Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
Spectrochim Acta A Mol Biomol Spectrosc ; 317: 124359, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-38704996

RESUMO

SERS (Surface Enhanced Raman Spectroscopy) is a new Raman spectroscopy which relies on Surface Plasmon Resonance (SPR) of metal nanoparticles. We have applied colloidal silver and gold nanoparticles as amplifier agents to enhance nucleotide Raman signals. It is observed that without these enhancing agents, it is impossible to investigate nucleotide spectrum due to weak Raman signals. Interaction mechanism of Melphalan, an anticancer drug with four nucleotides (Adenine, Cytosine, Guanine, Thymine) was investigated using SERS to detect and identify changes due to alkylating process in Raman spectra. After incubating Melphalan drug with nucleotides for 24 h at 37 °C, some changes occurred in SERS spectrum and interpretation of SERS spectra revealed the influence of the alkyl substitution on peaks and Raman shifts. After incubation of Melphalan with each nucleotide, intensity of relevant SERS signals assigned to Amid III group of Cytosine and Amid I of Thymine decreased significantly, confirming alkylating taking place. In this study, we also investigated the effect of nanoparticles type on nucleotide spectrum. We could not obtain useful information in the cases of guanine nucleotide. The SERS spectrum of Cytosine as an example of nucleotides in aqueous solution compared to solid state and results demonstrated that in solid state better signals were obtained than in liquid state.


Assuntos
Melfalan , Nanopartículas Metálicas , Nucleotídeos , Análise Espectral Raman , Análise Espectral Raman/métodos , Melfalan/química , Nucleotídeos/química , Nanopartículas Metálicas/química , Ouro/química , Alquilantes/química , Prata/química
2.
Biomolecules ; 14(4)2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38672511

RESUMO

TG2 is a unique member of the transglutaminase family as it undergoes a dramatic conformational change, allowing its mutually exclusive function as either a cross-linking enzyme or a G-protein. The enzyme's dysregulated activity has been implicated in a variety of pathologies (e.g., celiac disease, fibrosis, cancer), leading to the development of a wide range of inhibitors. Our group has primarily focused on the development of peptidomimetic targeted covalent inhibitors, the nature and size of which were thought to be important features to abolish TG2's conformational dynamism and ultimately inhibit both its activities. However, we recently demonstrated that the enzyme was unable to bind guanosine triphosphate (GTP) when catalytically inactivated by small molecule inhibitors. In this study, we designed a library of models targeting covalent inhibitors of progressively smaller sizes (15 to 4 atoms in length). We evaluated their ability to inactivate TG2 by measuring their respective kinetic parameters kinact and KI. Their impact on the enzyme's ability to bind GTP was then evaluated and subsequently correlated to the conformational state of the enzyme, as determined via native PAGE and capillary electrophoresis. All irreversible inhibitors evaluated herein locked TG2 in its open conformation and precluded GTP binding. Therefore, we conclude that steric bulk and structural complexity are not necessary factors to consider when designing TG2 inhibitors to abolish G-protein activity.


Assuntos
Alquilantes , Domínio Catalítico , Proteínas de Ligação ao GTP , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases , Transglutaminases/química , Transglutaminases/metabolismo , Transglutaminases/antagonistas & inibidores , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/metabolismo , Humanos , Alquilantes/química , Alquilantes/farmacologia , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Compostos de Sulfidrila/química , Compostos de Sulfidrila/farmacologia , Conformação Proteica , Cinética , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia
3.
Bioconjug Chem ; 33(11): 2097-2102, 2022 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-36315583

RESUMO

Alkylating agents for nucleic acids have been widely used in cancer chemotherapy, as well as in chemical biology for strong inhibitors and tagging methods. We provide a series of reactive OFF-ON type alkylating agents which enable the reactivity modulation toward G-quadruplex (G4) DNA and RNA. Due to the protonation-accelerated process and equilibrium elimination method, the amine leaving groups show highly reactive and storable properties in an extensive investigation of vinyl quinazolinone (VQ) precursors with different leaving groups.


Assuntos
Alquilantes , Quadruplex G , Alquilantes/química , DNA/química , RNA
4.
Anticancer Agents Med Chem ; 22(8): 1478-1495, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34382529

RESUMO

Cancer is considered one of the gruelling challenges and poses a grave health hazard across the globe. According to the International Agency for Research on Cancer (IARC), new cancer cases increased to 18.1 million in 2018, with 9.6 million deaths, bringing the global cancer rate to 23.6 million by 2030. In 1942, the discovery of nitrogen mustard as an alkylating agent was a tremendous breakthrough in cancer chemotherapy. It acts by binding to the DNA, and creating cross linkages between the two strands, leading to halt of DNA replication and eventual cell death. Nitrogen lone pairs of 'nitrogen mustard' produce an intermediate 'aziridinium ion' at the molecular level, which is very reactive towards DNA of tumour cells, resulting in multiple side effects with therapeutic consequences. Owing to its high reactivity and peripheral cytotoxicity, several improvements have been made with structural modifications for the past 75 years to enhance its efficacy and improve the direct transport of drugs to the tumour cells. Alkylating agents were among the first non-hormonal substances proven to be active against malignant cells and also the most valuable cytotoxic therapies available for the treatment of leukaemia and lymphoma patients. This review focus on the versatile use of alkylating agents and the Structure Activity Relationship (SAR) of each class of these compounds. This could provide an understanding for design and synthesis of new alkylating agents having enhanced target specificity and adequate bioavailability.


