Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int J Hematol ; 115(3): 428-434, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34704233

ABSTRACT

Shwachman-Diamond syndrome (SDS) is an autosomal recessive inherited disorder characterized by bone marrow failure, exocrine pancreatic dysfunction, and skeletal abnormalities. SDS is typically caused by a pathogenic mutation in the Shwachman-Bodian-Diamond Syndrome (SBDS) gene. Patients with SDS have an increased risk of developing acute myeloid leukemia (AML) and myelodysplastic syndromes. We identified germline biallelic SBDS mutations (p.K62X and p.I167M) in a 50-year-old AML patient who had never experienced the typical symptoms of SDS. The K62X mutation is one of the most common pathogenic mutations, whereas the significance of the I167M mutation was unclear. Based on cellular experiments, we concluded that the I167M mutation contributed to the development of AML, and chemotherapy including topoisomerase inhibitors, which induce DNA double-strand breaks, may have been toxic to this patient. Our experience indicates that some asymptomatic Shwachman-Bodian-Diamond syndrome mutations contribute to the development of leukemia, and that careful treatment selection may be warranted for patients harboring these mutations.


Subject(s)
Germ-Line Mutation/genetics , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Proteins/genetics , Shwachman-Diamond Syndrome/genetics , DNA/metabolism , DNA Repair/drug effects , Female , Genes, Recessive/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Middle Aged , Myelodysplastic Syndromes/etiology , Shwachman-Diamond Syndrome/complications , Topoisomerase Inhibitors/adverse effects
2.
In Vivo ; 32(4): 765-770, 2018.
Article in English | MEDLINE | ID: mdl-29936457

ABSTRACT

BACKGROUND/AIM: Although there are many reports of anticancer drug-induced neurotoxicity, most previous data have been derived from neuronal cell models grown in a variety of culture conditions. This has prevented accurate assessment of the potency of their neurotoxicity and of changes in drug sensitivity of neuronal cells during differentiation. In this study, a simple neuronal differentiation induction system was established and the relative potency of neurotoxicity of eight anticancer drugs was compared during neuronal cell differentiation. MATERIALS AND METHODS: Rat PC12 cells were induced to differentiate into neuronal cells by 50 ng/ml nerve growth factor in serum-free Dulbecco's modified Eagle's medium, followed by overlay of fresh nutrients at day 3, without medium change. Cell viability was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method. RESULTS: During differentiation, PC12 cells became 1.1-to more than 10,000-fold resistant to anticancer drugs. Topoisomerase inhibitors (doxorubicin, SN-38, etoposide) were the most toxic to differentiated PC12 cells, followed by docetaxel, gefitinib, melphalan, 5-fluorouracil and methotrexate. Docetaxel showed the highest cytotoxicity against undifferentiated PC12 cells, but its cytotoxicity was dramatically reduced during differentiation. CONCLUSION: The present study demonstrated considerable variation in the neurotoxicity of anticancer drugs during the cell differentiation process. The present simple assay system may be useful to search for neuroprotective substances.


Subject(s)
Cell Differentiation/drug effects , Neoplasms/drug therapy , Neurons/drug effects , Topoisomerase Inhibitors/adverse effects , Animals , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Etoposide/adverse effects , Etoposide/pharmacology , Humans , Irinotecan , Neoplasms/pathology , PC12 Cells , Rats , Topoisomerase Inhibitors/therapeutic use
3.
Expert Opin Drug Discov ; 12(7): 747-753, 2017 07.
Article in English | MEDLINE | ID: mdl-28504025

