Compounds that inhibit the activity of DNA TOPOISOMERASE I.
DNA TOPOISOMERASES that catalyze ATP-independent breakage of one of the two strands of DNA, passage of the unbroken strand through the break, and rejoining of the broken strand. DNA Topoisomerases, Type I enzymes reduce the topological stress in the DNA structure by relaxing the superhelical turns and knotted rings in the DNA helix.
An alkaloid isolated from the stem wood of the Chinese tree, Camptotheca acuminata. This compound selectively inhibits the nuclear enzyme DNA TOPOISOMERASES, TYPE I. Several semisynthetic analogs of camptothecin have demonstrated antitumor activity.
Compounds that inhibit the activity of DNA TOPOISOMERASE II. Included in this category are a variety of ANTINEOPLASTIC AGENTS which target the eukaryotic form of topoisomerase II and ANTIBACTERIAL AGENTS which target the prokaryotic form of topoisomerase II.
An antineoplastic agent used to treat ovarian cancer. It works by inhibiting DNA TOPOISOMERASES, TYPE I.
DNA TOPOISOMERASES that catalyze ATP-dependent breakage of both strands of DNA, passage of the unbroken strands through the breaks, and rejoining of the broken strands. These enzymes bring about relaxation of the supercoiled DNA and resolution of a knotted circular DNA duplex.
Compounds that inhibit the activity of DNA TOPOISOMERASES.
Agents obtained from higher plants that have demonstrable cytostatic or antineoplastic activity.
A family of fused-ring hydrocarbons isolated from coal tar that act as intermediates in various chemical reactions and are used in the production of coumarone-indene resins.
A semisynthetic derivative of PODOPHYLLOTOXIN that exhibits antitumor activity. Teniposide inhibits DNA synthesis by forming a complex with topoisomerase II and DNA. This complex induces breaks in double stranded DNA and prevents repair by topoisomerase II binding. Accumulated breaks in DNA prevent cells from entering into the mitotic phase of the cell cycle, and lead to cell death. Teniposide acts primarily in the G2 and S phases of the cycle.
Compounds or agents that combine with an enzyme in such a manner as to prevent the normal substrate-enzyme combination and the catalytic reaction.
Substances that inhibit or prevent the proliferation of NEOPLASMS.
Methods of investigating the effectiveness of anticancer cytotoxic drugs and biologic inhibitors. These include in vitro cell-kill models and cytostatic dye exclusion tests as well as in vivo measurement of tumor growth parameters in laboratory animals.
A reaction that severs one of the covalent sugar-phosphate linkages between NUCLEOTIDES that compose the sugar phosphate backbone of DNA. It is catalyzed enzymatically, chemically or by radiation. Cleavage may be exonucleolytic - removing the end nucleotide, or endonucleolytic - splitting the strand in two.
Circular duplex DNA isolated from viruses, bacteria and mitochondria in supercoiled or supertwisted form. This superhelical DNA is endowed with free energy. During transcription, the magnitude of RNA initiation is proportional to the DNA superhelicity.
Agents that are capable of inserting themselves between the successive bases in DNA, thus kinking, uncoiling or otherwise deforming it and therefore preventing its proper functioning. They are used in the study of DNA.
Injuries to DNA that introduce deviations from its normal, intact structure and which may, if left unrepaired, result in a MUTATION or a block of DNA REPLICATION. These deviations may be caused by physical or chemical agents and occur by natural or unnatural, introduced circumstances. They include the introduction of illegitimate bases during replication or by deamination or other modification of bases; the loss of a base from the DNA backbone leaving an abasic site; single-strand breaks; double strand breaks; and intrastrand (PYRIMIDINE DIMERS) or interstrand crosslinking. Damage can often be repaired (DNA REPAIR). If the damage is extensive, it can induce APOPTOSIS.
A semisynthetic derivative of PODOPHYLLOTOXIN that exhibits antitumor activity. Etoposide inhibits DNA synthesis by forming a complex with topoisomerase II and DNA. This complex induces breaks in double stranded DNA and prevents repair by topoisomerase II binding. Accumulated breaks in DNA prevent entry into the mitotic phase of cell division, and lead to cell death. Etoposide acts primarily in the G2 and S phases of the cell cycle.
Cells grown in vitro from neoplastic tissue. If they can be established as a TUMOR CELL LINE, they can be propagated in cell culture indefinitely.
A bacterial DNA topoisomerase II that catalyzes ATP-dependent breakage of both strands of DNA, passage of the unbroken strands through the breaks, and rejoining of the broken strands. Topoisomerase IV binds to DNA as a heterotetramer consisting 2 parC and 2 parE subunits. Topoisomerase IV is a decatenating enzyme that resolves interlinked daughter chromosomes following DNA replication.
An aminoacridine derivative that intercalates into DNA and is used as an antineoplastic agent.
A group of compounds with the heterocyclic ring structure of benzo(c)pyridine. The ring structure is characteristic of the group of opium alkaloids such as papaverine. (From Stedman, 25th ed)
A deoxyribonucleotide polymer that is the primary genetic material of all cells. Eukaryotic and prokaryotic organisms normally contain DNA in a double-stranded state, yet several important biological processes transiently involve single-stranded regions. DNA, which consists of a polysugar-phosphate backbone possessing projections of purines (adenine and guanine) and pyrimidines (thymine and cytosine), forms a double helix that is held together by hydrogen bonds between these purines and pyrimidines (adenine to thymine and guanine to cytosine).
Compounds based on benzene fused to oxole. They can be formed from methylated CATECHOLS such as EUGENOL.
Benzo-indoles similar to CARBOLINES which are pyrido-indoles. In plants, carbazoles are derived from indole and form some of the INDOLE ALKALOIDS.
Naphthyridines are a class of heterocyclic organic compounds containing a naphthyridine nucleus, which is a polycyclic aromatic hydrocarbon made up of two benzene rings fused to a pyridine ring, and they have been studied for their potential pharmacological properties, including as antimicrobial, antiviral, and anticancer agents.
A cell line derived from cultured tumor cells.
Time schedule for administration of a drug in order to achieve optimum effectiveness and convenience.
The relationship between the dose of an administered drug and the response of the organism to the drug.
Human colonic ADENOCARCINOMA cells that are able to express differentiation features characteristic of mature intestinal cells such as the GOBLET CELLS.
Resistance or diminished response of a neoplasm to an antineoplastic agent in humans, animals, or cell or tissue cultures.
The action of a drug in promoting or enhancing the effectiveness of another drug.
One of the mechanisms by which CELL DEATH occurs (compare with NECROSIS and AUTOPHAGOCYTOSIS). Apoptosis is the mechanism responsible for the physiological deletion of cells and appears to be intrinsically programmed. It is characterized by distinctive morphologic changes in the nucleus and cytoplasm, chromatin cleavage at regularly spaced sites, and the endonucleolytic cleavage of genomic DNA; (DNA FRAGMENTATION); at internucleosomal sites. This mode of cell death serves as a balance to mitosis in regulating the size of animal tissues and in mediating pathologic processes associated with tumor growth.
The span of viability of a cell characterized by the capacity to perform certain functions such as metabolism, growth, reproduction, some form of responsiveness, and adaptability.
Carboxylesterase is a serine-dependent esterase with wide substrate specificity. The enzyme is involved in the detoxification of XENOBIOTICS and the activation of ester and of amide PRODRUGS.
New abnormal growth of tissue. Malignant neoplasms show a greater degree of anaplasia and have the properties of invasion and metastasis, compared to benign neoplasms.
Enzymes that catalyze the transfer of multiple ADP-RIBOSE groups from nicotinamide-adenine dinucleotide (NAD) onto protein targets, thus building up a linear or branched homopolymer of repeating ADP-ribose units i.e., POLY ADENOSINE DIPHOSPHATE RIBOSE.
A botanical insecticide that is an inhibitor of mitochondrial electron transport.
Tumors or cancer of the COLON.
The complex series of phenomena, occurring between the end of one CELL DIVISION and the end of the next, by which cellular material is duplicated and then divided between two daughter cells. The cell cycle includes INTERPHASE, which includes G0 PHASE; G1 PHASE; S PHASE; and G2 PHASE, and CELL DIVISION PHASE.
The long-term (minutes to hours) administration of a fluid into the vein through venipuncture, either by letting the fluid flow by gravity or by pumping it.
Enzymes that regulate the topology of DNA by actions such as breaking, relaxing, passing, and rejoining strands of DNA in cells. These enzymes are important components of the DNA replication system. They are classified by their substrate specificities. DNA TOPOISOMERASE I enzymes act on a single strand of DNA. DNA TOPOISOMERASE II enzymes act on double strands of DNA.
The concentration of a compound needed to reduce population growth of organisms, including eukaryotic cells, by 50% in vitro. Though often expressed to denote in vitro antibacterial activity, it is also used as a benchmark for cytotoxicity to eukaryotic cells in culture.
Mutant mice homozygous for the recessive gene "nude" which fail to develop a thymus. They are useful in tumor studies and studies on immune responses.
The process by which a DNA molecule is duplicated.
Splitting the DNA into shorter pieces by endonucleolytic DNA CLEAVAGE at multiple sites. It includes the internucleosomal DNA fragmentation, which along with chromatin condensation, are considered to be the hallmarks of APOPTOSIS.
The relationship between the chemical structure of a compound and its biological or pharmacological activity. Compounds are often classed together because they have structural characteristics in common including shape, size, stereochemical arrangement, and distribution of functional groups.
An antibiotic compound derived from Streptomyces niveus. It has a chemical structure similar to coumarin. Novobiocin binds to DNA gyrase, and blocks adenosine triphosphatase (ATPase) activity. (From Reynolds, Martindale The Extra Pharmacopoeia, 30th ed, p189)
DNA present in neoplastic tissue.
The reconstruction of a continuous two-stranded DNA molecule without mismatch from a molecule which contained damaged regions. The major repair mechanisms are excision repair, in which defective regions in one strand are excised and resynthesized using the complementary base pairing information in the intact strand; photoreactivation repair, in which the lethal and mutagenic effects of ultraviolet light are eliminated; and post-replication repair, in which the primary lesions are not repaired, but the gaps in one daughter duplex are filled in by incorporation of portions of the other (undamaged) daughter duplex. Excision repair and post-replication repair are sometimes referred to as "dark repair" because they do not require light.
An anaplastic, highly malignant, and usually bronchogenic carcinoma composed of small ovoid cells with scanty neoplasm. It is characterized by a dominant, deeply basophilic nucleus, and absent or indistinct nucleoli. (From Stedman, 25th ed; Holland et al., Cancer Medicine, 3d ed, p1286-7)
Descriptions of specific amino acid, carbohydrate, or nucleotide sequences which have appeared in the published literature and/or are deposited in and maintained by databanks such as GENBANK, European Molecular Biology Laboratory (EMBL), National Biomedical Research Foundation (NBRF), or other sequence repositories.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
In vivo methods of screening investigative anticancer drugs, biologic response modifiers or radiotherapies. Human tumor tissue or cells are transplanted into mice or rats followed by tumor treatment regimens. A variety of outcomes are monitored to assess antitumor effectiveness.
The location of the atoms, groups or ions relative to one another in a molecule, as well as the number, type and location of covalent bonds.
Pyrido-CARBAZOLES originally discovered in the bark of OCHROSIA ELLIPTICA. They inhibit DNA and RNA synthesis and have immunosuppressive properties.
Transplantation between animals of different species.
Models used experimentally or theoretically to study molecular shape, electronic properties, or interactions; includes analogous molecules, computer-generated graphics, and mechanical structures.
The use of two or more chemicals simultaneously or sequentially in the drug therapy of neoplasms. The drugs need not be in the same dosage form.
A decrease in the number of NEUTROPHILS found in the blood.
The fission of a CELL. It includes CYTOKINESIS, when the CYTOPLASM of a cell is divided, and CELL NUCLEUS DIVISION.
Small chromosomal proteins (approx 12-20 kD) possessing an open, unfolded structure and attached to the DNA in cell nuclei by ionic linkages. Classification into the various types (designated histone I, histone II, etc.) is based on the relative amounts of arginine and lysine in each.
Established cell cultures that have the potential to propagate indefinitely.
Proteins, glycoprotein, or lipoprotein moieties on surfaces of tumor cells that are usually identified by monoclonal antibodies. Many of these are of either embryonic or viral origin.
Proteins which bind to DNA. The family includes proteins which bind to both double- and single-stranded DNA and also includes specific DNA binding proteins in serum which can be used as markers for malignant diseases.
The spatial arrangement of the atoms of a nucleic acid or polynucleotide that results in its characteristic 3-dimensional shape.
Nuclear phosphoprotein encoded by the p53 gene (GENES, P53) whose normal function is to control CELL PROLIFERATION and APOPTOSIS. A mutant or absent p53 protein has been found in LEUKEMIA; OSTEOSARCOMA; LUNG CANCER; and COLORECTAL CANCER.
CIRCULAR DNA that is interlaced together as links in a chain. It is used as an assay for the activity of DNA TOPOISOMERASES. Catenated DNA is attached loop to loop in contrast to CONCATENATED DNA which is attached end to end.
An antimitotic agent with immunosuppressive properties.
Tumors or cancer of the LUNG.
Extrachromosomal, usually CIRCULAR DNA molecules that are self-replicating and transferable from one organism to another. They are found in a variety of bacterial, archaeal, fungal, algal, and plant species. They are used in GENETIC ENGINEERING as CLONING VECTORS.
A flavoprotein and iron sulfur-containing oxidoreductase complex that catalyzes the conversion of UBIQUINONE to ubiquinol. In MITOCHONDRIA the complex also couples its reaction to the transport of PROTONS across the internal mitochondrial membrane. The NADH DEHYDROGENASE component of the complex can be isolated and is listed as EC 1.6.99.3.
Any detectable and heritable change in the genetic material that causes a change in the GENOTYPE and which is transmitted to daughter cells and to succeeding generations.
A lignan (LIGNANS) found in PODOPHYLLIN resin from the roots of PODOPHYLLUM plants. It is a potent spindle poison, toxic if taken internally, and has been used as a cathartic. It is very irritating to skin and mucous membranes, has keratolytic actions, has been used to treat warts and keratoses, and may have antineoplastic properties, as do some of its congeners and derivatives.
A species of gram-negative, facultatively anaerobic, rod-shaped bacteria (GRAM-NEGATIVE FACULTATIVELY ANAEROBIC RODS) commonly found in the lower part of the intestine of warm-blooded animals. It is usually nonpathogenic, but some strains are known to produce DIARRHEA and pyogenic infections. Pathogenic strains (virotypes) are classified by their specific pathogenic mechanisms such as toxins (ENTEROTOXIGENIC ESCHERICHIA COLI), etc.
The rate dynamics in chemical or physical systems.
Proteins whose abnormal expression (gain or loss) are associated with the development, growth, or progression of NEOPLASMS. Some neoplasm proteins are tumor antigens (ANTIGENS, NEOPLASM), i.e. they induce an immune reaction to their tumor. Many neoplasm proteins have been characterized and are used as tumor markers (BIOMARKERS, TUMOR) when they are detectable in cells and body fluids as monitors for the presence or growth of tumors. Abnormal expression of ONCOGENE PROTEINS is involved in neoplastic transformation, whereas the loss of expression of TUMOR SUPPRESSOR PROTEINS is involved with the loss of growth control and progression of the neoplasm.
A bacterial DNA topoisomerase II that catalyzes ATP-dependent breakage of both strands of DNA, passage of the unbroken strands through the breaks, and rejoining of the broken strands. Gyrase binds to DNA as a heterotetramer consisting of two A and two B subunits. In the presence of ATP, gyrase is able to convert the relaxed circular DNA duplex into a superhelix. In the absence of ATP, supercoiled DNA is relaxed by DNA gyrase.
The facilitation of a chemical reaction by material (catalyst) that is not consumed by the reaction.
The parts of a macromolecule that directly participate in its specific combination with another molecule.
Diminished or failed response of an organism, disease or tissue to the intended effectiveness of a chemical or drug. It should be differentiated from DRUG TOLERANCE which is the progressive diminution of the susceptibility of a human or animal to the effects of a drug, as a result of continued administration.
Tumors or cancer of the OVARY. These neoplasms can be benign or malignant. They are classified according to the tissue of origin, such as the surface EPITHELIUM, the stromal endocrine cells, and the totipotent GERM CELLS.
A characteristic feature of enzyme activity in relation to the kind of substrate on which the enzyme or catalytic molecule reacts.

