Subject(s)
Antineoplastic Agents , Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Arsenic Trioxide/pharmacology , Arsenic Trioxide/therapeutic use , Arsenicals/pharmacology , Arsenicals/therapeutic use , Oxides/pharmacology , Oxides/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Oncogene Proteins, Fusion/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
8.
J Cancer Res Ther ; 19(5): 1371-1378, 2023.
Article in English | MEDLINE | ID: mdl-37787311

ABSTRACT

Background: Acute promyelocytic leukemia (APL) comprises approximately 10% of acute myeloid leukemia (AML) cases. Material and Methods: Both options of treatment (ATRA-ATO and ATRA-chemotherapy) were discussed with patients with low- and intermediate-risk APL, pros and cons explained in details, and treatment regimen selected after getting informed written consent. Results: Total 71 patients were included in the study; among these patients, 3 were negative for both FISH for t (15,17) and RT-PCR for promyelocytic leukemia retinoic acid receptor alpha, and 36 patients with APL had white blood cell count at diagnosis >10 × 109/l. Total 30 patients with newly diagnosed as low- and intermediate-risk-APL fulfilled all inclusion criteria, treated and followed for a minimum period of 2 years up to June, 2016. Fifteen patients liked to be treated with all-trans retinoic acid (ATRA) and arsenic trioxide (ATO), while rest of the 15 patients preferred treatment with ATRA and chemotherapy. Conclusion: Combination of ATRA and ATO is equally effective, less toxic, and more feasible in comparison to ATRA and chemotherapy for patients with low- and intermediate-risk APL and is a viable option for this subset of patients, especially in countries with limited resources.


Subject(s)
Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/diagnosis , Arsenicals/therapeutic use , Oxides/therapeutic use , Tertiary Care Centers , Arsenic Trioxide/therapeutic use , Tretinoin/therapeutic use , Tretinoin/adverse effects , Treatment Outcome , Antineoplastic Combined Chemotherapy Protocols/adverse effects
9.
Anticancer Agents Med Chem ; 23(20): 2217-2224, 2023.
Article in English | MEDLINE | ID: mdl-37888819

ABSTRACT

BACKGROUND: Neuroblastoma (NB) remains associated with a low overall survival rate over the long term. Abnormal activation of the Hedgehog (HH) signaling pathway can activate the transcription of various downstream target genes that promote NB. Both arsenic trioxide (ATO) and itraconazole (ITRA) can inhibit tumor growth. OBJECTIVE: To determine whether ATO combined with ITRA can be used to treat NB with HH pathway activation, we examined the effects of ATO and ITRA monotherapy or combined inhibition of the HH pathway in NB. METHODS: Analysis of CCK8 and flow cytometry showed cell inhibition and cell cycle, respectively. Real-time PCR analysis was conducted to assess the mRNA expression of HH pathway. RESULTS: We revealed that as concentrations of ATO and ITRA increased, the killing effects of both agents on SK-N-BE(2) cells became more apparent. During G2/M, the cell cycle was largely arrested by ATO alone and combined with ITRA, and in the G0/G1 phase by ITRA alone. In the HH pathway, ATO inhibited the transcription of the SHH, PTCH1, SMO and GLI2 genes, however, ITRA did not. Instead of showing synergistic effects in a combined mode, ITRA decreased ATO inhibitory effects. CONCLUSION: We showed that ATO is an important inhibitor of HH pathway but ITRA can weaken the inhibitory effect of ATO. This study provides an experimental evidence for the clinical use of ATO and ITRA in the treatment of NB with HH pathway activation in cytology.