ABSTRACT

INTRODUCTION: Acute myeloid leukemia (AML) represents a disease with a very poor outcome and remains an area of significant unmet need necessitating novel therapeutic strategies. Among novel therapeutic agents, vosaroxin is a first-in-class anticancer quinolone derivative that targets topoisomerase II and induces site-selective double-strand breaks in DNA, leading to tumor cell apoptosis. Areas covered: Herein, the authors provide a comprehensive review of the preclinical development of vosaroxin. This includes coverage of vosaroxin's mechanism of action in addition to its pharmacology and of the main studies reported over the past few years with vosaroxin when used to treat adult AML. Expert opinion: Given that vosaroxin is associated with fewer potential side effects, it may be of benefit to elderly patients with relapsed/refractory AML and to those with additional comorbidities who have previously received an anthracycline and cytarabine combination. Furthermore, vosaroxin also was seen to be active in multidrug-resistant preclinical models. However, further studies have to be performed to better evaluate its place in the armamentarium against AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Naphthyridines/therapeutic use , Thiazoles/therapeutic use , Adult , Aged , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA Breaks, Double-Stranded/drug effects , Drug Evaluation, Preclinical/methods , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/pathology , Naphthyridines/adverse effects , Naphthyridines/pharmacology , Thiazoles/adverse effects , Thiazoles/pharmacology , Topoisomerase Inhibitors/adverse effects , Topoisomerase Inhibitors/pharmacology , Topoisomerase Inhibitors/therapeutic use
4.
J Med Chem ; 60(9): 3776-3794, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28406300

ABSTRACT

There is an urgent unmet medical need for novel antibiotics that are effective against a broad range of bacterial species, especially multidrug resistant ones. Tetrahydropyran-based inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) display potent activity against Gram-positive pathogens and no target-mediated cross-resistance with fluoroquinolones. We report our research efforts aimed at expanding the antibacterial spectrum of this class of molecules toward difficult-to-treat Gram-negative pathogens. Physicochemical properties (polarity and basicity) were considered to guide the design process. Dibasic tetrahydropyran-based compounds such as 6 and 21 are potent inhibitors of both DNA gyrase and topoisomerase IV, displaying antibacterial activities against Gram-positive and Gram-negative pathogens (Staphylococcus aureus, Enterobacteriaceae, Pseudomonas aeruginosa, and Acinetobacter baumannii). Compounds 6 and 21 are efficacious in clinically relevant murine infection models.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Pyrans/pharmacology , Topoisomerase Inhibitors/chemical synthesis , Topoisomerase Inhibitors/pharmacology , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemical synthesis , Guinea Pigs , Humans , Microbial Sensitivity Tests , Myocytes, Cardiac/drug effects , Pyrans/adverse effects , Pyrans/chemical synthesis , Topoisomerase Inhibitors/adverse effects
5.
Cardiovasc Toxicol ; 17(2): 130-139, 2017 04.
Article in English | MEDLINE | ID: mdl-27003392

ABSTRACT

Anthracycline (ANT) is a topoisomerase-interacting agent that is used in most malignancy treatments. We investigated the efficacy of enalapril (angiotensin-converting enzyme inhibitor) in the prevention of ANT-induced cardiomyopathy. In this randomized, single-blind, and placebo-controlled study, 69 patients with a newly diagnosed malignancy for which ANT therapy was planned were randomly assigned to either a group receiving enalapril (n = 34) or placebo (n = 35). Echocardiography studies were performed before chemotherapy and at 6 months after randomization. Additionally, troponin I and creatinine kinase-MB (CK-MB) were measured 1 month after the initiation of chemotherapy. In the enalapril group, the mean left ventricular ejection fraction (LVEF) (p = 0.58) was the same at baseline and 6 months after randomization. Conversely, LVEF significantly decreased in the control group (p < 0.001). Additionally, LV end systolic volume and left atrial diameter were significantly increased compared with the baseline measures in the control group. According to the tissue Doppler study, the mitral annuli early diastolic (e') and peak systolic (s') velocities were significantly reduced, and the E (the peak early diastolic velocity)/e' ratio was significantly increased in the control group. Furthermore, the TnI and CK-MB levels were significantly higher in the control group than in the enalapril group. Enalapril appears efficacious in preserving systolic and diastolic function in cancer patients treated with ANTs.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Cardiomyopathies/prevention & control , Enalapril/therapeutic use , Topoisomerase Inhibitors/adverse effects , Ventricular Dysfunction, Left/prevention & control , Adult , Biomarkers/blood , Cardiomyopathies/chemically induced , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Cardiotoxicity , Creatine Kinase, MB Form/blood , Cytoprotection , Echocardiography, Doppler , Female , Humans , Iran , Male , Middle Aged , Myocardial Contraction/drug effects , Single-Blind Method , Stroke Volume/drug effects , Time Factors , Treatment Outcome , Troponin I/blood , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/drug effects
7.
Dermatol Clin ; 33(4): 787-805, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26433850