Novel selective inhibitors for human topoisomerase I, BM2419-1 and -2 derived from saintopin. (1/722)

Compounds BM2419-1 and -2 were isolated from a culture broth of a fungus Paecilomyces sp. BM2419. It was shown that these novel compounds were artifacts derived from saintopin, a dual inhibitor of topoisomerase I and II by independent processes. In the human topoisomerase I inhibition assay using the recombinant Saccharomyces cerevisiae, BM2419-1 and -2 inhibited selectively the yeast growth dependent on human topoisomerase I induction with IC50 values of 0.3 ng/ml and 6.0 ng/ml, respectively.  (+info)

Detection of poly(ADP-ribose) polymerase cleavage in response to treatment with topoisomerase I inhibitors: a potential surrogate end point to assess treatment effectiveness. (2/722)

Cleavage of poly(ADP-ribose) polymerase (PARP) by caspases is a prominent characteristic of apoptosis or programmed cell death shown to be induced by topoisomerase (Topo) inhibitors. Because Topo I inhibitors have been shown to be effective in the treatment of some patients with colon cancer, we considered the possibility of using PARP cleavage as an early predictor of responsiveness to this class of agents. We show cleavage of PARP in response to treatment with Topo I inhibitors in colon cancer both in vitro and in vivo: (a) in vitro in SW480, HCT116, VACO5, VACO6, VACO8, VACO411, VACO425, and VACO451 human colon cancer cell lines treated with topotecan (TPT) or CPT-11; (b) in vivo in SW480, VACO451, and VRC5 colon cancer xenografts grown in athymic mice treated with TPT or CPT-11; and (c) in vivo in colon cancer samples from patients undergoing a Phase II clinical trial with CPT-11. Our results show a strong correlation between percentage of PARP cleavage and percentage of acridine orange-positive cells in colon cancer cell lines treated with 0.1 microM TPT for 24 and 48 h, confirming that PARP cleavage is a useful marker for programmed cell death in colon cancer cell lines. Results from experiments performed on colon cancer xenografts also show an association between PARP cleavage and response to treatment with TPT or CPT-11. The increase of PARP cleavage in xenografts and in clinical samples corresponding to treatment with Topo I inhibitors suggests that this procedure may have early predictive value to assess effectiveness of treatment. These results provide the basis for determining the validity of using PARP cleavage as an early marker of chemotherapeutic effectiveness in human samples.  (+info)

Degradation of topoisomerase I induced by topoisomerase I inhibitors is dependent on inhibitor structure but independent of cell death. (3/722)

DNA topoisomerase I (top I) is the target of the antitumor drug camptothecin (CPT) and its analogs. CPT induces dose- and time-dependent degradation of top I. Degradation of top I also occurs in a CPT-resistant cell line and, therefore, is not a consequence of cell death. Top I degradation is preceded by the appearance of a high molecular weight ladder of top I immunoreactivity and can be blocked by specific inhibitors of the proteasome. We compared the effects of five top I poisons [CPT, topotecan, 6-N-formylamino-12,13-dihydro-1, 11-dihydroxy-13-(beta-D-glucopyranosyl)-5H-indolo[2,3-a]pyrrolo[3, 4-c]carbazole-5,7(6H)-dione (NB506), camptothecin-(para)-4beta-amino-4'-O-demethyl Epipodophyllotoxin (W1), and camptothecin-(ortho)-4beta-amino-4'-O-demethyl Epipodophyllotoxin (W2)] on cleavable complex formation and top I degradation. Although all five drugs induced cleavable complex formation, two of the drugs, NB506 and W1 did not induce top I degradation.  (+info)

Inhibition of inducible nitric oxide synthase by beta-lapachone in rat alveolar macrophages and aorta. (4/722)

Beta-lapachone, a plant product, has been shown to be a novel inhibitor of DNA topoisomerase. In this study, we performed experiments to examine the effects of beta-lapachone on lipopolysaccharide (LPS)-induced inducible nitric oxide (NO) synthase (iNOS) in rat alveolar macrophages and aortic rings. In alveolar macrophages, incubation with LPS (10 microg ml(-1)) for various time intervals resulted in a significant increase in nitrite production and iNOS protein synthesis, that was inhibited by coincubation with beta-lapachone (1-4.5 microM) without any cytotoxic effects. However, addition of beta-lapachone after induction of NO synthase by LPS failed to affect the nitrite production. Treatment with LPS (10 microg ml(-1)) for 6 h resulted in significant expression of mRNA for iNOS which was significantly inhibited in the presence of beta-lapachone (3 microM) in alveolar macrophages. In endothelium-intact rings of thoracic aorta, beta-lapachone (1 and 3 microM) markedly inhibited the hypocontractility to phenylephrine in aortic rings treated with LPS (10 microg ml(-1)) for 4 h. When beta-lapachone was added 3 h after LPS into the medium, the contractions evoked by phenylephrine were not significantly different in the presence or absence of beta-lapachone. Treatment with LPS (10 microg ml(-1)) for 4 h resulted in a significant increase in iNOS protein synthesis which was inhibited in the presence of beta-lapachone (3 microM), but did not affect the constitutive (endothelial and neuronal) NOS forms in aortic rings. These results indicate that beta-lapachone is capable of inhibiting expression and function of iNOS in rat alveolar macrophages and aortic rings. It is considered that beta-lapachone can be developed as a potential anti-inflammatory agent in the future.  (+info)

Bax-alpha promotes apoptosis induced by cancer chemotherapy and accelerates the activation of caspase 3-like cysteine proteases in p53 double mutant B lymphoma Namalwa cells. (5/722)

Stable transfected human p53 (mt/mt) B lymphoma Namalwa variant lines showing differential expression of the Bax-alpha protein were derived under hygromycin selection. Overexpression of Bax-alpha in these variant cells accelerates cell death induced by short or continuous treatments with various concentrations of camptothecin, etoposide, vinblastine and shows no accelerating cell death activity in cis-platinum and paclitaxel-treated cells. Activation of apoptosis and oligonucleosome-sized DNA fragmentation was observed in the variant lines with more pronounced effect in cells containing high level of Bax-alpha protein. These results suggest that increased cell death mediated by anticancer drugs correlates with Bax-alpha level of expression and that Bax-alpha sensitizes Namalwa cells treated at low drug concentrations. The extent of DNA synthesis inhibition following DNA topoisomerase inhibitor treatments was similar in control and all transfected Namalwa cells suggesting that Bax-alpha acts downstream of DNA topoisomerase-mediated DNA strand breaks. To define further the relation between Bax-alpha expression and apoptosis activation, kinetics of caspase activation was measured in drug-treated cells. Caspase activities were measured using specific fluorogenic peptide derivatives DABCYL-YVADAPV-EDANS and Ac-DEVD-AMC, substrates of the caspase 1-like and caspase 3-like families, respectively. In control and Bax-alpha transfected Namalwa cells no increase in caspase 1-like activity was detected following camptothecin and etoposide treatments. In contrast, a significant difference in Ac-DEVD-AMC hydrolysis activity was observed in Bax-alpha transfected Namalwa cells compared to that of control Namalwa cells after camptothecin and etoposide treatment. Increased caspase 3-like activity correlated also with poly(ADPribosyl) polymerase cleavage. Taken together, these results suggest that Bax-alpha sensitize B lymphoma cells to series of anticancer drugs and accelerates the activation of apoptotic protease cascade.  (+info)

Escherichia coli DNA topoisomerase I copurifies with Tn5 transposase, and Tn5 transposase inhibits topoisomerase I. (6/722)

Tn5 transposase (Tnp) overproduction is lethal to Escherichia coli. Genetic evidence suggested that this killing involves titration of E. coli topoisomerase I (Topo I). Here, we present biochemical evidence that supports this model. Tn5 Tnp copurifies with Topo I while nonkilling derivatives of Tnp, Delta37Tnp and Delta55Tnp (Inhibitor [Inh]), show reduced affinity or no affinity, respectively, for Topo I. In agreement with these results, the presence of Tnp, but not Delta37 or Inh derivatives of Tnp, inhibits the DNA relaxation activity of Topo I in vivo as well as in vitro. Other proteins, including RNA polymerase, are also found to copurify with Tnp. For RNA polymerase, reduced copurification with Tnp is observed in extracts from a topA mutant strain, suggesting that RNA polymerase interacts with Topo I and not Tnp.  (+info)

Calories from carbohydrates: energetic contribution of the carbohydrate moiety of rebeccamycin to DNA binding and the effect of its orientation on topoisomerase I inhibition. (7/722)

BACKGROUND: Only a few antitumor drugs inhibit the DNA breakage-reunion reaction catalyzed by topoisomerase. One is the camptothecin derivative topotecan that has recently been used clinically. Others are the glycosylated antibiotic rebeccamycin and its synthetic analog NB-506, which is presently in phase I of clinical trials. Unlike the camptothecins, rebeccamycin-type compounds bind to DNA. We set out to elucidate the molecular basis of their interaction with duplex DNA, with particular emphasis on the role of the carbohydrate residue. RESULTS: We compared the DNA-binding and topoisomerase-I-inhibition activities of two isomers of rebeccamycin that contain a galactose residue attached to the indolocarbazole chromophore via an alpha (axial) or a beta (equatorial) glycosidic linkage. The modification of the stereochemistry of the chromophore-sugar linkage results in a marked change of the DNA-binding and topoisomerase-I- poisoning activities. The inverted configuration at the C-1' of the carbohydrate residue abolishes intercalative binding of the drug to DNA thereby drastically reducing the binding affinity. Consequently, the alpha isomer has lost the capacity to induce topoisomerase-I-mediated cleavage of DNA. Comparison with the aglycone allowed us to determine the energetic contribution of the sugar residue. CONCLUSIONS: The optimal interaction of rebeccamycin analogs with DNA is controlled to a large extent by the stereochemistry of the sugar residue. The results clarify the role of carbohydrates in stereospecific drug-DNA interactions and provide valuable information for the rational design of new rebeccamycin-type antitumor agents.  (+info)

The ATM protein is required for sustained activation of NF-kappaB following DNA damage. (8/722)

Cells lacking an intact ATM gene are hypersensitive to ionizing radiation and show multiple defects in the cell cycle-coupled checkpoints. DNA damage usually triggers cell cycle arrest through, among other things, the activation of p53. Another DNA-damage responsive factor is NF-kappaB. It is activated by various stress situations, including oxidative stress, and by DNA-damaging compounds such as topoisomerase poisons. We found that cells from Ataxia Telangiectasia patients exhibit a defect in NF-kappaB activation in response to treatment with camptothecin, a topoisomerase I poison. In AT cells, this activation is shortened or suppressed, compared to that observed in normal cells. Ectopic expression of the ATM protein in AT cells increases the activation of NF-kappaB in response to camptothecin. MO59J glioblastoma cells that do not express the DNA-PK catalytic subunit respond normally to camptothecin. These results support the hypothesis that NF-kappaB is a DNA damage-responsive transcription factor and that its activation pathway by DNA damage shares some components with the one leading to p53 activation.  (+info)

Topoisomerase I inhibitors are a class of anticancer drugs that work by inhibiting the function of topoisomerase I, an enzyme that plays a crucial role in the relaxation and replication of DNA. By inhibiting this enzyme's activity, these drugs interfere with the normal unwinding and separation of DNA strands, leading to DNA damage and ultimately cell death. Topoisomerase I inhibitors are used in the treatment of various types of cancer, including colon, small cell lung, ovarian, and cervical cancers. Examples of topoisomerase I inhibitors include camptothecin, irinotecan, and topotecan.

DNA topoisomerases are enzymes that modify the topological structure of DNA by regulating the number of twists or supercoils in the double helix. There are two main types of DNA topoisomerases: type I and type II.

Type I DNA topoisomerases function by cutting one strand of the DNA duplex, allowing the uncut strand to rotate around the break, and then resealing the break. This process can relieve both positive and negative supercoiling in DNA, as well as introduce single-stranded breaks into the DNA molecule.

Type I topoisomerases are further divided into three subtypes: type IA, type IB, and type IC. These subtypes differ in their mechanism of action and the structure of the active site tyrosine residue that makes the transient break in the DNA strand.

Overall, DNA topoisomerases play a crucial role in many cellular processes involving DNA, including replication, transcription, recombination, and chromosome segregation. Dysregulation of these enzymes has been implicated in various human diseases, including cancer and genetic disorders.

Camptothecin is a topoisomerase I inhibitor, which is a type of chemotherapeutic agent used in cancer treatment. It works by interfering with the function of an enzyme called topoisomerase I, which helps to uncoil DNA during cell division. By inhibiting this enzyme, camptothecin prevents the cancer cells from dividing and growing, ultimately leading to their death.

Camptothecin is found naturally in the bark and stem of the Camptotheca acuminata tree, also known as the "happy tree," which is native to China. It was first isolated in 1966 and has since been developed into several synthetic derivatives, including irinotecan and topotecan, which are used clinically to treat various types of cancer, such as colon, lung, and ovarian cancers.