Subject(s)
Arsenicals , Neuroblastoma , Humans , Arsenic Trioxide/pharmacology , Hedgehog Proteins/metabolism , Itraconazole/pharmacology , Oxides/pharmacology , Oxides/therapeutic use , Arsenicals/pharmacology , Arsenicals/therapeutic use , Cell Line, Tumor , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Apoptosis
10.
Cancer Med ; 12(17): 17766-17775, 2023 09.
Article in English | MEDLINE | ID: mdl-37584196

ABSTRACT

BACKGROUND: The innovative combination of all-trans retinoic acid (ATRA) and arsenic trioxide (ATO) has established a new chapter of curative approach in acute promyelocytic leukemia (APL). The disease characteristics and prognostic influence of additional cytogenetic abnormalities (ACA) in APL with modern therapeutic strategy need to be elucidated. METHODS: In the present study, we retrospectively investigated disease features and prognostic power of ACA in 171 APL patients treated with ATRA-ATO-containing regimens. RESULTS: Patients with ACA had markedly decreased hemoglobin levels than that without ACA (p = 0.021). Risk stratification in the ACA group was significantly worse than that in the non-ACA group (p = 0.032). With a median follow-up period of 62.0 months, worse event-free survival (EFS) was demonstrated in patients harboring ACA. Multivariate analysis showed that ACA was an independent adverse factor for EFS (p = 0.033). By further subgroup analysis, in CD34 and CD56 negative APL, patients harboring ACA had inferior EFS (p = 0.017; p = 0.037). CONCLUSIONS: To sum up, ACA remains the independent prognostic value for EFS, we should build risk-adapted therapeutic strategies in the long-term management of APL when such abnormalities are detected.


Subject(s)
Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Leukemia, Promyelocytic, Acute/diagnosis , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Progression-Free Survival , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Tretinoin/therapeutic use , Chromosome Aberrations , Oxides/therapeutic use , Arsenicals/therapeutic use , Treatment Outcome
11.
J Ethnopharmacol ; 317: 116895, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37467822

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Realgar-Indigo naturalis formula (RIF), a first-line drug for the treatment of acute promyelocytic leukemia (APL),is also a TCM formula entirely designed based on TCM theories. There have been studies that explain the scientific connotation of the compatibility of RIF from the perspective of pharmacodynamics. However, as one of the arsenic-containing preparations, the safety of realgar is widely concerned, and there has not been systematic studies to explain the scientific connotation of RIF from the perspective of toxicology. AIM OF THIS STUDY: Dissection of scientific compatibility of Chinese medicinal formula Realgar-Indigo naturalis as an effective treatment for promyelocytic leukemia from the perspective of toxicology. MATERIALS AND METHODS: We used normal mice and an APL model to explore (i) the effects of different components on intestinal permeability, (ii) the changes in intestinal flora, and (iii) toxic effects. At the same time, a bionic extraction method was used to study the effects of different components on the dissolution of soluble arsenic in realgar under the acidic environment in the stomach and the alkaline environment in the intestinal tract. RESULTS: Salvia miltiorrhiza Bunge can repair the intestinal mucosal barrier, maintain the homeostasis of intestinal flora, intervene in the dissolution process of realgar, reverse the increase in intestinal permeability and the disturbance of intestinal flora caused by realgar, and reduce toxicity. CONCLUSION: From the perspective of toxicology, we propose new insights into the definition of the roles of each component in the RIF formula, namely realgar is the monarch, Indigo naturalis is the minister, Salvia miltiorrhiza Bungeis the assistant.


Subject(s)
Arsenic , Arsenicals , Leukemia, Promyelocytic, Acute , Mice , Animals , Leukemia, Promyelocytic, Acute/drug therapy , Arsenicals/therapeutic use , Arsenicals/pharmacology
12.
Leuk Lymphoma ; 64(10): 1673-1680, 2023 10.
Article in English | MEDLINE | ID: mdl-37493540

ABSTRACT

Significant variations exist related to the end of induction practices in the management of Acute Promyelocytic Leukemia (APL). These variations include all-trans retinoic acid (ATRA)-arsenic trioxide (ATO) in fixed doses versus continuation until hematologic complete remission (CR) and performance versus omission of post-induction bone marrow biopsy to confirm morphological CR. A retrospective chart review was conducted of 61 patients (42 low/intermediate-risk and 19 high-risk) aged ≥ 18 years with newly diagnosed APL treated with fixed duration ATRA-ATO +/- cytoreduction at a tertiary medical center from December 2012 through March 2020. Of the 54 patients with post-induction bone marrow biopsy results, 52 (96%) demonstrated no morphologic evidence of APL while the remaining were equivocal. After 2.6 years median follow-up, no relapses occurred. The estimated 2-year overall survival rate of 95% suggests excellent outcomes with a fixed ATO induction regimen and safe omission of post-induction bone marrow biopsy irrespective of hematologic parameters.