ABSTRACT

Traditional chemotherapies, interleukins, phosphorylase inhibitors, and proteasome inhibitors are important therapies available to patients with cutaneous T-cell lymphoma (CTCL). Traditional chemotherapies, both in combination and as single agents, are commonly used in relapsed, refractory CTCLs that behave in an aggressive manner. Interleukins, phosphorylase inhibitors, and proteasome inhibitors are less commonly used but data support a role in patients with more refractory disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma, T-Cell, Cutaneous/drug therapy , Skin Neoplasms/drug therapy , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/classification , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Interleukins/adverse effects , Interleukins/therapeutic use , Mycosis Fungoides/drug therapy , Proteasome Inhibitors/adverse effects , Proteasome Inhibitors/therapeutic use , Sezary Syndrome/drug therapy , Topoisomerase Inhibitors/adverse effects , Topoisomerase Inhibitors/therapeutic use
8.
Gan To Kagaku Ryoho ; 42(7): 821-5, 2015 Jul.
Article in Japanese | MEDLINE | ID: mdl-26197743

ABSTRACT

Nogitecan hydrochloride(topotecan)has shown efficacy in patients with recurrent ovarian cancer, but has not been used widely in Japan. We evaluated the efficacy and adverse effects of topotecan in 12 patients (median age, 62 years) treated for recurrent ovarian cancer between 2000 and 2013. Four patients had relapsed after primary treatment, and 8 had relapsed at least twice. Seven patients had been treated with more than 3 prior regimens. Initial treatment of the 12 patients consisted of intravenous topotecan (1.0-1.4 mg/m2/day) for 5 consecutive days. Initial doses were based on previous chemotherapy and/ or renal function, with reduced doses administered to patients with severe adverse effects during prior courses of treatment. The 12 patients received a total of 54 courses of topotecan(range, 1-15 courses). Of these 12 patients, one achieved a partial response and 6 had stable disease. The median time to progression was 14.4 weeks. All 12 patients had grade 3-4 myelosuppression, while none had febrile neutropenia or severe non-hematologic toxicities. Patients who received higher doses or increased courses of chemotherapy had apparently more severe adverse events. These findings suggested that topotecan should be used as a second- or third-line treatment, rather than later, in patients with tumor recurrence, with its dose reduced according to the physical status of each patient. Such strategies may enhance both the efficacy and safety of topotecan in patients with recurrent ovarian cancer.


Subject(s)
Ovarian Neoplasms/drug therapy , Topoisomerase Inhibitors/therapeutic use , Topotecan/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/pathology , Recurrence , Tomography, X-Ray Computed , Topoisomerase Inhibitors/adverse effects , Topotecan/adverse effects , Treatment Outcome
9.
Mutat Res Rev Mutat Res ; 763: 181-201, 2015.
Article in English | MEDLINE | ID: mdl-25795120

ABSTRACT

In genetic toxicology, risk assessment has traditionally adopted linear dose-responses for any compound that causes genotoxic effects. Increasing evidence of non-linear dose-responses, however, suggests potential cellular tolerance to low levels of many genotoxicants with diverse modes of action. Such putative non-linear dose-responses need to be substantiated by strong mechanistic data that identifies the mechanisms responsible for the tolerance to low doses. This can be achieved by experimental demonstration of cytoprotective mechanisms and by providing experimental support for the existence of tolerance mechanisms against low dose effects. By highlighting key experiments into low dose mechanisms, this review aims to clarify which mechanistic data are required to support the use of non-linear dose-response models in risk assessment. Such key experiments are presented and discussed for alkylating agents, oxidants, particulate matter, nucleoside analogues, topoisomerase inhibitors and aneugens and exemplify the use of gene knockout models or transgenic models as well as chemical modulators of key effectors of relevant pathways and their impact on dose-response relationships. In vitro studies are particularly valuable to elucidate mechanisms of low-dose protection or lack thereof, while in vivo experiments are most appropriate for deriving a safe dose. In order to evaluate the existence of non-linear dose-response relationships for genotoxicants, we suggest that careful attention should be given to the mode of genotoxic action, relevant biomarkers of exposure, as well as to the existence and impact of potential cytoprotective mechanisms like detoxifying metabolism and DNA repair.