Like other chemotherapeutic agents, camptothecin can have significant side effects, including nausea, vomiting, diarrhea, and myelosuppression (suppression of bone marrow function). It is important for patients receiving camptothecin-based therapies to be closely monitored by their healthcare team to manage these side effects effectively.

Topoisomerase II inhibitors are a class of anticancer drugs that work by interfering with the enzyme topoisomerase II, which is essential for DNA replication and transcription. These inhibitors bind to the enzyme-DNA complex, preventing the relaxation of supercoiled DNA and causing DNA strand breaks. This results in the accumulation of double-stranded DNA breaks, which can lead to apoptosis (programmed cell death) in rapidly dividing cells, such as cancer cells. Examples of topoisomerase II inhibitors include etoposide, doxorubicin, and mitoxantrone.

Topotecan is a chemotherapeutic agent, specifically a topoisomerase I inhibitor. It is a semi-synthetic derivative of camptothecin and works by interfering with the function of topoisomerase I, an enzyme that helps to relax supercoiled DNA during transcription and replication. By inhibiting this enzyme, topotecan causes DNA damage and apoptosis (programmed cell death) in rapidly dividing cells, such as cancer cells. It is used in the treatment of various types of cancer, including small cell lung cancer and ovarian cancer.

DNA topoisomerases are enzymes that regulate the topological state of DNA during various cellular processes such as replication, transcription, and repair. They do this by introducing temporary breaks in the DNA strands and allowing the strands to rotate around each other, thereby relieving torsional stress and supercoiling. Topoisomerases are classified into two types: type I and type II.

Type II topoisomerases are further divided into two subtypes: type IIA and type IIB. These enzymes function by forming a covalent bond with the DNA strands, cleaving them, and then passing another segment of DNA through the break before resealing the original strands. This process allows for the removal of both positive and negative supercoils from DNA as well as the separation of interlinked circular DNA molecules (catenanes) or knotted DNA structures.

Type II topoisomerases are essential for cell viability, and their dysfunction has been linked to various human diseases, including cancer and neurodegenerative disorders. They have also emerged as important targets for the development of anticancer drugs that inhibit their activity and induce DNA damage leading to cell death. Examples of type II topoisomerase inhibitors include etoposide, doxorubicin, and mitoxantrone.

Topoisomerase inhibitors are a class of anticancer drugs that work by interfering with the function of topoisomerases, which are enzymes responsible for relaxing supercoiled DNA during processes such as replication and transcription. Topoisomerase I inhibitors selectively bind to and stabilize the cleavage complex formed between topoisomerase I and DNA, preventing the relegation of the broken DNA strand and resulting in DNA damage and cell death. Examples include irinotecan and topotecan. Topoisomerase II inhibitors, on the other hand, bind to and stabilize the cleavage complex formed between topoisomerase II and DNA, leading to double-stranded DNA breaks and cell death. Examples include doxorubicin, etoposide, and mitoxantrone. These drugs are used in the treatment of various types of cancer.

Antineoplastic agents, phytogenic, also known as plant-derived anticancer drugs, are medications that are derived from plants and used to treat cancer. These agents have natural origins and work by interfering with the growth and multiplication of cancer cells, helping to slow or stop the spread of the disease. Some examples of antineoplastic agents, phytogenic include paclitaxel (Taxol), vincristine, vinblastine, and etoposide. These drugs are often used in combination with other treatments such as surgery, radiation therapy, and other medications to provide a comprehensive approach to cancer care.

I'm sorry for any confusion, but "Indenes" is not a recognized medical term or concept in the field of medicine or healthcare. It may be that there is a spelling mistake or typo in your question. If you are referring to "Indenes" as a chemical compound, it is a polycyclic aromatic hydrocarbon (PAH) with the molecular formula C9H8. However, I would recommend consulting a chemistry or toxicology resource for information on its non-medical uses and properties.

Teniposide is a synthetic podophyllotoxin derivative, which is an antineoplastic agent. It works by interfering with the DNA synthesis and function of cancer cells, leading to cell cycle arrest and apoptosis (programmed cell death). Teniposide is primarily used in the treatment of acute lymphoblastic leukemia (ALL) and other malignancies in children. It is often administered through intravenous infusion and is typically used in combination with other chemotherapeutic agents.

The medical definition of Teniposide can be stated as:

Teniposide, chemically known as (4'-demethylepipodophyllotoxin 9-[4,6-O-(R)-benzylidene-α-L-glucopyranoside]), is a semi-synthetic podophyllotoxin derivative with antineoplastic activity. It inhibits DNA topoisomerase II, leading to the formation of DNA-topoisomerase II cleavable complexes, G2 arrest, and apoptosis in cancer cells. Teniposide is primarily used in the treatment of acute lymphoblastic leukemia (ALL) and other malignancies in children, often administered through intravenous infusion and typically used in combination with other chemotherapeutic agents.

Enzyme inhibitors are substances that bind to an enzyme and decrease its activity, preventing it from catalyzing a chemical reaction in the body. They can work by several mechanisms, including blocking the active site where the substrate binds, or binding to another site on the enzyme to change its shape and prevent substrate binding. Enzyme inhibitors are often used as drugs to treat various medical conditions, such as high blood pressure, abnormal heart rhythms, and bacterial infections. They can also be found naturally in some foods and plants, and can be used in research to understand enzyme function and regulation.

Antineoplastic agents are a class of drugs used to treat malignant neoplasms or cancer. These agents work by inhibiting the growth and proliferation of cancer cells, either by killing them or preventing their division and replication. Antineoplastic agents can be classified based on their mechanism of action, such as alkylating agents, antimetabolites, topoisomerase inhibitors, mitotic inhibitors, and targeted therapy agents.

Alkylating agents work by adding alkyl groups to DNA, which can cause cross-linking of DNA strands and ultimately lead to cell death. Antimetabolites interfere with the metabolic processes necessary for DNA synthesis and replication, while topoisomerase inhibitors prevent the relaxation of supercoiled DNA during replication. Mitotic inhibitors disrupt the normal functioning of the mitotic spindle, which is essential for cell division. Targeted therapy agents are designed to target specific molecular abnormalities in cancer cells, such as mutated oncogenes or dysregulated signaling pathways.

It's important to note that antineoplastic agents can also affect normal cells and tissues, leading to various side effects such as nausea, vomiting, hair loss, and myelosuppression (suppression of bone marrow function). Therefore, the use of these drugs requires careful monitoring and management of their potential adverse effects.

Drug screening assays for antitumor agents are laboratory tests used to identify and evaluate the effectiveness of potential drugs or compounds that can inhibit the growth of tumor cells or induce their death. These assays are typically performed in vitro (in a test tube or petri dish) using cell cultures of various types of cancer cells.

The assays measure different parameters such as cell viability, proliferation, apoptosis (programmed cell death), and cytotoxicity to determine the ability of the drug to kill or inhibit the growth of tumor cells. The results of these assays can help researchers identify promising antitumor agents that can be further developed for clinical use in cancer treatment.

There are different types of drug screening assays for antitumor agents, including high-throughput screening (HTS) assays, which allow for the rapid and automated testing of a large number of compounds against various cancer cell lines. Other types of assays include phenotypic screening assays, target-based screening assays, and functional screening assays, each with its own advantages and limitations.

Overall, drug screening assays for antitumor agents play a critical role in the development of new cancer therapies by providing valuable information on the activity and safety of potential drugs, helping to identify effective treatments and reduce the time and cost associated with bringing new drugs to market.

DNA cleavage is the breaking of the phosphodiester bonds in the DNA molecule, resulting in the separation of the two strands of the double helix. This process can occur through chemical or enzymatic reactions and can result in various types of damage to the DNA molecule, including single-strand breaks, double-strand breaks, and base modifications.

Enzymatic DNA cleavage is typically carried out by endonucleases, which are enzymes that cut DNA molecules at specific sequences or structures. There are two main types of endonucleases: restriction endonucleases and repair endonucleases. Restriction endonucleases recognize and cleave specific DNA sequences, often used in molecular biology techniques such as genetic engineering and cloning. Repair endonucleases, on the other hand, are involved in DNA repair processes and recognize and cleave damaged or abnormal DNA structures.

Chemical DNA cleavage can occur through various mechanisms, including oxidation, alkylation, or hydrolysis of the phosphodiester bonds. Chemical agents such as hydrogen peroxide, formaldehyde, or hydrazine can induce chemical DNA cleavage and are often used in laboratory settings for various purposes, such as DNA fragmentation or labeling.

Overall, DNA cleavage is an essential process in many biological functions, including DNA replication, repair, and recombination. However, excessive or improper DNA cleavage can lead to genomic instability, mutations, and cell death.

Superhelical DNA refers to a type of DNA structure that is formed when the double helix is twisted around itself. This occurs due to the presence of negative supercoiling, which results in an overtwisted state that can be described as having a greater number of helical turns than a relaxed circular DNA molecule.

Superhelical DNA is often found in bacterial and viral genomes, where it plays important roles in compacting the genome into a smaller volume and facilitating processes such as replication and transcription. The degree of supercoiling can affect the structure and function of DNA, with varying levels of supercoiling influencing the accessibility of specific regions of the genome to proteins and other regulatory factors.

Superhelical DNA is typically maintained in a stable state by topoisomerase enzymes, which introduce or remove twists in the double helix to regulate its supercoiling level. Changes in supercoiling can have significant consequences for cellular processes, as they can impact the expression of genes and the regulation of chromosome structure and function.

Intercalating agents are chemical substances that can be inserted between the stacked bases of DNA, creating a separation or "intercalation" of the base pairs. This property is often exploited in cancer chemotherapy, where intercalating agents like doxorubicin and daunorubicin are used to inhibit the replication and transcription of cancer cells by preventing the normal functioning of their DNA. However, these agents can also have toxic effects on normal cells, particularly those that divide rapidly, such as bone marrow and gut epithelial cells. Therefore, their use must be carefully monitored and balanced against their therapeutic benefits.

DNA damage refers to any alteration in the structure or composition of deoxyribonucleic acid (DNA), which is the genetic material present in cells. DNA damage can result from various internal and external factors, including environmental exposures such as ultraviolet radiation, tobacco smoke, and certain chemicals, as well as normal cellular processes such as replication and oxidative metabolism.

Examples of DNA damage include base modifications, base deletions or insertions, single-strand breaks, double-strand breaks, and crosslinks between the two strands of the DNA helix. These types of damage can lead to mutations, genomic instability, and chromosomal aberrations, which can contribute to the development of diseases such as cancer, neurodegenerative disorders, and aging-related conditions.

The body has several mechanisms for repairing DNA damage, including base excision repair, nucleotide excision repair, mismatch repair, and double-strand break repair. However, if the damage is too extensive or the repair mechanisms are impaired, the cell may undergo apoptosis (programmed cell death) to prevent the propagation of potentially harmful mutations.

Etoposide is a chemotherapy medication used to treat various types of cancer, including lung cancer, testicular cancer, and certain types of leukemia. It works by inhibiting the activity of an enzyme called topoisomerase II, which is involved in DNA replication and transcription. By doing so, etoposide can interfere with the growth and multiplication of cancer cells.

Etoposide is often administered intravenously in a hospital or clinic setting, although it may also be given orally in some cases. The medication can cause a range of side effects, including nausea, vomiting, hair loss, and an increased risk of infection. It can also have more serious side effects, such as bone marrow suppression, which can lead to anemia, bleeding, and a weakened immune system.

Like all chemotherapy drugs, etoposide is not without risks and should only be used under the close supervision of a qualified healthcare provider. It is important for patients to discuss the potential benefits and risks of this medication with their doctor before starting treatment.

'Tumor cells, cultured' refers to the process of removing cancerous cells from a tumor and growing them in controlled laboratory conditions. This is typically done by isolating the tumor cells from a patient's tissue sample, then placing them in a nutrient-rich environment that promotes their growth and multiplication.

The resulting cultured tumor cells can be used for various research purposes, including the study of cancer biology, drug development, and toxicity testing. They provide a valuable tool for researchers to better understand the behavior and characteristics of cancer cells outside of the human body, which can lead to the development of more effective cancer treatments.

It is important to note that cultured tumor cells may not always behave exactly the same way as they do in the human body, so findings from cell culture studies must be validated through further research, such as animal models or clinical trials.

DNA Topoisomerase IV is a type of enzyme that plays a crucial role in the relaxation and manipulation of supercoiled DNA during processes such as replication, transcription, and chromosome segregation. It functions by temporarily cleaving and rejoining the DNA strands to allow for the unlinking and separation of DNA molecules. This enzyme primarily targets double-stranded DNA and is especially important in bacteria, where it helps to resolve the topological challenges that arise during DNA replication and segregation of daughter chromosomes during cell division. Inhibition of DNA Topoisomerase IV has been explored as a strategy for developing antibacterial drugs, as this enzyme is essential for bacterial survival and is not found in humans.

Amsacrine is a chemotherapeutic agent, which means it is a medication used to treat cancer. It is classified as an antineoplastic drug, and more specifically, as an intercalating agent and a topoisomerase II inhibitor. Amsacrine works by intercalating, or inserting itself, into the DNA of cancer cells, which prevents the DNA from replicating and ultimately leads to the death of the cancer cell. It is primarily used in the treatment of acute myeloid leukemia (AML) and other hematologic malignancies.

The chemical name for Amsacrine is 5-[3-amino-1-(3-aminopropyl)-2-hydroxybut-1-yloxy]-8-chloro-1,4-naphthoquinone. It has a molecular formula of C16H17ClNO5 and a molecular weight of 359.8 g/mol.

Amsacrine is typically administered intravenously, and its use is usually reserved for patients who have not responded to other forms of chemotherapy. It may be used in combination with other anticancer drugs as part of a treatment regimen. As with any chemotherapeutic agent, Amsacrine can have significant side effects, including nausea, vomiting, and hair loss. It can also cause damage to the heart and other organs, so it is important for patients to be closely monitored during treatment.

It's worth noting that while Amsacrine can be an effective treatment for some types of cancer, it is not a cure-all, and its use must be carefully considered in the context of each individual patient's medical history and current health status.

Isoquinolines are not a medical term per se, but a chemical classification. They refer to a class of organic compounds that consist of a benzene ring fused to a piperidine ring. This structure is similar to that of quinoline, but with the nitrogen atom located at a different position in the ring.

Isoquinolines have various biological activities and can be found in some natural products, including certain alkaloids. Some isoquinoline derivatives have been developed as drugs for the treatment of various conditions, such as cardiovascular diseases, neurological disorders, and cancer. However, specific medical definitions related to isoquinolines typically refer to the use or effects of these specific drugs rather than the broader class of compounds.