Subject(s)
Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Leukemia, Promyelocytic, Acute/diagnosis , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/etiology , Bone Marrow , Retrospective Studies , Arsenicals/therapeutic use , Oxides/therapeutic use , Treatment Outcome , Arsenic Trioxide/therapeutic use , Tretinoin/therapeutic use , Remission Induction , Antineoplastic Combined Chemotherapy Protocols/adverse effects
14.
Anticancer Agents Med Chem ; 23(16): 1829-1837, 2023.
Article in English | MEDLINE | ID: mdl-37259217

ABSTRACT

INTRODUCTION: Small cell lung cancer (SCLC) is featured by a high TP53 mutant rate. Our previous research found that arsenic trioxide (As2O3) could significantly inhibit the growth and metastasis of SCLC. Studies have shown that the degradation of mutant p53 mediated by murine double minute 2 (MDM2) can be induced by As2O3, which probably contributes to the inhibition of SCLC, but the detailed mechanism is still unclear. We aimed to testify that As2O3 can inhibit the growth of SCLC cells by degrading mutant p53 protein via binding to MDM2. METHODS: CCK-8 assay, cell cycle analysis, and western blot of apoptosis markers were used to evaluate the inhibitory effect of As2O3 on NCI-H446 cells (containing mutant p53) and NCI-H1299 cells (p53 null). The effects of As2O3 on p53 and its downstream proteins were identified by western blot using mut-p53-knockdown and overexpressed cell models. MDM2-knockdown cell models were constructed, and western blot, co-IP of mut-p53, and ubiquitin were carried out to explore the mediating effect of MDM2 in As2O3 induced mut-p53 degradation. RESULTS: As2O3 inhibited proliferation and induced cell cycle arrest and apoptosis of SCLC cells in a dose- and timedependent manner. After mut-p53 knockdown or overexpressed, the inhibitory effect of As2O3 was dampened or enhanced. Additionally, As2O3-induced mut-p53 ubiquitination was significantly weakened after MDM2 knockdown. CONCLUSION: As2O3 could inhibit SCLC cells by inhibiting proliferation and inducing cell cycle arrest and apoptosis. These inhibitory effects were achieved at least in part by upregulating MDM2, which, in turn, promotes ubiquitination and degradation of mut-p53.


Subject(s)
Antineoplastic Agents , Arsenicals , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Animals , Mice , Arsenic Trioxide/pharmacology , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Oxides/pharmacology , Oxides/metabolism , Oxides/therapeutic use , Arsenicals/pharmacology , Arsenicals/metabolism , Arsenicals/therapeutic use , Cell Line, Tumor , Apoptosis , Lung Neoplasms/pathology , Cell Proliferation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/pharmacology , Proto-Oncogene Proteins c-mdm2/therapeutic use
15.
Biomed Pharmacother ; 163: 114858, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37172335

ABSTRACT

Torsemide is commonly used to relieve edema during the treatment of acute promyelocytic leukemia (APL) with arsenic trioxide (ATO). We explored the effect of torsemide on the plasma concentrations of inorganic arsenic (iAs), monomethylarsonic acid (MMAV) and dimethyarsinic acid (DMAV) in APL patients treated with ATO and clarified its molecular mechanism in rats and cells. The study included 146 APL patients treated with ATO. 60(41.1 %) of these 146 patients were co-administered with torsemide. The treatment of torsemide increased plasma concentrations of iAs (P < 0.05) and DMAV (P < 0.05) in APL patients. The single co-administration of ATO and torsemide in rats significantly increased the plasma concentrations and AUC(0-t) of iAs (P < 0.05) and MMAV (P < 0.05), decreased the urinary excretion rates and the urine concentrations of iAs (P < 0.05) and DMAV (P < 0.05), and enhanced iAs (P < 0.05) and MMAV (P < 0.05) concentrations in the kidneys of rats. In addition, torsemide decreased the expression of multidrug resistance protein 4 (MRP4) in rat kidneys after 7 days of continuous co-administration (P < 0.05). We also treated MRP4-overexpressing HEK293T cells with ATO and different concentrations of torsemide. Torsemide markedly increased the concentrations of iAs, MMAV and DMAV by inhibiting MRP4 compared with ATO alone (P < 0.05). In conclusion, torsemide increased the plasma concentrations of arsenic metabolites in APL patients treated with ATO by inhibiting the transporter MRP4 in a dose-dependent manner.