Subject(s)
DNA Damage , Mutagenicity Tests/methods , Mutagens/adverse effects , Alkylating Agents/toxicity , Aneugens/adverse effects , Animals , Dose-Response Relationship, Drug , Humans , Models, Chemical , Nucleosides/adverse effects , Oxidants/adverse effects , Particulate Matter/adverse effects , Risk Assessment , Topoisomerase Inhibitors/adverse effects
10.
Am J Hematol ; 90(5): E80-5, 2015 May.
Article in English | MEDLINE | ID: mdl-25653205

ABSTRACT

Therapy-related myeloid neoplasms (t-MN) are a complication of cytotoxic treatment for primary tumors and autoimmune diseases. We report data on 277 t-MN patients, recruited between 1999 and 2013 by the Italian Network on Secondary Leukemias (104 retrospectively and 173 prospectively registered). Median age at t-MN diagnosis was 64 years (range, 21-87). Most frequent primary malignancies (PMs) were lymphoproliferative diseases and breast cancer. One hundred and thirty-three patients had received chemotherapy (CHT), 43 patients radiotherapy (RT), and 101 patients combined CHT/RT for PM. Median time between cytotoxic treatment and t-MN was 5.7 years, with t-MN following RT alone associated with significantly longer latency, compared to CHT or combined CHT/RT (mean, 11.2 vs. 7.1 years, P = 0.0005). The addition of topoisomerase-II inhibitors to alkylating agents was associated with shorter latency compared to alkylating agents alone (median, 6 vs. 8.4 years, P = 0.02). Median survival was 14.6 months from t-MN diagnosis, and was significantly longer in patients treated with allogeneic stem cell transplantation. Significant factors for survival at the multivariable analysis included age, adverse karyotype, and degree of anemia. Our data underline the prognostic importance of karyotype and age in t-MN, similar to de novo acute myeloid leukemia. Treatment approaches should not preclude the use of conventional treatments for younger t-MN patients, including allogeneic stem cell transplantation as potentially curative approach.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Gamma Rays/adverse effects , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/diagnosis , Topoisomerase Inhibitors/adverse effects , Adult , Age Factors , Aged , Aged, 80 and over , Anemia/genetics , Anemia/mortality , Anemia/pathology , Anemia/therapy , Antineoplastic Agents, Alkylating/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Female , Humans , Karyotype , Leukemia, Myeloid, Acute/chemically induced , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/mortality , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/therapy , Male , Middle Aged , Prospective Studies , Retrospective Studies , Severity of Illness Index , Survival Analysis , Topoisomerase Inhibitors/administration & dosage , Transplantation, Homologous
11.
Semin Liver Dis ; 34(2): 162-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24879981

ABSTRACT

Drug-induced liver injury (DILI) due to chemotherapeutic drugs is a significant cause of morbidity and mortality. Most cases of chemotherapy-induced hepatotoxicity are idiosyncratic and do not have a unique clinical or histological signature that is distinct from other agents that cause DILI. The major mechanisms underlying chemotherapy-related hepatotoxicity are based on the production of reactive metabolites generated by phase I oxidation reactions, immunological injury, or alterations in mitochondrial function. Underlying liver disease and hepatic involvement by tumor are important modifiers of liver injury, and reversibility is not universal after drug cessation. Chemotherapy can also exacerbate underlying liver disease, particularly hepatitis B, leading to worsening hepatic function. Diagnosing DILI due to chemotherapeutic agents is particularly challenging because competing etiologies, such as hepatotoxicity from other medications, opportunistic infections, radiation therapy, and pre-existing liver disease, are frequent.