Deoxyribonucleic acid (DNA) is the genetic material present in the cells of organisms where it is responsible for the storage and transmission of hereditary information. DNA is a long molecule that consists of two strands coiled together to form a double helix. Each strand is made up of a series of four nucleotide bases - adenine (A), guanine (G), cytosine (C), and thymine (T) - that are linked together by phosphate and sugar groups. The sequence of these bases along the length of the molecule encodes genetic information, with A always pairing with T and C always pairing with G. This base-pairing allows for the replication and transcription of DNA, which are essential processes in the functioning and reproduction of all living organisms.

Benzodioxoles are chemical compounds that consist of a benzene ring (a six-carbon cyclic structure with alternating double bonds) linked to two oxide groups through methane bridges. They can be found naturally in some plants, such as nutmeg and tea, but they are also synthesized for use in various pharmaceuticals and illicit drugs.

In the medical field, benzodioxoles are used in the synthesis of certain drugs, including some antimicrobials, antihelmintics (drugs that treat parasitic worm infections), and muscle relaxants. However, they are perhaps best known for their use as a structural component in certain illicit drugs, such as ecstasy (MDMA) and related substances.

It's important to note that while benzodioxoles themselves may have some medical uses, many of the drugs that contain this structure can be dangerous when used improperly or without medical supervision.

Carbazoles are aromatic organic compounds that consist of a tricyclic structure with two benzene rings fused to a five-membered ring containing two nitrogen atoms. The chemical formula for carbazole is C12H9N. Carbazoles are found in various natural sources, including coal tar and certain plants. They also have various industrial applications, such as in the production of dyes, pigments, and pharmaceuticals. In a medical context, carbazoles are not typically referred to as a single entity but rather as a class of compounds with potential therapeutic activity. Some carbazole derivatives have been studied for their anti-cancer, anti-inflammatory, and anti-microbial properties.

Naphthyridines are a class of heterocyclic organic compounds that contain a naphthyridine core structure, which is a polycyclic aromatic hydrocarbon made up of two benzene rings fused to a tetrahydropyridine ring. They have a variety of pharmacological activities and are used in the development of various therapeutic agents, including antibiotics, antivirals, and anticancer drugs.

In medical terms, naphthyridines do not have a specific clinical definition or application, but they are rather a chemical class that is utilized in the design and synthesis of drugs with potential therapeutic benefits. The unique structure and properties of naphthyridines make them attractive candidates for drug development, particularly in areas where new treatments are needed to overcome drug resistance or improve efficacy.

It's worth noting that while naphthyridines have shown promise in preclinical studies, further research is needed to fully understand their safety and effectiveness in humans before they can be approved as therapeutic agents.

A cell line that is derived from tumor cells and has been adapted to grow in culture. These cell lines are often used in research to study the characteristics of cancer cells, including their growth patterns, genetic changes, and responses to various treatments. They can be established from many different types of tumors, such as carcinomas, sarcomas, and leukemias. Once established, these cell lines can be grown and maintained indefinitely in the laboratory, allowing researchers to conduct experiments and studies that would not be feasible using primary tumor cells. It is important to note that tumor cell lines may not always accurately represent the behavior of the original tumor, as they can undergo genetic changes during their time in culture.

A "Drug Administration Schedule" refers to the plan for when and how a medication should be given to a patient. It includes details such as the dose, frequency (how often it should be taken), route (how it should be administered, such as orally, intravenously, etc.), and duration (how long it should be taken) of the medication. This schedule is often created and prescribed by healthcare professionals, such as doctors or pharmacists, to ensure that the medication is taken safely and effectively. It may also include instructions for missed doses or changes in the dosage.

A dose-response relationship in the context of drugs refers to the changes in the effects or symptoms that occur as the dose of a drug is increased or decreased. Generally, as the dose of a drug is increased, the severity or intensity of its effects also increases. Conversely, as the dose is decreased, the effects of the drug become less severe or may disappear altogether.

The dose-response relationship is an important concept in pharmacology and toxicology because it helps to establish the safe and effective dosage range for a drug. By understanding how changes in the dose of a drug affect its therapeutic and adverse effects, healthcare providers can optimize treatment plans for their patients while minimizing the risk of harm.

The dose-response relationship is typically depicted as a curve that shows the relationship between the dose of a drug and its effect. The shape of the curve may vary depending on the drug and the specific effect being measured. Some drugs may have a steep dose-response curve, meaning that small changes in the dose can result in large differences in the effect. Other drugs may have a more gradual dose-response curve, where larger changes in the dose are needed to produce significant effects.

In addition to helping establish safe and effective dosages, the dose-response relationship is also used to evaluate the potential therapeutic benefits and risks of new drugs during clinical trials. By systematically testing different doses of a drug in controlled studies, researchers can identify the optimal dosage range for the drug and assess its safety and efficacy.

HT-29 is a human colon adenocarcinoma cell line that is commonly used in research. These cells are derived from a colorectal cancer tumor and have the ability to differentiate into various cell types found in the intestinal mucosa, such as absorptive enterocytes and mucus-secreting goblet cells. HT-29 cells are often used to study the biology of colon cancer, including the effects of drugs on cancer cell growth and survival, as well as the role of various genes and signaling pathways in colorectal tumorigenesis.

It is important to note that when working with cell lines like HT-29, it is essential to use proper laboratory techniques and follow established protocols to ensure the integrity and reproducibility of experimental results. Additionally, researchers should regularly authenticate their cell lines to confirm their identity and verify that they are free from contamination with other cell types.

Drug resistance in neoplasms (also known as cancer drug resistance) refers to the ability of cancer cells to withstand the effects of chemotherapeutic agents or medications designed to kill or inhibit the growth of cancer cells. This can occur due to various mechanisms, including changes in the cancer cell's genetic makeup, alterations in drug targets, increased activity of drug efflux pumps, and activation of survival pathways.

Drug resistance can be intrinsic (present at the beginning of treatment) or acquired (developed during the course of treatment). It is a significant challenge in cancer therapy as it often leads to reduced treatment effectiveness, disease progression, and poor patient outcomes. Strategies to overcome drug resistance include the use of combination therapies, development of new drugs that target different mechanisms, and personalized medicine approaches that consider individual patient and tumor characteristics.

Drug synergism is a pharmacological concept that refers to the interaction between two or more drugs, where the combined effect of the drugs is greater than the sum of their individual effects. This means that when these drugs are administered together, they produce an enhanced therapeutic response compared to when they are given separately.

Drug synergism can occur through various mechanisms, such as:

1. Pharmacodynamic synergism - When two or more drugs interact with the same target site in the body and enhance each other's effects.
2. Pharmacokinetic synergism - When one drug affects the metabolism, absorption, distribution, or excretion of another drug, leading to an increased concentration of the second drug in the body and enhanced therapeutic effect.
3. Physiochemical synergism - When two drugs interact physically, such as when one drug enhances the solubility or permeability of another drug, leading to improved absorption and bioavailability.

It is important to note that while drug synergism can result in enhanced therapeutic effects, it can also increase the risk of adverse reactions and toxicity. Therefore, healthcare providers must carefully consider the potential benefits and risks when prescribing combinations of drugs with known or potential synergistic effects.

Apoptosis is a programmed and controlled cell death process that occurs in multicellular organisms. It is a natural process that helps maintain tissue homeostasis by eliminating damaged, infected, or unwanted cells. During apoptosis, the cell undergoes a series of morphological changes, including cell shrinkage, chromatin condensation, and fragmentation into membrane-bound vesicles called apoptotic bodies. These bodies are then recognized and engulfed by neighboring cells or phagocytic cells, preventing an inflammatory response. Apoptosis is regulated by a complex network of intracellular signaling pathways that involve proteins such as caspases, Bcl-2 family members, and inhibitors of apoptosis (IAPs).

Cell survival refers to the ability of a cell to continue living and functioning normally, despite being exposed to potentially harmful conditions or treatments. This can include exposure to toxins, radiation, chemotherapeutic drugs, or other stressors that can damage cells or interfere with their normal processes.

In scientific research, measures of cell survival are often used to evaluate the effectiveness of various therapies or treatments. For example, researchers may expose cells to a particular drug or treatment and then measure the percentage of cells that survive to assess its potential therapeutic value. Similarly, in toxicology studies, measures of cell survival can help to determine the safety of various chemicals or substances.

It's important to note that cell survival is not the same as cell proliferation, which refers to the ability of cells to divide and multiply. While some treatments may promote cell survival, they may also inhibit cell proliferation, making them useful for treating diseases such as cancer. Conversely, other treatments may be designed to specifically target and kill cancer cells, even if it means sacrificing some healthy cells in the process.

Carboxylesterase is a type of enzyme that catalyzes the hydrolysis of ester bonds in carboxylic acid esters, producing alcohol and carboxylate products. These enzymes are widely distributed in various tissues, including the liver, intestines, and plasma. They play important roles in detoxification, metabolism, and the breakdown of xenobiotics (foreign substances) in the body.

Carboxylesterases can also catalyze the reverse reaction, forming esters from alcohols and carboxylates, which is known as transesterification or esterification. This activity has applications in industrial processes and biotechnology.

There are several families of carboxylesterases, with different substrate specificities, kinetic properties, and tissue distributions. These enzymes have been studied for their potential use in therapeutics, diagnostics, and drug delivery systems.

Neoplasms are abnormal growths of cells or tissues in the body that serve no physiological function. They can be benign (non-cancerous) or malignant (cancerous). Benign neoplasms are typically slow growing and do not spread to other parts of the body, while malignant neoplasms are aggressive, invasive, and can metastasize to distant sites.

Neoplasms occur when there is a dysregulation in the normal process of cell division and differentiation, leading to uncontrolled growth and accumulation of cells. This can result from genetic mutations or other factors such as viral infections, environmental exposures, or hormonal imbalances.

Neoplasms can develop in any organ or tissue of the body and can cause various symptoms depending on their size, location, and type. Treatment options for neoplasms include surgery, radiation therapy, chemotherapy, immunotherapy, and targeted therapy, among others.

Rotenone is not strictly a medical term, but it is a pesticide that is used in some medical situations. According to the National Pesticide Information Center, rotenone is a pesticide derived from the roots and stems of several plants, including Derris Eliptica, Lonchocarpus utilis, and Tephrosia vogelii. It is used as a pesticide to control insects, mites, and fish in both agricultural and residential settings.

In medical contexts, rotenone has been studied for its potential effects on human health, particularly in relation to Parkinson's disease. Some research suggests that exposure to rotenone may increase the risk of developing Parkinson's disease, although more studies are needed to confirm this link. Rotenone works by inhibiting the mitochondria in cells, which can lead to cell death and neurodegeneration.

It is important to note that rotenone is highly toxic and should be handled with care. It can cause skin and eye irritation, respiratory problems, and gastrointestinal symptoms if ingested or inhaled. Therefore, it is recommended to use personal protective equipment when handling rotenone and to follow all label instructions carefully.

Colonic neoplasms refer to abnormal growths in the large intestine, also known as the colon. These growths can be benign (non-cancerous) or malignant (cancerous). The two most common types of colonic neoplasms are adenomas and carcinomas.

Adenomas are benign tumors that can develop into cancer over time if left untreated. They are often found during routine colonoscopies and can be removed during the procedure.

Carcinomas, on the other hand, are malignant tumors that invade surrounding tissues and can spread to other parts of the body. Colorectal cancer is the third leading cause of cancer-related deaths in the United States, and colonic neoplasms are a significant risk factor for developing this type of cancer.

Regular screenings for colonic neoplasms are recommended for individuals over the age of 50 or those with a family history of colorectal cancer or other risk factors. Early detection and removal of colonic neoplasms can significantly reduce the risk of developing colorectal cancer.

The cell cycle is a series of events that take place in a cell leading to its division and duplication. It consists of four main phases: G1 phase, S phase, G2 phase, and M phase.

During the G1 phase, the cell grows in size and synthesizes mRNA and proteins in preparation for DNA replication. In the S phase, the cell's DNA is copied, resulting in two complete sets of chromosomes. During the G2 phase, the cell continues to grow and produces more proteins and organelles necessary for cell division.

The M phase is the final stage of the cell cycle and consists of mitosis (nuclear division) and cytokinesis (cytoplasmic division). Mitosis results in two genetically identical daughter nuclei, while cytokinesis divides the cytoplasm and creates two separate daughter cells.

The cell cycle is regulated by various checkpoints that ensure the proper completion of each phase before progressing to the next. These checkpoints help prevent errors in DNA replication and division, which can lead to mutations and cancer.

Intravenous (IV) infusion is a medical procedure in which liquids, such as medications, nutrients, or fluids, are delivered directly into a patient's vein through a needle or a catheter. This route of administration allows for rapid absorption and distribution of the infused substance throughout the body. IV infusions can be used for various purposes, including resuscitation, hydration, nutrition support, medication delivery, and blood product transfusion. The rate and volume of the infusion are carefully controlled to ensure patient safety and efficacy of treatment.

DNA topoisomerases are enzymes that play a crucial role in the regulation of DNA topology, which refers to the three-dimensional arrangement of the DNA molecule. These enzymes control the number of twists or coils in the DNA helix by creating temporary breaks in the strands and allowing them to rotate around each other, thereby relieving the torsional stress that builds up during processes such as replication and transcription.

There are two main types of DNA topoisomerases: type I and type II. Type I enzymes create a single-stranded break in the DNA helix, while type II enzymes create a double-stranded break. Both types of enzymes can change the linking number (Lk) of the DNA molecule, which is a topological invariant that describes the overall degree of twist in the helix.

Type I topoisomerases are further divided into two subtypes: type IA and type IB. Type IA enzymes, such as topo I from Escherichia coli, create a transient break in one DNA strand and then pass the other strand through the break before resealing it. In contrast, type IB enzymes, such as human topo I, create a covalent bond with the 3'-phosphate end of the broken strand and then pass the 5'-end through the break before rejoining the ends.

Type II topoisomerases are also divided into two subtypes: type IIA and type IIB. Type IIA enzymes, such as bacterial topo IV and eukaryotic topo II, create a double-stranded break in the DNA helix and then pass another segment of double-stranded DNA through the break before resealing it. Type IIB enzymes, such as bacterial topo III and eukaryotic topo IIIα and β, create a double-stranded break and then pass a single strand of DNA through the break before resealing it.

DNA topoisomerases are important targets for cancer chemotherapy because they are essential for cell division and can be inhibited by drugs such as doxorubicin, etoposide, and irinotecan. However, these drugs can also have significant side effects, including cardiotoxicity and myelosuppression. Therefore, there is ongoing research to develop new topoisomerase inhibitors with improved efficacy and safety profiles.

Inhibitory Concentration 50 (IC50) is a measure used in pharmacology, toxicology, and virology to describe the potency of a drug or chemical compound. It refers to the concentration needed to reduce the biological or biochemical activity of a given substance by half. Specifically, it is most commonly used in reference to the inhibition of an enzyme or receptor.