Assuntos
Antineoplásicos , Leucemia , Neoplasias , Alquilantes/química , Alquilantes/farmacologia , Alquilantes/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , DNA/química , Humanos , Leucemia/tratamento farmacológico , Mecloretamina/farmacologia , Mecloretamina/uso terapêutico , Neoplasias/tratamento farmacológico
5.
Int J Biol Macromol ; 193(Pt B): 1332-1339, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34742849

RESUMO

Forisomes are giant polyprotein complexes that undergo reversible conformational rearrangements from a spindle-like to a plug-like state in response to Ca2+ or changes in pH. They act as valves in the plant vasculature, and reproduce this function in vitro to regulate flow in microfluidic capillaries controlled by electro-titration. Heterologous expression in yeast or plants allows the large-scale production of tailor-made artificial forisomes for technical applications. Here we investigated the unexpected disintegration of artificial forisomes in response to Ca2+ following the deletion of the M1 motif in the MtSEO-F1 protein or the replacement of all four conserved cysteine residues therein. This phenomenon could be mimicked in wild-type forisomes under reducing conditions by adding a thiol alkylating agent. We propose a model in which reversible changes in forisome structure depend on cysteine residues with ambiguous redox states, allowing the formation of intermolecular disulfide bridges (confirmed by mass spectrometry) as well as noncovalent thiol interactions to connect forisome substructures in the dispersed state. This is facilitated by the projection of the M1 motif from the MtSEO-F1 protein as part of an extended loop. Our findings support the rational engineering of disintegrating forisomes to control the release of peptides or enzymes in microfluidic systems.


Assuntos
Cisteína/química , Proteínas de Plantas/química , Plantas/química , Alquilantes/química , Dissulfetos/química , Oxirredução , Compostos de Sulfidrila/química
6.
J Am Chem Soc ; 143(45): 18960-18976, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34726902

RESUMO

DNA interstrand cross-links (ICLs) are extremely deleterious and structurally diverse, driving the evolution of ICL repair pathways. Discovering ICL-inducing agents is, thus, crucial for the characterization of ICL repair pathways and Fanconi anemia, a genetic disease caused by mutations in ICL repair genes. Although several studies point to oxidative stress as a cause of ICLs, oxidative stress-induced cross-linking events remain poorly characterized. Also, polycyclic aromatic amines, potent environmental carcinogens, have been implicated in producing ICLs, but their identities and sequences are unknown. To close this knowledge gap, we tested whether ICLs arise by the oxidation of 8-arylamino-2'-deoxyadenosine (ArNHdA) lesions, adducts produced by arylamino carcinogens. Herein, we report that ArNHdA acts as a latent cross-linking agent to generate ICLs under oxidative conditions. The formation of an ICL from 8-aminoadenine, but not from 8-aminoguanine, highlights the specificity of 8-aminopurine-mediated ICL production. Under the influence of the reactive oxygen species (ROS) nitrosoperoxycarbonate, ArNHdA (Ar = biphenyl, fluorenyl) lesions were selectively oxidized to generate ICLs. The cross-linking reaction may occur between the C2-ArNHdA and N2-dG, presumably via oxidation of ArNHdA into a reactive diiminoadenine intermediate followed by the nucleophilic attack of the N2-dG on the diiminoadenine. Overall, ArNHdA-mediated ICLs represent rare examples of ROS-induced ICLs and polycyclic aromatic amine-mediated ICLs. These results reveal novel cross-linking chemistry and the genotoxic effects of arylamino carcinogens and support the hypothesis that C8-modified adenines with low redox potential can cause ICLs in oxidative stress.


Assuntos
Alquilantes/química , Compostos de Anilina/química , Reagentes de Ligações Cruzadas/química , Dano ao DNA/efeitos dos fármacos , DNA/química , Desoxiadenosinas/química , Carbonatos/química , Adutos de DNA/síntese química , Nitratos/química , Oxirredução
7.
Molecules ; 26(18)2021 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-34576931

RESUMO

Post-translational modifications (PTMs) are used by organisms to control protein structure and function after protein translation, but their study is complicated and their roles are not often well understood as PTMs are difficult to introduce onto proteins selectively. Designing reagents that are both good mimics of PTMs, but also only modify select amino acid residues in proteins is challenging. Frequently, both a chemical warhead and linker are used, creating a product that is a misrepresentation of the natural modification. We have previously shown that biotin-chloromethyl-triazole is an effective reagent for cysteine modification to give S-Lys derivatives where the triazole is a good mimic of natural lysine acylation. Here, we demonstrate both how the reactivity of the alkylating reagents can be increased and how the range of triazole PTM mimics can be expanded. These new iodomethyl-triazole reagents are able to modify a cysteine residue on a histone protein with excellent selectivity in 30 min to give PTM mimics of acylated lysine side-chains. Studies on the more complicated, folded protein SCP-2L showed promising reactivity, but also suggested the halomethyl-triazoles are potent alkylators of methionine residues.