Subject(s)
Antineoplastic Agents/adverse effects , Chemical and Drug Induced Liver Injury/diagnosis , Chemical and Drug Induced Liver Injury/etiology , Protein Kinase Inhibitors/adverse effects , Antibiotics, Antineoplastic/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Antimetabolites, Antineoplastic/adverse effects , Antineoplastic Agents/metabolism , Antineoplastic Agents, Alkylating/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Humans , Platinum Compounds/adverse effects , Taxoids/adverse effects , Topoisomerase Inhibitors/adverse effects
12.
Crit Rev Oncol Hematol ; 91(3): 283-91, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24787275

ABSTRACT

Rearrangements of the MLL gene involve multiple partners and are implicated in both therapy related acute leukemia [tAL] and infant acute leukemia. For these diseases, recently compiled clinical data confirms an elevated frequency of such breakpoints within a 4 kb tract between exon 11 and a region of structural instability adjacent to exon 12. Linked primarily to cases of tAL, interference with topoisomerase II activity may either contribute to the initial DNA lesion directly or indirectly by, for example, providing a physical block to transcription progression. Alternatively, sites of fragmentation may be mis-repaired, guided by intergenic spliced transcripts of the participating genes. Co-transcription of MLL and potential fusion partners may provide the localization that enhances the probability of gene interaction. An indirect role for the leukemogenic activity of topoisomerase II inhibitors would imply that the negative consequences of their use may be separated from their therapeutic effects.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Biphenotypic, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasms, Second Primary/genetics , Oncogene Proteins, Fusion/genetics , Adult , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Epistasis, Genetic , Exons , Histone-Lysine N-Methyltransferase , Humans , Infant , Introns , Leukemia, Biphenotypic, Acute/metabolism , Leukemia, Biphenotypic, Acute/pathology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology , Oncogene Proteins, Fusion/metabolism , Topoisomerase Inhibitors/adverse effects , Translocation, Genetic
13.
Genes Chromosomes Cancer ; 53(3): 248-54, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24310817

ABSTRACT

Genomic characterization of translocation breakpoints is relevant to identify possible mechanisms underlying their origin. The consistent association of anthracylines (e.g., epirubicin and idarubicin) in inducing therapy-related acute leukemias (t-AL) with mixed lineage leukemia (MLL) gene rearrangement suggests that MLL translocations are causative events for t-AL. Using asymmetric multiplex PCR strategy followed by direct DNA sequencing, we characterized the genomic breakpoints of the MLL and AFF1 genes in two patients who developed t-AL with t(4;11)(q21;q23). Chemotherapeutic treatment of the primary disease in both patients included topoisomerase II (topo II) targeting agents. In one case, the MLL breakpoint was located in intron 9 at nucleotide position chr11:118354284 while the AFF1 breakpoint was in intron 3 at nucleotide position chr4:87992070. The breakpoint junction sequences revealed an insertion of two nucleotides at the MLL-AFF1 junction. In the other patient, the MLL breakpoint was located in intron 11 at nucleotide position chr11:118359130-32 and the AFF1 break was in intron 3 at nucleotide position chr4:87996215-17. The MLL breakpoint found in the latter patient was identical to that of two previously reported cases, strongly suggesting the presence of a preferential site of DNA cleavage in the presence of topo II inhibitor. In addition, microhomologies at the breakpoint junctions were indicative of DNA repair by the non-homologous end joining (NHEJ) pathway. This study further supports the evidence that MLL breakpoints in therapy-related acute leukemia with MLL-AFF1 are clustered in the telomeric half of the breakpoint cluster region that contains topo II recognition sites.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Chromosome Breakpoints , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 4/genetics , Genetic Loci , Leukemia, Biphenotypic, Acute/genetics , Topoisomerase Inhibitors/adverse effects , Base Sequence , Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , Epirubicin/adverse effects , Female , Humans , Idarubicin/adverse effects , Leukemia, Biphenotypic, Acute/chemically induced , Leukemia, Promyelocytic, Acute/drug therapy , Middle Aged , Molecular Sequence Data , Multigene Family , Translocation, Genetic
14.
Curr Drug Metab ; 14(10): 1042-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24261706