In the context of infectious diseases, IC50 values are often used to compare the effectiveness of antiviral drugs against a particular virus. A lower IC50 value indicates that less of the drug is needed to achieve the desired effect, suggesting greater potency and potentially fewer side effects. Conversely, a higher IC50 value suggests that more of the drug is required to achieve the same effect, indicating lower potency.

It's important to note that IC50 values can vary depending on the specific assay or experimental conditions used, so they should be interpreted with caution and in conjunction with other measures of drug efficacy.

"Nude mice" is a term used in the field of laboratory research to describe a strain of mice that have been genetically engineered to lack a functional immune system. Specifically, nude mice lack a thymus gland and have a mutation in the FOXN1 gene, which results in a failure to develop a mature T-cell population. This means that they are unable to mount an effective immune response against foreign substances or organisms.

The name "nude" refers to the fact that these mice also have a lack of functional hair follicles, resulting in a hairless or partially hairless phenotype. This feature is actually a secondary consequence of the same genetic mutation that causes their immune deficiency.

Nude mice are commonly used in research because their weakened immune system makes them an ideal host for transplanted tumors, tissues, and cells from other species, including humans. This allows researchers to study the behavior of these foreign substances in a living organism without the complication of an immune response. However, it's important to note that because nude mice lack a functional immune system, they must be kept in sterile conditions and are more susceptible to infection than normal mice.

DNA replication is the biological process by which DNA makes an identical copy of itself during cell division. It is a fundamental mechanism that allows genetic information to be passed down from one generation of cells to the next. During DNA replication, each strand of the double helix serves as a template for the synthesis of a new complementary strand. This results in the creation of two identical DNA molecules. The enzymes responsible for DNA replication include helicase, which unwinds the double helix, and polymerase, which adds nucleotides to the growing strands.

DNA fragmentation is the breaking of DNA strands into smaller pieces. This process can occur naturally during apoptosis, or programmed cell death, where the DNA is broken down and packaged into apoptotic bodies to be safely eliminated from the body. However, excessive or abnormal DNA fragmentation can also occur due to various factors such as oxidative stress, exposure to genotoxic agents, or certain medical conditions. This can lead to genetic instability, cellular dysfunction, and increased risk of diseases such as cancer. In the context of reproductive medicine, high levels of DNA fragmentation in sperm cells have been linked to male infertility and poor assisted reproductive technology outcomes.

A Structure-Activity Relationship (SAR) in the context of medicinal chemistry and pharmacology refers to the relationship between the chemical structure of a drug or molecule and its biological activity or effect on a target protein, cell, or organism. SAR studies aim to identify patterns and correlations between structural features of a compound and its ability to interact with a specific biological target, leading to a desired therapeutic response or undesired side effects.

By analyzing the SAR, researchers can optimize the chemical structure of lead compounds to enhance their potency, selectivity, safety, and pharmacokinetic properties, ultimately guiding the design and development of novel drugs with improved efficacy and reduced toxicity.

Novobiocin is an antibiotic derived from the actinomycete species Streptomyces niveus. It belongs to the class of drugs known as aminocoumarins, which function by inhibiting bacterial DNA gyrase, thereby preventing DNA replication and transcription. Novobiocin has activity against a narrow range of gram-positive bacteria, including some strains of Staphylococcus aureus (particularly those resistant to penicillin and methicillin), Streptococcus pneumoniae, and certain mycobacteria. It is used primarily in the treatment of serious staphylococcal infections and is administered orally or intravenously.

It's important to note that Novobiocin has been largely replaced by other antibiotics due to its narrow spectrum of activity, potential for drug interactions, and adverse effects. It is not widely used in clinical practice today.

The term "DNA, neoplasm" is not a standard medical term or concept. DNA refers to deoxyribonucleic acid, which is the genetic material present in the cells of living organisms. A neoplasm, on the other hand, is a tumor or growth of abnormal tissue that can be benign (non-cancerous) or malignant (cancerous).

In some contexts, "DNA, neoplasm" may refer to genetic alterations found in cancer cells. These genetic changes can include mutations, amplifications, deletions, or rearrangements of DNA sequences that contribute to the development and progression of cancer. Identifying these genetic abnormalities can help doctors diagnose and treat certain types of cancer more effectively.

However, it's important to note that "DNA, neoplasm" is not a term that would typically be used in medical reports or research papers without further clarification. If you have any specific questions about DNA changes in cancer cells or neoplasms, I would recommend consulting with a healthcare professional or conducting further research on the topic.

DNA repair is the process by which cells identify and correct damage to the DNA molecules that encode their genome. DNA can be damaged by a variety of internal and external factors, such as radiation, chemicals, and metabolic byproducts. If left unrepaired, this damage can lead to mutations, which may in turn lead to cancer and other diseases.

There are several different mechanisms for repairing DNA damage, including:

1. Base excision repair (BER): This process repairs damage to a single base in the DNA molecule. An enzyme called a glycosylase removes the damaged base, leaving a gap that is then filled in by other enzymes.
2. Nucleotide excision repair (NER): This process repairs more severe damage, such as bulky adducts or crosslinks between the two strands of the DNA molecule. An enzyme cuts out a section of the damaged DNA, and the gap is then filled in by other enzymes.
3. Mismatch repair (MMR): This process repairs errors that occur during DNA replication, such as mismatched bases or small insertions or deletions. Specialized enzymes recognize the error and remove a section of the newly synthesized strand, which is then replaced by new nucleotides.
4. Double-strand break repair (DSBR): This process repairs breaks in both strands of the DNA molecule. There are two main pathways for DSBR: non-homologous end joining (NHEJ) and homologous recombination (HR). NHEJ directly rejoins the broken ends, while HR uses a template from a sister chromatid to repair the break.

Overall, DNA repair is a crucial process that helps maintain genome stability and prevent the development of diseases caused by genetic mutations.

Carcinoma, small cell is a type of lung cancer that typically starts in the bronchi (the airways that lead to the lungs). It is called "small cell" because the cancer cells are small and appear round or oval in shape. This type of lung cancer is also sometimes referred to as "oat cell carcinoma" due to the distinctive appearance of the cells, which can resemble oats when viewed under a microscope.

Small cell carcinoma is a particularly aggressive form of lung cancer that tends to spread quickly to other parts of the body. It is strongly associated with smoking and is less common than non-small cell lung cancer (NSCLC), which accounts for about 85% of all lung cancers.

Like other types of lung cancer, small cell carcinoma may not cause any symptoms in its early stages. However, as the tumor grows and spreads, it can cause a variety of symptoms, including coughing, chest pain, shortness of breath, hoarseness, and weight loss. Treatment for small cell carcinoma typically involves a combination of chemotherapy, radiation therapy, and sometimes surgery.

Molecular sequence data refers to the specific arrangement of molecules, most commonly nucleotides in DNA or RNA, or amino acids in proteins, that make up a biological macromolecule. This data is generated through laboratory techniques such as sequencing, and provides information about the exact order of the constituent molecules. This data is crucial in various fields of biology, including genetics, evolution, and molecular biology, allowing for comparisons between different organisms, identification of genetic variations, and studies of gene function and regulation.

A base sequence in the context of molecular biology refers to the specific order of nucleotides in a DNA or RNA molecule. In DNA, these nucleotides are adenine (A), guanine (G), cytosine (C), and thymine (T). In RNA, uracil (U) takes the place of thymine. The base sequence contains genetic information that is transcribed into RNA and ultimately translated into proteins. It is the exact order of these bases that determines the genetic code and thus the function of the DNA or RNA molecule.

A xenograft model antitumor assay is a type of preclinical cancer research study that involves transplanting human tumor cells or tissues into an immunodeficient mouse. This model allows researchers to study the effects of various treatments, such as drugs or immune therapies, on human tumors in a living organism.

In this assay, human tumor cells or tissues are implanted into the mouse, typically under the skin or in another organ, where they grow and form a tumor. Once the tumor has established, the mouse is treated with the experimental therapy, and the tumor's growth is monitored over time. The response of the tumor to the treatment is then assessed by measuring changes in tumor size or weight, as well as other parameters such as survival rate and metastasis.

Xenograft model antitumor assays are useful for evaluating the efficacy and safety of new cancer therapies before they are tested in human clinical trials. They provide valuable information on how the tumors respond to treatment, drug pharmacokinetics, and toxicity, which can help researchers optimize dosing regimens and identify potential side effects. However, it is important to note that xenograft models have limitations, such as differences in tumor biology between mice and humans, and may not always predict how well a therapy will work in human patients.

Molecular structure, in the context of biochemistry and molecular biology, refers to the arrangement and organization of atoms and chemical bonds within a molecule. It describes the three-dimensional layout of the constituent elements, including their spatial relationships, bond lengths, and angles. Understanding molecular structure is crucial for elucidating the functions and reactivities of biological macromolecules such as proteins, nucleic acids, lipids, and carbohydrates. Various experimental techniques, like X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, and cryo-electron microscopy (cryo-EM), are employed to determine molecular structures at atomic resolution, providing valuable insights into their biological roles and potential therapeutic targets.

Ellipticines are a class of naturally occurring alkaloids that have been isolated from various plants, including those in the family Apocynaceae. These compounds have been found to exhibit various biological activities, including anti-cancer and anti-microbial properties.

Ellipticines have a unique chemical structure, characterized by a planar, aromatic core with two side chains that contain nitrogen atoms. This structure allows ellipticines to intercalate into DNA, disrupting its normal function and leading to cell death. As a result, ellipticines have been studied as potential anti-cancer agents, particularly for the treatment of drug-resistant cancers.

In addition to their anti-cancer properties, ellipticines have also been found to exhibit antibacterial, antifungal, and antiparasitic activities. However, further research is needed to fully understand the mechanisms behind these effects and to determine the safety and efficacy of ellipticines as therapeutic agents.

Heterologous transplantation is a type of transplantation where an organ or tissue is transferred from one species to another. This is in contrast to allogeneic transplantation, where the donor and recipient are of the same species, or autologous transplantation, where the donor and recipient are the same individual.

In heterologous transplantation, the immune systems of the donor and recipient are significantly different, which can lead to a strong immune response against the transplanted organ or tissue. This is known as a graft-versus-host disease (GVHD), where the immune cells in the transplanted tissue attack the recipient's body.

Heterologous transplantation is not commonly performed in clinical medicine due to the high risk of rejection and GVHD. However, it may be used in research settings to study the biology of transplantation and to develop new therapies for transplant rejection.

Molecular models are three-dimensional representations of molecular structures that are used in the field of molecular biology and chemistry to visualize and understand the spatial arrangement of atoms and bonds within a molecule. These models can be physical or computer-generated and allow researchers to study the shape, size, and behavior of molecules, which is crucial for understanding their function and interactions with other molecules.

Physical molecular models are often made up of balls (representing atoms) connected by rods or sticks (representing bonds). These models can be constructed manually using materials such as plastic or wooden balls and rods, or they can be created using 3D printing technology.

Computer-generated molecular models, on the other hand, are created using specialized software that allows researchers to visualize and manipulate molecular structures in three dimensions. These models can be used to simulate molecular interactions, predict molecular behavior, and design new drugs or chemicals with specific properties. Overall, molecular models play a critical role in advancing our understanding of molecular structures and their functions.

Antineoplastic combined chemotherapy protocols refer to a treatment plan for cancer that involves the use of more than one antineoplastic (chemotherapy) drug given in a specific sequence and schedule. The combination of drugs is used because they may work better together to destroy cancer cells compared to using a single agent alone. This approach can also help to reduce the likelihood of cancer cells becoming resistant to the treatment.

The choice of drugs, dose, duration, and frequency are determined by various factors such as the type and stage of cancer, patient's overall health, and potential side effects. Combination chemotherapy protocols can be used in various settings, including as a primary treatment, adjuvant therapy (given after surgery or radiation to kill any remaining cancer cells), neoadjuvant therapy (given before surgery or radiation to shrink the tumor), or palliative care (to alleviate symptoms and prolong survival).

It is important to note that while combined chemotherapy protocols can be effective in treating certain types of cancer, they can also cause significant side effects, including nausea, vomiting, hair loss, fatigue, and an increased risk of infection. Therefore, patients undergoing such treatment should be closely monitored and managed by a healthcare team experienced in administering chemotherapy.

Neutropenia is a condition characterized by an abnormally low concentration (less than 1500 cells/mm3) of neutrophils, a type of white blood cell that plays a crucial role in fighting off bacterial and fungal infections. Neutrophils are essential components of the innate immune system, and their main function is to engulf and destroy microorganisms that can cause harm to the body.

Neutropenia can be classified as mild, moderate, or severe based on the severity of the neutrophil count reduction:

* Mild neutropenia: Neutrophil count between 1000-1500 cells/mm3
* Moderate neutropenia: Neutrophil count between 500-1000 cells/mm3
* Severe neutropenia: Neutrophil count below 500 cells/mm3

Severe neutropenia significantly increases the risk of developing infections, as the body's ability to fight off microorganisms is severely compromised. Common causes of neutropenia include viral infections, certain medications (such as chemotherapy or antibiotics), autoimmune disorders, and congenital conditions affecting bone marrow function. Treatment for neutropenia typically involves addressing the underlying cause, administering granulocyte-colony stimulating factors to boost neutrophil production, and providing appropriate antimicrobial therapy to prevent or treat infections.

Cell division is the process by which a single eukaryotic cell (a cell with a true nucleus) divides into two identical daughter cells. This complex process involves several stages, including replication of DNA, separation of chromosomes, and division of the cytoplasm. There are two main types of cell division: mitosis and meiosis.

Mitosis is the type of cell division that results in two genetically identical daughter cells. It is a fundamental process for growth, development, and tissue repair in multicellular organisms. The stages of mitosis include prophase, prometaphase, metaphase, anaphase, and telophase, followed by cytokinesis, which divides the cytoplasm.

Meiosis, on the other hand, is a type of cell division that occurs in the gonads (ovaries and testes) during the production of gametes (sex cells). Meiosis results in four genetically unique daughter cells, each with half the number of chromosomes as the parent cell. This process is essential for sexual reproduction and genetic diversity. The stages of meiosis include meiosis I and meiosis II, which are further divided into prophase, prometaphase, metaphase, anaphase, and telophase.

In summary, cell division is the process by which a single cell divides into two daughter cells, either through mitosis or meiosis. This process is critical for growth, development, tissue repair, and sexual reproduction in multicellular organisms.

Histones are highly alkaline proteins found in the chromatin of eukaryotic cells. They are rich in basic amino acid residues, such as arginine and lysine, which give them their positive charge. Histones play a crucial role in packaging DNA into a more compact structure within the nucleus by forming a complex with it called a nucleosome. Each nucleosome contains about 146 base pairs of DNA wrapped around an octamer of eight histone proteins (two each of H2A, H2B, H3, and H4). The N-terminal tails of these histones are subject to various post-translational modifications, such as methylation, acetylation, and phosphorylation, which can influence chromatin structure and gene expression. Histone variants also exist, which can contribute to the regulation of specific genes and other nuclear processes.