Subject(s)
Antineoplastic Agents , Arsenic , Arsenicals , Leukemia, Promyelocytic, Acute , Animals , Humans , Rats , Antineoplastic Agents/adverse effects , Arsenic/metabolism , Arsenic Trioxide/pharmacology , Arsenicals/pharmacology , Arsenicals/therapeutic use , Drug Resistance, Multiple , HEK293 Cells , Leukemia, Promyelocytic, Acute/drug therapy , Multidrug Resistance-Associated Proteins , Oxides , Torsemide/therapeutic use
16.
Drug Res (Stuttg) ; 73(7): 417-425, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37230480

ABSTRACT

INTRODUCTION: Lung cancer is one of the deadliest cancers globally. Arsenic trioxide (ATO) is still present as a highly effective drug in treating acute promyelocytic leukemia (APL). Chemotherapy resistance is one of the major problems in cancer therapy. Necroptosis, can overcomes resistance to apoptosis, and can promote cancer treatment. This study examines the necroptosis pathway in A549 cancer cells exposed to ATO. METHODS: We used the MTT test to determine the ATO effects on the viability of A549 cells at three different time intervals. Also, the reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were performed in three-time intervals. The effect of ATO on apoptosis was evaluated by Annexin V / PI staining and, the RIPK1 and MLKL gene expression were measured by Real-Time PCR. RESULTS: The ATO has dose and time-dependent cytotoxic effects, so at 24, 48, and 72 h, the IC50 doses were 33.81 '11.44 '2.535 µM respectively. A 50 µM ATO is the most appropriate to increase the MMP loss significantly at all three times. At 24 and 48 h after exposure of cells to ATO, the ROS levels increased. The RIPK1 gene expression increased significantly compared to the control group at concentrations of 50 and 100 µM; however, MLKL gene expression decreased. CONCLUSIONS: The A549 cells, after 48 h exposure to ATO at 50 and 100 µM, induces apoptosis and necroptosis. Due to the reduced expression of MLKL, it can be concluded that ATO is probably effective in the metastatic stage of cancer cells.


Subject(s)
Antineoplastic Agents , Arsenicals , Humans , Arsenic Trioxide/pharmacology , A549 Cells , Reactive Oxygen Species/metabolism , Necroptosis , Arsenicals/pharmacology , Arsenicals/therapeutic use , Oxides/pharmacology , Oxides/therapeutic use , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor
17.
Cell Death Differ ; 30(5): 1320-1333, 2023 05.
Article in English | MEDLINE | ID: mdl-36894687

ABSTRACT

Acute promyelocytic leukemia (APL) is driven by the oncoprotein PML-RARα, which recruits corepressor complexes, including histone deacetylases (HDACs), to suppress cell differentiation and promote APL initiation. All-trans retinoic acid (ATRA) combined with arsenic trioxide (ATO) or chemotherapy highly improves the prognosis of APL patients. However, refractoriness to ATRA and ATO may occur, which leads to relapsed disease in a group of patients. Here, we report that HDAC3 was highly expressed in the APL subtype of AML, and the protein level of HDAC3 was positively associated with PML-RARα. Mechanistically, we found that HDAC3 deacetylated PML-RARα at lysine 394, which reduced PIAS1-mediated PML-RARα SUMOylation and subsequent RNF4-induced ubiquitylation. HDAC3 inhibition promoted PML-RARα ubiquitylation and degradation and reduced the expression of PML-RARα in both wild-type and ATRA- or ATO-resistant APL cells. Furthermore, genetic or pharmacological inhibition of HDAC3 induced differentiation, apoptosis, and decreased cellular self-renewal of APL cells, including primary leukemia cells from patients with resistant APL. Using both cell line- and patient-derived xenograft models, we demonstrated that treatment with an HDAC3 inhibitor or combination of ATRA/ATO reduced APL progression. In conclusion, our study identifies the role of HDAC3 as a positive regulator of the PML-RARα oncoprotein by deacetylating PML-RARα and suggests that targeting HDAC3 could be a promising strategy to treat relapsed/refractory APL.