ABSTRACT

Enrofloxacin is a fluorquinolone exclusively developed for use in veterinary medicine (1980). The kinetics of enrofloxacin are characterized, in general terms, by high bioavailability in most species and rapid absorption after IM, SC or oral administration. However, several studies reported that enrofloxacin showed low bioavailability after oral administration in ruminants. This drug has a broad distribution in the organism, excellent tissue penetration and long serum half-life. Also, enrofloxacin is characterized by a low host toxicity, a broad antibacterial spectrum and high bactericidal activity against major pathogenic bacteria (both Gram-positive and Gram-negative), and intracellular organisms found in diseased animals. The kinetics vary according to the route of administration, formulation, animal species, age, body condition, and physiological status, all of which contribute to differences in drug efficacy. The pharmacokinetic properties of drugs are closely related to their pharmacological efficiency, so it is important to know their behavior in each species that is used. This article reviews the pharmacokinetics of enrofloxacin in several domestic animal species.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Fluoroquinolones/pharmacokinetics , Animals , Animals, Domestic , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Infections/metabolism , Bacterial Infections/veterinary , Biological Availability , Biotransformation , Enrofloxacin , Fluoroquinolones/administration & dosage , Fluoroquinolones/adverse effects , Fluoroquinolones/therapeutic use , Half-Life , Humans , Intestinal Absorption , Metabolic Clearance Rate , Tissue Distribution , Topoisomerase Inhibitors/administration & dosage , Topoisomerase Inhibitors/adverse effects , Topoisomerase Inhibitors/pharmacokinetics , Topoisomerase Inhibitors/therapeutic use
15.
Neoplasia ; 13(11): 1043-57, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22131880

ABSTRACT

We report that daurinol, a novel arylnaphthalene lignan, is a promising potential anticancer agent with adverse effects that are less severe than those of etoposide, a clinical anticancer agent. Despite its potent antitumor activity, clinical use of etoposide is limited because of its adverse effects, including myelosuppression and the development of secondary leukemia. Here, we comprehensively compared the mechanistic differences between daurinol and etoposide because they have similar chemical structures. Etoposide, a topoisomerase II poison, is known to attenuate cancer cell proliferation through the inhibition of DNA synthesis. Etoposide treatment induces G(2)/M arrest, severe DNA damage, and the formation of giant nuclei in HCT116 cells. We hypothesized that the induction of DNA damage and nuclear enlargement due to abnormal chromosomal conditions could give rise to genomic instability in both tumor cells and in actively dividing normal cells, resulting in the toxic adverse effects of etoposide. We found that daurinol is a catalytic inhibitor of human topoisomerase IIa, and it induces S-phase arrest through the enhanced expression of cyclins E and A and by activation of the ATM/Chk/Cdc25A pathway in HCT116 cells. However, daurinol treatment did not cause DNA damage or nuclear enlargement in vitro. Finally, we confirmed the in vivo antitumor effects and adverse effects of daurinol and etoposide in nude mice xenograft models. Daurinol displayed potent antitumor effects without any significant loss of body weight or changes in hematological parameters, whereas etoposide treatment led to decreased body weight and white blood cell, red blood cell, and hemoglobin concentration.


Subject(s)
Carcinoma/pathology , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Etoposide/pharmacology , Hematologic Diseases/chemically induced , Topoisomerase Inhibitors/adverse effects , Topoisomerase Inhibitors/pharmacology , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzodioxoles/adverse effects , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Carcinoma/drug therapy , Colorectal Neoplasms/drug therapy , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Etoposide/adverse effects , Etoposide/therapeutic use , HCT116 Cells , Hematologic Diseases/epidemiology , Hep G2 Cells , Humans , Male , Mice , Mice, Nude , Models, Biological , Naphthalenes/adverse effects , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Topoisomerase Inhibitors/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Acta Clin Belg ; 66(4): 260-6, 2011.
Article in English | MEDLINE | ID: mdl-21938980

ABSTRACT

Drugs with potential cardiac toxicity are prominent in cancer treatment, not only the old chemotherapeutic agents, but also the newer targeted drugs and biologic agents. As the long-term survival of patients with malignancies has improved and cancer turned into a chronic disease, physicians must take into account the short-term as well as long-term consequences of cancer treatment, such as chemotherapy-induced cardiotoxicity. We summarize some general characteristics and subsequently review specific antineoplastic agents that are associated with cardiac toxicity.


Subject(s)
Cardiovascular System/drug effects , Heart/drug effects , Neoplasms/drug therapy , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/adverse effects , Humans , Microtubules/drug effects , Quality of Life , Topoisomerase Inhibitors/adverse effects , Tubulin Modulators/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...