A cell line is a culture of cells that are grown in a laboratory for use in research. These cells are usually taken from a single cell or group of cells, and they are able to divide and grow continuously in the lab. Cell lines can come from many different sources, including animals, plants, and humans. They are often used in scientific research to study cellular processes, disease mechanisms, and to test new drugs or treatments. Some common types of human cell lines include HeLa cells (which come from a cancer patient named Henrietta Lacks), HEK293 cells (which come from embryonic kidney cells), and HUVEC cells (which come from umbilical vein endothelial cells). It is important to note that cell lines are not the same as primary cells, which are cells that are taken directly from a living organism and have not been grown in the lab.

Neoplasm antigens, also known as tumor antigens, are substances that are produced by cancer cells (neoplasms) and can stimulate an immune response. These antigens can be proteins, carbohydrates, or other molecules that are either unique to the cancer cells or are overexpressed or mutated versions of normal cellular proteins.

Neoplasm antigens can be classified into two main categories: tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs). TSAs are unique to cancer cells and are not expressed by normal cells, while TAAs are present at low levels in normal cells but are overexpressed or altered in cancer cells.

TSAs can be further divided into viral antigens and mutated antigens. Viral antigens are produced when cancer is caused by a virus, such as human papillomavirus (HPV) in cervical cancer. Mutated antigens are the result of genetic mutations that occur during cancer development and are unique to each patient's tumor.

Neoplasm antigens play an important role in the immune response against cancer. They can be recognized by the immune system, leading to the activation of immune cells such as T cells and natural killer (NK) cells, which can then attack and destroy cancer cells. However, cancer cells often develop mechanisms to evade the immune response, allowing them to continue growing and spreading.

Understanding neoplasm antigens is important for the development of cancer immunotherapies, which aim to enhance the body's natural immune response against cancer. These therapies include checkpoint inhibitors, which block proteins that inhibit T cell activation, and therapeutic vaccines, which stimulate an immune response against specific tumor antigens.

DNA-binding proteins are a type of protein that have the ability to bind to DNA (deoxyribonucleic acid), the genetic material of organisms. These proteins play crucial roles in various biological processes, such as regulation of gene expression, DNA replication, repair and recombination.

The binding of DNA-binding proteins to specific DNA sequences is mediated by non-covalent interactions, including electrostatic, hydrogen bonding, and van der Waals forces. The specificity of binding is determined by the recognition of particular nucleotide sequences or structural features of the DNA molecule.

DNA-binding proteins can be classified into several categories based on their structure and function, such as transcription factors, histones, and restriction enzymes. Transcription factors are a major class of DNA-binding proteins that regulate gene expression by binding to specific DNA sequences in the promoter region of genes and recruiting other proteins to modulate transcription. Histones are DNA-binding proteins that package DNA into nucleosomes, the basic unit of chromatin structure. Restriction enzymes are DNA-binding proteins that recognize and cleave specific DNA sequences, and are widely used in molecular biology research and biotechnology applications.

Nucleic acid conformation refers to the three-dimensional structure that nucleic acids (DNA and RNA) adopt as a result of the bonding patterns between the atoms within the molecule. The primary structure of nucleic acids is determined by the sequence of nucleotides, while the conformation is influenced by factors such as the sugar-phosphate backbone, base stacking, and hydrogen bonding.

Two common conformations of DNA are the B-form and the A-form. The B-form is a right-handed helix with a diameter of about 20 Å and a pitch of 34 Å, while the A-form has a smaller diameter (about 18 Å) and a shorter pitch (about 25 Å). RNA typically adopts an A-form conformation.

The conformation of nucleic acids can have significant implications for their function, as it can affect their ability to interact with other molecules such as proteins or drugs. Understanding the conformational properties of nucleic acids is therefore an important area of research in molecular biology and medicine.

Tumor suppressor protein p53, also known as p53 or tumor protein p53, is a nuclear phosphoprotein that plays a crucial role in preventing cancer development and maintaining genomic stability. It does so by regulating the cell cycle and acting as a transcription factor for various genes involved in apoptosis (programmed cell death), DNA repair, and cell senescence (permanent cell growth arrest).

In response to cellular stress, such as DNA damage or oncogene activation, p53 becomes activated and accumulates in the nucleus. Activated p53 can then bind to specific DNA sequences and promote the transcription of target genes that help prevent the proliferation of potentially cancerous cells. These targets include genes involved in cell cycle arrest (e.g., CDKN1A/p21), apoptosis (e.g., BAX, PUMA), and DNA repair (e.g., GADD45).

Mutations in the TP53 gene, which encodes p53, are among the most common genetic alterations found in human cancers. These mutations often lead to a loss or reduction of p53's tumor suppressive functions, allowing cancer cells to proliferate uncontrollably and evade apoptosis. As a result, p53 has been referred to as "the guardian of the genome" due to its essential role in preventing tumorigenesis.

Catenated DNA refers to the linking or interlocking of two or more DNA molecules in a circular form, where the circles are topologically entangled. This occurs during DNA replication when the sister chromatids (identical copies of DNA) are formed and remain interlinked before they are separated during cell division. The term "catenane" is used to describe this interlocking structure. It is important to note that in linear DNA, the term "catenated" does not apply since there is no circular formation.

I'm sorry for any confusion, but "Razoxane" is not a medical term that has a widely accepted or specific definition in the field of medicine. It is possible that you may be referring to "razoxane," which is a medication used in the treatment of certain types of cancer. Razoxane is an antineoplastic agent, which means it is a drug that is used to treat cancer. It works by interfering with the formation of blood vessels that supply tumors, which can help to slow or stop the growth of the tumor.

It is important to note that the use of razoxane is not widely accepted and it is not a commonly used cancer treatment. It is typically used only in certain specific circumstances and when other treatments have not been effective. As with any medication, razoxane should be used under the close supervision of a healthcare professional, and it is important to be aware of the potential risks and benefits.

Lung neoplasms refer to abnormal growths or tumors in the lung tissue. These tumors can be benign (non-cancerous) or malignant (cancerous). Malignant lung neoplasms are further classified into two main types: small cell lung carcinoma and non-small cell lung carcinoma. Lung neoplasms can cause symptoms such as cough, chest pain, shortness of breath, and weight loss. They are often caused by smoking or exposure to secondhand smoke, but can also occur due to genetic factors, radiation exposure, and other environmental carcinogens. Early detection and treatment of lung neoplasms is crucial for improving outcomes and survival rates.

A plasmid is a small, circular, double-stranded DNA molecule that is separate from the chromosomal DNA of a bacterium or other organism. Plasmids are typically not essential for the survival of the organism, but they can confer beneficial traits such as antibiotic resistance or the ability to degrade certain types of pollutants.

Plasmids are capable of replicating independently of the chromosomal DNA and can be transferred between bacteria through a process called conjugation. They often contain genes that provide resistance to antibiotics, heavy metals, and other environmental stressors. Plasmids have also been engineered for use in molecular biology as cloning vectors, allowing scientists to replicate and manipulate specific DNA sequences.

Plasmids are important tools in genetic engineering and biotechnology because they can be easily manipulated and transferred between organisms. They have been used to produce vaccines, diagnostic tests, and genetically modified organisms (GMOs) for various applications, including agriculture, medicine, and industry.

Electron Transport Complex I, also known as NADH:ubiquinone oxidoreductase, is a large protein complex located in the inner mitochondrial membrane of eukaryotic cells and the cytoplasmic membrane of prokaryotic cells. It is the first complex in the electron transport chain, a series of protein complexes that transfer electrons from NADH to oxygen, driving the synthesis of ATP through chemiosmosis.

Complex I consists of multiple subunits, including a flavin mononucleotide (FMN) cofactor and several iron-sulfur clusters, which facilitate the oxidation of NADH and the reduction of ubiquinone (coenzyme Q). The energy released during this electron transfer process is used to pump protons across the membrane, creating a proton gradient that drives ATP synthesis.

Defects in Complex I can lead to various mitochondrial diseases, including neurological disorders and muscle weakness.

A mutation is a permanent change in the DNA sequence of an organism's genome. Mutations can occur spontaneously or be caused by environmental factors such as exposure to radiation, chemicals, or viruses. They may have various effects on the organism, ranging from benign to harmful, depending on where they occur and whether they alter the function of essential proteins. In some cases, mutations can increase an individual's susceptibility to certain diseases or disorders, while in others, they may confer a survival advantage. Mutations are the driving force behind evolution, as they introduce new genetic variability into populations, which can then be acted upon by natural selection.

Podophyllotoxin is a pharmaceutical agent derived from the podophyllum plant. It is an antimitotic compound that inhibits microtubule assembly, leading to cell cycle arrest and apoptosis. It is primarily used in topical form as a treatment for genital warts, caused by certain types of human papillomavirus (HPV). Podophyllotoxin works by interfering with the growth of the wart cells, eventually causing them to die off.

It's important to note that podophyllotoxin is a potent cytotoxic agent and should only be used under the supervision of a healthcare professional. It should not be taken orally or applied to open wounds, and it should be kept out of reach of children.

'Escherichia coli' (E. coli) is a type of gram-negative, facultatively anaerobic, rod-shaped bacterium that commonly inhabits the intestinal tract of humans and warm-blooded animals. It is a member of the family Enterobacteriaceae and one of the most well-studied prokaryotic model organisms in molecular biology.

While most E. coli strains are harmless and even beneficial to their hosts, some serotypes can cause various forms of gastrointestinal and extraintestinal illnesses in humans and animals. These pathogenic strains possess virulence factors that enable them to colonize and damage host tissues, leading to diseases such as diarrhea, urinary tract infections, pneumonia, and sepsis.

E. coli is a versatile organism with remarkable genetic diversity, which allows it to adapt to various environmental niches. It can be found in water, soil, food, and various man-made environments, making it an essential indicator of fecal contamination and a common cause of foodborne illnesses. The study of E. coli has contributed significantly to our understanding of fundamental biological processes, including DNA replication, gene regulation, and protein synthesis.

In the context of medicine and pharmacology, "kinetics" refers to the study of how a drug moves throughout the body, including its absorption, distribution, metabolism, and excretion (often abbreviated as ADME). This field is called "pharmacokinetics."

1. Absorption: This is the process of a drug moving from its site of administration into the bloodstream. Factors such as the route of administration (e.g., oral, intravenous, etc.), formulation, and individual physiological differences can affect absorption.

2. Distribution: Once a drug is in the bloodstream, it gets distributed throughout the body to various tissues and organs. This process is influenced by factors like blood flow, protein binding, and lipid solubility of the drug.

3. Metabolism: Drugs are often chemically modified in the body, typically in the liver, through processes known as metabolism. These changes can lead to the formation of active or inactive metabolites, which may then be further distributed, excreted, or undergo additional metabolic transformations.

4. Excretion: This is the process by which drugs and their metabolites are eliminated from the body, primarily through the kidneys (urine) and the liver (bile).

Understanding the kinetics of a drug is crucial for determining its optimal dosing regimen, potential interactions with other medications or foods, and any necessary adjustments for special populations like pediatric or geriatric patients, or those with impaired renal or hepatic function.

A neoplasm is a tumor or growth that is formed by an abnormal and excessive proliferation of cells, which can be benign or malignant. Neoplasm proteins are therefore any proteins that are expressed or produced in these neoplastic cells. These proteins can play various roles in the development, progression, and maintenance of neoplasms.

Some neoplasm proteins may contribute to the uncontrolled cell growth and division seen in cancer, such as oncogenic proteins that promote cell cycle progression or inhibit apoptosis (programmed cell death). Others may help the neoplastic cells evade the immune system, allowing them to proliferate undetected. Still others may be involved in angiogenesis, the formation of new blood vessels that supply the tumor with nutrients and oxygen.

Neoplasm proteins can also serve as biomarkers for cancer diagnosis, prognosis, or treatment response. For example, the presence or level of certain neoplasm proteins in biological samples such as blood or tissue may indicate the presence of a specific type of cancer, help predict the likelihood of cancer recurrence, or suggest whether a particular therapy will be effective.

Overall, understanding the roles and behaviors of neoplasm proteins can provide valuable insights into the biology of cancer and inform the development of new diagnostic and therapeutic strategies.

DNA gyrase is a type II topoisomerase enzyme that plays a crucial role in the negative supercoiling and relaxation of DNA in bacteria. It functions by introducing transient double-stranded breaks into the DNA helix, allowing the strands to pass through one another and thereby reducing positive supercoils or introducing negative supercoils as required for proper DNA function, replication, and transcription.

DNA gyrase is composed of two subunits, GyrA and GyrB, which form a heterotetrameric structure (AB-BA) in the functional enzyme. The enzyme's activity is targeted by several antibiotics, such as fluoroquinolones and novobiocin, making it an essential target for antibacterial drug development.

In summary, DNA gyrase is a bacterial topoisomerase responsible for maintaining the correct supercoiling of DNA during replication and transcription, which can be inhibited by specific antibiotics to combat bacterial infections.

Catalysis is the process of increasing the rate of a chemical reaction by adding a substance known as a catalyst, which remains unchanged at the end of the reaction. A catalyst lowers the activation energy required for the reaction to occur, thereby allowing the reaction to proceed more quickly and efficiently. This can be particularly important in biological systems, where enzymes act as catalysts to speed up metabolic reactions that are essential for life.

In the context of medical and biological sciences, a "binding site" refers to a specific location on a protein, molecule, or cell where another molecule can attach or bind. This binding interaction can lead to various functional changes in the original protein or molecule. The other molecule that binds to the binding site is often referred to as a ligand, which can be a small molecule, ion, or even another protein.

The binding between a ligand and its target binding site can be specific and selective, meaning that only certain ligands can bind to particular binding sites with high affinity. This specificity plays a crucial role in various biological processes, such as signal transduction, enzyme catalysis, or drug action.

In the case of drug development, understanding the location and properties of binding sites on target proteins is essential for designing drugs that can selectively bind to these sites and modulate protein function. This knowledge can help create more effective and safer therapeutic options for various diseases.

Drug resistance, also known as antimicrobial resistance, is the ability of a microorganism (such as bacteria, viruses, fungi, or parasites) to withstand the effects of a drug that was originally designed to inhibit or kill it. This occurs when the microorganism undergoes genetic changes that allow it to survive in the presence of the drug. As a result, the drug becomes less effective or even completely ineffective at treating infections caused by these resistant organisms.