Subject(s)
Antineoplastic Agents , Arsenic , Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Arsenic/metabolism , Arsenic/pharmacology , Arsenic/therapeutic use , Arsenic Trioxide/pharmacology , Arsenic Trioxide/metabolism , Arsenic Trioxide/therapeutic use , Arsenicals/metabolism , Arsenicals/pharmacology , Arsenicals/therapeutic use , Cell Differentiation , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Proteins/metabolism , Oxides/metabolism , Oxides/pharmacology , Oxides/therapeutic use , Transcription Factors/metabolism , Tretinoin/pharmacology , Ubiquitination
19.
Acta Haematol ; 146(3): 240-244, 2023.
Article in English | MEDLINE | ID: mdl-36538909

ABSTRACT

The association of glomerulonephritis and malignant hematological disease is uncommon, but well known in chronic leukemias, lymphomas, and monoclonal gammopathies. However, only a few cases of glomerulonephritis and acute myeloid leukemia have been reported in the literature. We describe the first case of a genetically diagnosed acute promyelocytic leukemia presenting with nephrotic range proteinuria that resolved with induction therapy with ATRA and ATO and performed a comprehensive review of the literature.


Subject(s)
Arsenicals , Glomerulonephritis , Leukemia, Promyelocytic, Acute , Humans , Leukemia, Promyelocytic, Acute/complications , Leukemia, Promyelocytic, Acute/diagnosis , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/therapeutic use , Induction Chemotherapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Glomerulonephritis/diagnosis , Glomerulonephritis/drug therapy , Glomerulonephritis/etiology , Oxides/therapeutic use , Arsenicals/therapeutic use
20.
Cell Mol Immunol ; 20(1): 51-64, 2023 01.
Article in English | MEDLINE | ID: mdl-36447031

ABSTRACT

Boosting tumor immunosurveillance with vaccines has been proven to be a feasible and cost-effective strategy to fight cancer. Although major breakthroughs have been achieved in preventative tumor vaccines targeting oncogenic viruses, limited advances have been made in curative vaccines for virus-irrelevant malignancies. Accumulating evidence suggests that preconditioning tumor cells with certain cytotoxic drugs can generate whole-cell tumor vaccines with strong prophylactic activities. However, the immunogenicity of these vaccines is not sufficient to restrain the outgrowth of existing tumors. In this study, we identified arsenic trioxide (ATO) as a wide-spectrum cytotoxic and highly immunogenic drug through multiparameter screening. ATO preconditioning could generate whole-cell tumor vaccines with potent antineoplastic effects in both prophylactic and therapeutic settings. The tumor-preventive or tumor-suppressive benefits of these vaccines relied on CD8+ T cells and type I and II interferon signaling and could be linked to the release of immunostimulatory danger molecules. Unexpectedly, following ATO-induced oxidative stress, multiple cell death pathways were activated, including autophagy, apoptosis, necroptosis, and ferroptosis. CRISPR‒Cas9-mediated knockout of cell death executors revealed that the absence of Rip3, Mlkl, or Acsl4 largely abolished the efficacy of ATO-based prophylactic and therapeutic cancer vaccines. This therapeutic failure could be rescued by coadministration of danger molecule analogs. In addition, PD-1 blockade synergistically improved the therapeutic efficacy of ATO-based cancer vaccines by augmenting local IFN-γ production.


Subject(s)
Antineoplastic Agents , Arsenicals , Cancer Vaccines , Ferroptosis , Neoplasms , Humans , Arsenic Trioxide/pharmacology , CD8-Positive T-Lymphocytes , Necroptosis , Arsenicals/pharmacology , Arsenicals/therapeutic use , Oxides/pharmacology , Oxides/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Neoplasms/drug therapy , Immunity , Cell Line, Tumor
SELECTION OF CITATIONS
SEARCH DETAIL
...