Drug resistance can develop through various mechanisms, including mutations in the genes responsible for producing the target protein of the drug, alteration of the drug's target site, modification or destruction of the drug by enzymes produced by the microorganism, and active efflux of the drug from the cell.

The emergence and spread of drug-resistant microorganisms pose significant challenges in medical treatment, as they can lead to increased morbidity, mortality, and healthcare costs. The overuse and misuse of antimicrobial agents, as well as poor infection control practices, contribute to the development and dissemination of drug-resistant strains. To address this issue, it is crucial to promote prudent use of antimicrobials, enhance surveillance and monitoring of resistance patterns, invest in research and development of new antimicrobial agents, and strengthen infection prevention and control measures.

Ovarian neoplasms refer to abnormal growths or tumors in the ovary, which can be benign (non-cancerous) or malignant (cancerous). These growths can originate from various cell types within the ovary, including epithelial cells, germ cells, and stromal cells. Ovarian neoplasms are often classified based on their cell type of origin, histological features, and potential for invasive or metastatic behavior.

Epithelial ovarian neoplasms are the most common type and can be further categorized into several subtypes, such as serous, mucinous, endometrioid, clear cell, and Brenner tumors. Some of these epithelial tumors have a higher risk of becoming malignant and spreading to other parts of the body.

Germ cell ovarian neoplasms arise from the cells that give rise to eggs (oocytes) and can include teratomas, dysgerminomas, yolk sac tumors, and embryonal carcinomas. Stromal ovarian neoplasms develop from the connective tissue cells supporting the ovary and can include granulosa cell tumors, thecomas, and fibromas.

It is essential to diagnose and treat ovarian neoplasms promptly, as some malignant forms can be aggressive and potentially life-threatening if not managed appropriately. Regular gynecological exams, imaging studies, and tumor marker tests are often used for early detection and monitoring of ovarian neoplasms. Treatment options may include surgery, chemotherapy, or radiation therapy, depending on the type, stage, and patient's overall health condition.

Substrate specificity in the context of medical biochemistry and enzymology refers to the ability of an enzyme to selectively bind and catalyze a chemical reaction with a particular substrate (or a group of similar substrates) while discriminating against other molecules that are not substrates. This specificity arises from the three-dimensional structure of the enzyme, which has evolved to match the shape, charge distribution, and functional groups of its physiological substrate(s).

Substrate specificity is a fundamental property of enzymes that enables them to carry out highly selective chemical transformations in the complex cellular environment. The active site of an enzyme, where the catalysis takes place, has a unique conformation that complements the shape and charge distribution of its substrate(s). This ensures efficient recognition, binding, and conversion of the substrate into the desired product while minimizing unwanted side reactions with other molecules.

Substrate specificity can be categorized as:

1. Absolute specificity: An enzyme that can only act on a single substrate or a very narrow group of structurally related substrates, showing no activity towards any other molecule.
2. Group specificity: An enzyme that prefers to act on a particular functional group or class of compounds but can still accommodate minor structural variations within the substrate.
3. Broad or promiscuous specificity: An enzyme that can act on a wide range of structurally diverse substrates, albeit with varying catalytic efficiencies.

Understanding substrate specificity is crucial for elucidating enzymatic mechanisms, designing drugs that target specific enzymes or pathways, and developing biotechnological applications that rely on the controlled manipulation of enzyme activities.

Topoisomerase inhibitors influence these essential cellular processes. Some topoisomerase inhibitors prevent topoisomerases ... Topoisomerase inhibitors are chemical compounds that block the action of topoisomerases, which are broken into two broad ... Catalytic inhibitors are the other main identification of TopII inhibitors. Common catalytic inhibitors are Bisdioxopiperazine ... topoisomerase inhibitors hold a prominent place among antibiotics and anticancer drugs in active medical use, as inhibitors ...
Camptothecin analogs as potent inhibitors of topoisomerase I. Wall, M., & Wani, M. (1992). Camptothecin analogs as potent ... The camptothecin analogs are effective inhibitors of topoisomerase I and show anti-leukemic and anti-tumor activity. ... inhibitors of topoisomerase I. (U.S. Patent No. 5106742). http://patft.uspto.gov/netacgi/nph-Parser?patentnumber=5106742 ...
Zymeworks Topoisomerase-1 inhibitor technology uses a novel fit-for-purpose camptothecin payload with moderate potency and ... and lysosomal trafficking releases the topoisomerase inhibitor payload ZD06519. ...
Antineoplastics, Topoisomerase Inhibitors. Class Summary. Topoisomerase-inhibiting antineoplastic agents prevent cell growth ... Antineoplastics, VEGF Inhibitor. Class Summary. Vascular endothelial growth factor (VEGF) is crucial to angiogenesis. VEGF ... PD-1/PD-L1 Inhibitors. Class Summary. Monoclonal antibodies that bind the programmed cell death-1 protein (PD-1) ligands, PD-L1 ... Topotecan inhibits topoisomerase I, inhibiting DNA replication. It acts in the S phase of the cell cycle. Patients who have ...
Antineoplastics, Topoisomerase Inhibitors. Class Summary. Topoisomerase inhibitors inhibit cell growth and proliferation. ... Antineoplastics, VEGF Inhibitor. Class Summary. Vascular endothelial growth factor inhibitors reduce tumor vascularity and ... which is composed of a cleavable linker and topoisomerase inhibitor (DXd). The drug binds to HER2 on tumors and undergoes ... PD-1/PD-L1 Inhibitors. Class Summary. PD-1 and related target PD-ligand 1 (PD-L1) are expressed on the surface of activated T ...
Synthesis and Evaluation of Indenoisoquinoline Non-Camptothecin Topoisomerase I Inhibitors (NIH-Only) ... Synthesis and Evaluation of Indenoisoquinoline Non-Camptothecin Topoisomerase I Inhibitors (NIH-Only) ...
EP-0431076-B1 chemical patent summary.
... a topoisomerase I inhibitor, and etoposide, a topoisomerase II inhibitor. Sensitivity to these drugs cannot be predicted by ... Furthermore, the sensitivity to topoisomerase inhibitors correlates neither with endogenous levels of either topoisomerase I or ... Cytotoxicity of Topoisomerase Inhibitors to Human Colorectal Cancer Cell Lines.. The cytotoxicity of CPT and ETP to colorectal ... Cytotoxicity of Topoisomerase Inhibitors to hMLH1-expressing HCT116 Transfectants.. The cytotoxicity of CPT and ETP toward ...
Reversal of Physiological Stress-Induced Resistance to Topoisomerase II Inhibitors Using an Inducible Phosphorylation Site- ... Reversal of Physiological Stress-Induced Resistance to Topoisomerase II Inhibitors Using an Inducible Phosphorylation Site- ... Reversal of Physiological Stress-Induced Resistance to Topoisomerase II Inhibitors Using an Inducible Phosphorylation Site- ... Reversal of Physiological Stress-Induced Resistance to Topoisomerase II Inhibitors Using an Inducible Phosphorylation Site- ...
Topoisomerase inhibitors. Topoisomerase inhibitors are chemical compounds that block the action of topoisomerases. ... Action of topoisomerase begins by breaking one of DNA strands. *then they induce a topological change in DNA such as relaxing ... Intercalating drugs block the reconnection step, freezing the topoisomerase after it has broken the DNA. ...
Topoisomerase inhibitors can be divided into topoisomerase I and topoisomerase II depending on which enzyme they affect. ... Topoisomerase inhibitors. Topoisomerase inhibitors block cancer cells from dividing and spreading by interfering with enzymes ... topoisomerase inhibitors. leukemia. pancreatic cancer. ovarian cancer. gastrointestinal cancer. lung cancer. plant alkaloids: ... Drugs that fall in the subgroup called topoisomerase II inhibitors can increase your risk of developing a second cancer. ...
Topoisomerase Inhibitor. Topoisomerase inhibitors have gained interest as therapeutics against infectious and cancerous cells. ... Posted in Topoisomerase Signaling , Leave a reply 6 to 0 5, unit B from 6 9 to 1 6, and unit C from 7 8 to 0 6 Sec. Posted on ... Posted in Topoisomerase Signaling , Leave a reply The use of NH3 (6 M) allowed the recovery of Cd(II), with a PF of. Posted on ... Posted in Topoisomerase Signaling , Leave a reply We used robust-design models and Cormack-Jolly-Seber models to es. Posted on ...
... Read more For this function, we used a lysosomal inhibitor, leupeptin, to arrest the autophagic pathway ... Topoisomerase Read more This is accompanied by normal-level T B and cell cell responses This is accompanied by normal-level T ... Topoisomerase Read more Data on the transient inactivation of c-MYC indicate selective effects on cancer cells,18 but exactly ... Topoisomerase Read more These findings provide fresh evidence for the mechanism where this FDA-approved course of substances ...
Novel fluoroindenoisoquinoline non-camptothecin topoisomerase I inhibitors. / Marzi, Laetitia; Agama, Keli; Murai, Junko et al. ... Novel fluoroindenoisoquinoline non-camptothecin topoisomerase I inhibitors. Laetitia Marzi, Keli Agama, Junko Murai, Simone ... Novel fluoroindenoisoquinoline non-camptothecin topoisomerase I inhibitors. In: Molecular Cancer Therapeutics. 2018 ; Vol. 17, ... Novel fluoroindenoisoquinoline non-camptothecin topoisomerase I inhibitors. Molecular Cancer Therapeutics. 2018 Aug;17(8):1694- ...
Topoisomerase I/II inhibitor 3 (compound 7) is a potent topoisomerase I (Topo I) and II (Topo II) dual inhibitor. Topoisomerase ... We do not sell to patients.Topoisomerase I/II inhibitor 3Biological Activity: ... I/II inhibitor 3 can inhibit cell proliferation, invasion and migration, and induce apoptosis by inhibiting PI3K/Akt/mTOR ... Agonist Inhibitor,Antagonist Inhibitor,Inducer Inhibitor,MDM2 Inhibitor Inhibitor,Modulator MDM2 Inhibitor,Inhibitor Modulator ...
Topoisomerase Inhibitor. Topoisomerase inhibitors have gained interest as therapeutics against infectious and cancerous cells. ... This entry was posted in Topoisomerase Signaling by topo5813. Bookmark the permalink. Leave a Reply Cancel reply. Your email ... Cell Cycle inhibitor Sleep disturbance was classified into four sleep complaints: difficulty initiating AZD8055 supplier sleep ...
TOPOISOMERASE INHIBITORS. Used to treat:. *Leukemia. *Lung, ovarian, gastrointestinal, and other cancers ...
Antineoplastics, Topoisomerase Inhibitors. Class Summary. These agents prevent cell growth and proliferation. They work by ... Irinotecan binds reversibly to the topoisomerase I-DNA complex and prevents the ligation of the cleaved DNA strand. It can be ... They intercalate between DNA base pairs and trigger DNA cleavage by topoisomerase II. ...
Control contaminated cells without inhibitors, but with added DMSO amounts equal to those found in the wells with inhibitors, ... Background Proton pump inhibitors (PPIs) are influenced by cytochrome P450 2C19. * Post author By conferencedequebec ... Background Proton pump inhibitors (PPIs) are influenced by cytochrome P450 2C19 (CYP2C19) polymorphisms. higher with omeprazole ...
Repair of topoisomerase 1 (TOP1) DNA protein crosslinks (DPC) limits the efficacy of the TOP1 inhibitor irinotecan in cancer ... Targeting neddylation sensitizes colorectal cancer to topoisomerase I inhibitors by inactivating the DCAF13-CRL4 ubiquitin ... Here, the authors identify pevonedistat, NEDD8 inhibitor, as synergistic with irinotecan by blocking neddylation-activated ...
Control contaminated cells without inhibitors, but with added DMSO amounts equal to those found in the wells with inhibitors, ... Proton pump inhibitors (PPIs) will be the strongest gastric acidity suppressing. * Post author By conferencedequebec ... Proton pump inhibitors (PPIs) will be the strongest gastric acidity suppressing drugs obtainable, and their make use of is ...
Chalcones, inhibitors for topoisomerase i and cathepsin B and L, as potential anti-cancer agents. Authors. Kim S.-H.; Lee E.; ... Chalcones, inhibitors for topoisomerase i and cathepsin B and L, as potential anti-cancer agents ... In the topoisomerase inhibition test, most analogues in group III and IV except compound 11 exhibited more efficient ... Overall, although more compounds should be tested and analyzed for clear SAR against topoisomerase I and cathepsin B and L, ...
Some type I topoisomerase inhibitors include camptothecins: irinotecan and topotecan.. *Examples of type II inhibitors include ... Topoisomerase inhibitors (L01CB and L01XX). Topoisomerases are essential enzymes that maintain the topology of DNA. Inhibition ... topoisomerase inhibitors, and other antitumour agents. All of these drugs affect cell division or DNA synthesis and function in ... Spindle poison/mitotic inhibitor. Taxane: (Docetaxel, Larotaxel, Paclitaxel). Vinca: (Vinblastine, Vincristine, Vindesine, ...
Topoisomerase I Inhibitors / administration & dosage* * Topoisomerase I Inhibitors / adverse effects * Topotecan / ...
The Evolving Evidence-Based And Guideline-Supported Role For Nanoliposomal Topoisomerase Inhibitors In Metastatic Pancreatic ... The Evolving Evidence-Based And Guideline-Supported Role For Nanoliposomal Topoisomerase Inhibitors In Metastatic Pancreatic ...
Topoisomerase-I Inhibitors in Gynaecologic Tumours. C F Verschraegen, A P Kudelka, J J Kavanagh ... Angiotensin converting enzyme inhibitors (ACEI) and angiotensin II receptor antagonists (ATRA) are both well established drugs ... Topoisomerases are essential nuclear enzymes with a multiplicity of cellular functions involving DNA replication, RNA ... Angiotensin-Converting Enzyme Inhibitor versus Angiotensin 2 Receptor Antagonist Therapy and the Influence of Angiotensin- ...
Effect of safranal on the response of cancer cells to topoisomerase I inhibitors : Does sequence matter?. In: Frontiers in ... Effect of safranal on the response of cancer cells to topoisomerase I inhibitors: Does sequence matter? / Lozon, Lama; Saleh, ... Dive into the research topics of Effect of safranal on the response of cancer cells to topoisomerase I inhibitors: Does ... Effect of safranal on the response of cancer cells to topoisomerase I inhibitors: Does sequence matter?. ...
A new series of triazolophthalazine derivatives was designed and synthesized as topoisomerase II (Topo II) inhibitors and DNA ... Discovery of novel triazolophthalazine derivatives as DNA intercalators and topoisomerase II inhibitors. Sakr, Helmy,Ayyad, ... as inhibitors of NF-κB activation. We showed that compounds 6h (6-(2,4-dichlorophenoxy)-[1,2,4]triazolo[3,4-a]phthalazine-3- ...
... bacterial topoisomerase inhibitors and regulatory proteins may inspire the discovery of novel human topoisomerase inhibitors. ... The Effect of Topoisomerase I Inhibitors on the Efficacy of T-Cell-Based Cancer Immunotherapy.. J Natl Cancer Inst. 2018; 110(7 ... It may also be possible to design catalytic inhibitors of topoisomerases by targeting certain inactive conformations of these ... The initial sequence used for this oligonucleotide-linked topoisomerase inhibitor (OTI) was identified as part of the ...
TOPOISOMERASE INHIBITORS. Used to treat:. *Leukemia. *Lung, ovarian, gastrointestinal, and other cancers ...
  • Topotecan inhibits topoisomerase I, inhibiting DNA replication. (medscape.com)
  • Contrary to other anticancer targets, topoisomerase I (TOP1) is targeted by only one chemical class of FDA-approved drugs: topotecan and irinotecan, the derivatives of the plant alkaloid, camptothecin. (usuhs.edu)
  • Although topotecan (TPT), a topoisomerase1 inhibitor, is a first- and second-line drug for lung and colon cancers, the development of drug resistance and toxicity still remain as a major obstacle to chemotherapeutic success. (uaeu.ac.ae)
  • The research, published in the Proceedings of the National Academy of Sciences , shows how the common chemotherapy drug topotecan can drastically suppress the expression of Topoisomerase-1, a gene that triggers the creation of proteins essential for normal brain function. (sciencedaily.com)
  • This discovery led them to investigate how topotecan affects the specific topoisomerase enzymes in cancer cells and in neurons. (sciencedaily.com)
  • In the PNAS paper, the researchers describe how topotecan hits its intended target -- the topoisomerase proteins that are integral for cell division, a hallmark of cancer cells. (sciencedaily.com)
  • These experiments used only topotecan, but there's an entire class of topoisomerase inhibitors. (sciencedaily.com)
  • Studies searching for antibiotic and anticancer agents in the mid to late 20th century have illuminated the existence of numerous unique families of both TopI and TopII inhibitors, with the 1960s alone resulting in the discovery of the camptothecin, anthracycline and epipodophyllotoxin classes. (wikipedia.org)
  • Topoisomerase inhibitor classes have been derived from a wide variety of disparate sources, with some being natural products first extracted from plants (camptothecin, etoposide) or bacterial samples (doxorubicin, indolocarbazole), while others possess purely synthetic, and often accidental, origins (quinolone, indenoisoquinoline). (wikipedia.org)
  • Currently, topoisomerase inhibitors hold a prominent place among antibiotics and anticancer drugs in active medical use, as inhibitors like doxorubicin (anthracycline, TopII inhibitor), etoposide (TopII inhibitor), ciprofloxaxin (fluoroquinolone, TopII inhibitor), and irinotecan (camptothecin derivative, TopI inhibitor) were all included in the 2019 WHO Model List for Essential Medicines. (wikipedia.org)
  • Camptothecin-derived TopI inhibitors function by forming a ternary complex with TopI-DNA and are able to stack between the base pairs that flank the cleavage site due to their planar structure. (wikipedia.org)
  • Camptothecin analogs as potent inhibitors of topoisomerase I . (U.S. Patent No. 5106742 ). (rti.org)
  • The camptothecin analogs are effective inhibitors of topoisomerase I and show anti-leukemic and anti-tumor activity. (rti.org)
  • Zymeworks Topoisomerase-1 inhibitor technology uses a novel fit-for-purpose camptothecin payload with moderate potency and strong bystander activity in conjunction with a traceless, plasma-stable, cleavable peptide linker. (zymeworks.com)
  • We have shown that colorectal cancer cell lines defective in DNA MMR exhibit an increased sensitivity to both camptothecin, a topoisomerase I inhibitor, and etoposide, a topoisomerase II inhibitor. (aacrjournals.org)
  • Genz-644282 is a non-camptothecin inhibitor of topoisomerase I with potential antineoplastic activity. (adooq.com)
  • Drugs called mitotic inhibitors stop cancer cells from making more copies of themselves. (webmd.com)
  • Topoisomerase inhibitors have been used as important experimental tools that have contributed to the discovery of some topoisomerases, as the quinolone nalidixic acid helped elucidate the bacterial TopII proteins it binds to. (wikipedia.org)
  • Occupational exposure limits with proteins such as topoisomerase inhibitors, and mitotic and meiotic spindle poisons. (cdc.gov)
  • Repair of topoisomerase 1 (TOP1) DNA protein crosslinks (DPC) limits the efficacy of the TOP1 inhibitor irinotecan in cancer therapy. (nature.com)
  • Here, the authors identify pevonedistat, NEDD8 inhibitor, as synergistic with irinotecan by blocking neddylation-activated ubiquitin/proteasomal degradation of TOP1-DPC. (nature.com)
  • Krop noted that the drug is 10 times more potent than topoisomerase I inhibitors such as irinotecan, one of the more typically-used drugs in this class. (medpagetoday.com)
  • Knowledge of the first topoisomerase inhibitors, and their medical potential as anticancer drugs and antibiotics, predates the discovery of the first topoisomerase (Escherichia. (wikipedia.org)
  • Bortezomib is the first drug approved in the group of anticancer agents known as proteasome inhibitors. (medscape.com)
  • Dr. Yalowich's lab focuses on the mechanisms of action and resistance to DNA topoisomerase II (topo II)-targeted agents, such as the anticancer agent etoposide (VP-16). (osu.edu)
  • In this study, treatment of EMT6 mouse mammary tumor cells with hypoxia or the chemical stress agents brefeldin A (BFA) or okadaic acid (OA) causes the development of resistance to the topoisomerase II inhibitor etoposide. (aspetjournals.org)
  • CRISPR/Cas9 Genome Editing of the Human Topoisomerase II Intron 19 5' Splice Site Circumvents Etoposide Resistance in Human Leukemia K562 Cells. (osu.edu)
  • hsa-miR-9-3p and hsa-miR-9-5p as Post-Transcriptional Modulators of DNA Topoisomerase II in Human Leukemia K562 Cells with Acquired Resistance to Etoposide. (osu.edu)
  • Our results also indicate that neither p53 status, nor cell cycle alterations correlate with the sensitivity of colorectal cancer cells to topoisomerase inhibitors. (aacrjournals.org)
  • Biological Activity: Topoisomerase I/II inhibitor 3 (compound 7) is a potent topoisomerase I (Topo I) and II (Topo II) dual inhibitor. (epigenetics-modulation-frontier.com)
  • RHPS4 is a potent inhibitor of Telomerase at submicromolar. (adooq.com)
  • Catabolism of the ADC following target engagement, internalization, and lysosomal trafficking releases the topoisomerase inhibitor payload ZD06519. (zymeworks.com)
  • Trastuzumab deruxtecan is an antibody drug conjugate with three key components: an anti-HER2 monoclonal antibody with the same amino acid as trastuzumab, a topoisomerase I inhibitor payload, and a tetra-peptide-based cleavable linker. (medpagetoday.com)
  • The ADCs sacituzumab govitecan (Trodelvy) and trastuzumab deruxtecan (Enhertu), for instance, both incorporate a topoisomerase inhibitor as their toxic payload. (medscape.com)
  • Similarly, trastuzumab emtansine (Kadcyla), enfortumab vedotin (Padcev), mirvetuximab soravtansine (Elahere), and tisotumab vedotin (Tivdak), which have a microtubule inhibitor as the payload, come with similar toxicities to the microtubule-inhibiting drugs. (medscape.com)
  • These topoisomerase-DNA-inhibitor complexes are cytotoxic agents, as the un-repaired single- and double stranded DNA breaks they cause can lead to apoptosis and cell death. (wikipedia.org)
  • Because of this ability to induce apoptosis, topoisomerase inhibitors have gained interest as therapeutics against infectious and cancerous cells. (wikipedia.org)
  • Topoisomerase I/II inhibitor 3 can inhibit cell proliferation, invasion and migration, and induce apoptosis by inhibiting PI3K/Akt/mTOR signaling pathway. (epigenetics-modulation-frontier.com)
  • SN-38 binds to and stabilizes the topoisomerase I-DNA complex and prevents the relegation of DNA after it has been cleaved by topoisomerase I, inhibiting DNA replication. (medscape.com)
  • Topoisomerases are nuclear enzymes that modify the topological state of DNA and participate in fundamental metabolic processes such as replication, transcription, repair, recombination, and chromosome segregation (1) . (aacrjournals.org)
  • Topoisomerases are essential nuclear enzymes with a multiplicity of cellular functions involving DNA replication, RNA transcription, mitosis, and chromosome condensation. (annals.edu.sg)
  • Another type of medicine, called topoisomerase inhibitors, also attacks enzymes that help cancer cells divide and grow. (webmd.com)
  • Pretreatment with the proteasome inhibitor carbobenzyoxyl-leucinyl-leucinyl-leucinal inhibits stress-induced NF-κB activation and reverses BFA-induced drug resistance. (aspetjournals.org)
  • Reversible proteasome inhibitor. (medscape.com)
  • detailed the discovery of the bacterial TopII DNA gyrase and discussed its inhibition when introduced to coumarin and quinolone class inhibitors, sparking greater interest in topoisomerase-targeting antibiotic and antitumor agents. (wikipedia.org)
  • Oral Pharmacokinetics and Efficacy of BWC0977, a Novel Bacterial Topoisomerase Inhibitor. (asm.org)
  • Dioxane-Linked Amide Derivatives as Novel Bacterial Topoisomerase Inhibitors against Gram-Positive Staphylococcus aureus. (osu.edu)
  • Novel bacterial topoisomerase inhibitors derived from isomannide. (osu.edu)
  • Gepotidacin (GlaxoSmithKline) is another bacterial topoisomerase II inhibitor with good in vitro activity against a wide range of drug-resistant bacteria, including methicillin-resistant Staphylococcus aureus , extended-spectrum beta-lactamase-producing Enterobacteriaceae , and N gonorrhoeae . (medscape.com)
  • The indenoisoquinolines LMP400, LMP744, and LMP776 are novel noncamptothecin TOP1 inhibitors in clinical trial, which overcome the limitations of camptothecins. (usuhs.edu)
  • Some topoisomerase inhibitors prevent topoisomerases from performing DNA strand breaks while others, deemed topoisomerase poisons, associate with topoisomerase-DNA complexes and prevent the re-ligation step of the topoisomerase mechanism. (wikipedia.org)
  • TMPyP4 tosylate (TMP 1363) is a telomerase inhibitor with antitumor effects in osteosarcoma cell lines. (adooq.com)
  • Furthermore, they imply that coadministration of agents that inhibit NF-κB may enhance the efficacy of topoisomerase II inhibitors in clinical cancer chemotherapy. (aspetjournals.org)
  • VEGF inhibitors directly bind to the VEGF protein to disrupt angiogenesis. (medscape.com)
  • Topoisomerase inhibitors are chemical compounds that block the action of topoisomerases, which are broken into two broad subtypes: type I topoisomerases (TopI) and type II topoisomerases (TopII). (wikipedia.org)
  • Treatment with TopI inhibitors stabilizes the intermediate cleavable complex, preventing DNA re-ligation, and inducing lethal DNA strand breaks. (wikipedia.org)
  • In cancer cells, however, these checkpoints are typically inactivated, making them selectively sensitive to TopI inhibitors. (wikipedia.org)
  • Exploring Potential MCR-1 Inhibitors: Restoring In Vitro Polymyxin Activity in MCR-1-Producing Enterobacteriales. (asm.org)
  • Topoisomerase inhibitors inhibit cell growth and proliferation. (medscape.com)
  • Effects of DNA topoisomerase IIα splice variants on acquired drug resistance. (osu.edu)
  • Topoisomerase plays important roles in cellular reproduction and DNA organization, as they mediate the cleavage of single and double stranded DNA to relax supercoils, untangle catenanes, and condense chromosomes in eukaryotic cells. (wikipedia.org)
  • It was identified that the nuclear division cycle 80, cyclin B2 and topoisomerase 2‑α may serve important roles in adrenocortical tumor development. (cancerindex.org)
  • It is described as an angiogenesis inhibitor and a phytoestrogen . (wikipedia.org)
  • Topoisomerase inhibitors are chemical compounds that block the action of topoisomerases . (github.io)
  • Resistance to fluoroquinolones in S . pneu- with MIC 4-8 µg/mL) was caused by a reserpine-sensitive moniae can be acquired by point mutations, intraspecific efflux phenotype (n = 4) or single topoisomerase IV ( parC [n = 24] or parE [n = 1]) changes. (cdc.gov)
  • Topoisomerase inhibitors have gained interest as therapeutics against infectious and cancerous cells. (topoisomerasesignaling.com)
  • Zoliflodacin (Entasis Therapeutics), a first-in-class spiropyrimidinetrione topoisomerase II inhibitor with activity against several pathogens, including N gonorrhoeae and C trachomatis . (medscape.com)
  • Biological evaluation, docking studies, and in silico ADME prediction of some pyrimidine and pyridine derivatives as potential EGFR WT and EGFR T790M inhibitors. (bvsalud.org)
  • Sensitivity to these drugs cannot be predicted by measuring endogenous levels of topoisomerase I and II. (aacrjournals.org)
  • Intercalating drugs block the reconnection step, freezing the topoisomerase after it has broken the DNA. (github.io)
  • It acts throughout the entire cell cycle and by direct intercalating into DNA triggers DNA breakage by topoisomerase II, causing subsequent cytocydal activity. (medscape.com)
  • Costunolide is an Inhibitor of human telomerase activity (IC50 = 65 μM in MCF-7 breast cancer cells). (adooq.com)
  • Topoisomerase inhibitors influence these essential cellular processes. (wikipedia.org)
  • Topoisomerase-inhibiting antineoplastic agents prevent cell growth and proliferation. (medscape.com)
  • Because MMR is assumed to modulate cytotoxicity to various chemotherapeutic agents that act upon DNA, our objectives have been to define its possible involvement in the cytotoxicity of topoisomerase inhibitors. (aacrjournals.org)
  • The primary toxicities associated with these agents - alopecia, neutropenia, and diarrhea - align with the toxicities seen with unconjugated topoisomerase inhibitors. (medscape.com)
  • Cell Cycle inhibitor Sleep disturbance was classified into four sleep complaints: difficulty initiating AZD8055 supplier sleep, difficulty maintaining sleep, early-morning awakening, and non-restorative sleep. (topoisomerasesignaling.com)
  • Effect of safranal on the response of cancer cells to topoisomerase I inhibitors: Does sequence matter? (uaeu.ac.ae)
  • We think drug developers should be aware of this when testing their next generation of topoisomerase inhibitors. (sciencedaily.com)

No images available that match "topoisomerase i inhibitors"