Assuntos
Proteínas/química , Proteínas/metabolismo , Triazóis/química , Alquilantes/química , Cisteína/química , Glicosilação , Histonas/química , Metionina/química , Processamento de Proteína Pós-Traducional , Triazóis/síntese química
8.
Artigo em Inglês | MEDLINE | ID: mdl-33429128

RESUMO

A novel analytical method was developed for the quantification of glutathione hydropersulfide (G-SSH) in biological samples by high-performance liquid chromatography (HPLC) with post-column derivatization. G-SSH was treated with iodoacetamide as an alkylating agent for 5 min at 37 °C, and the resultant acetamide-labeled G-SSH (G-SS-acetamide) was subjected to HPLC. After separation on a reversed-phase column, G-SS-acetamide was quantified by detection using a post-column reaction with orthophthalaldehyde under alkaline conditions. The standard G-SS-acetamide was synthesized through the S-S exchange reaction between oxidized glutathione and 2-mercaptoacetamide. It should be noted that some types of alkylating agents, including N-ethylmaleimide and monobromobimane, cleave the polysulfide chains of polysulfides that consist of glutathione, resulting in the production of alkylated G-SSHs. We confirmed that iodoacetamide did not enhance the cleavage of acetamide-labeled glutathione trihydropersulfide (G-SSS-acetamide). The lowest quantification limit was estimated to be 25 nM for G-SS-acetamide. This method can be useful for studying the dynamics of sulfane sulfur in glutathione-containing matrices.


Assuntos
Alquilantes/química , Cromatografia Líquida de Alta Pressão/métodos , Dissulfetos , Glutationa/análogos & derivados , Iodoacetamida/química , Linhagem Celular Tumoral , Dissulfetos/análise , Dissulfetos/química , Dissulfetos/metabolismo , Glutationa/análise , Glutationa/química , Glutationa/metabolismo , Humanos , o-Ftalaldeído/química
9.
Antioxid Redox Signal ; 34(18): 1407-1419, 2021 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-33198504

RESUMO

Aims: Persulfides and other reactive sulfur species are endogenously produced in large amounts in vivo and participate in multiple cellular functions underlying physiological and pathological conditions. In the current study, we aimed to develop an ideal alkylating agent for use in sulfur metabolomics, particularly targeting persulfides and other reactive sulfur species, with minimal artifactual decomposition. Results: We synthesized a tyrosine-based iodoacetamide derivative, N-iodoacetyl l-tyrosine methyl ester (TME-IAM), which reacts with the thiol residue of cysteine identically to that of ß-(4-hydroxyphenyl)ethyl iodoacetamide (HPE-IAM), a commercially available reagent. Our previous study revealed that although various electrophilic alkylating agents readily decomposed polysulfides, HPE-IAM exceptionally stabilized the polysulfides by inhibiting their alkaline hydrolysis. The newly synthesized TME-IAM stabilizes oxidized glutathione tetrasulfide more efficiently than other alkylating agents, including HPE-IAM, iodoacetamide, and monobromobimane. In fact, our quantitative sulfur-related metabolome analysis showed that TME-IAM is a more efficient trapping agent for endogenous persulfides/polysulfides containing a larger number of sulfur atoms in mouse liver and brain tissues compared with HPE-IAM. Innovation and Conclusions: We developed a novel iodoacetamide derivative, which is the most ideal reagent developed to date for detecting endogenous persulfides/polysulfides formed in biological samples, such as cultured cells, tissues, and plasma. This new probe may be useful for investigating the unique chemical properties of reactive persulfides, thereby enabling identification of novel reactive sulfur metabolites that remain unidentified because of their instability, and thus can be applied in high-precision sulfur metabolomics in redox biology and medicine. We did not perform any clinical experiments in this study. Antioxid. Redox Signal. 34, 1407-1419.


Assuntos
Acetamidas/síntese química , Alquilantes/síntese química , Metabolômica/métodos , Sulfetos/análise , Acetamidas/química , Alquilantes/química , Animais , Cromatografia Líquida , Células HEK293 , Humanos , Iodoacetamida/química , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Espécies Reativas de Nitrogênio
10.
Sci Rep ; 10(1): 12772, 2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32728172

RESUMO

Folate receptor (FR)-targeted small molecule drug conjugates (SMDCs) have shown promising results in early stage clinical trials with microtubule destabilizing agents, such as vintafolide and EC1456. In our effort to develop FR-targeted SMDCs with varying mechanisms of action, we synthesized EC2629, a folate conjugate of a DNA crosslinking agent based on a novel DNA-alkylating moiety. This agent was found to be extremely potent with an in vitro IC50 ~ 100× lower than folate SMDCs constructed with various microtubule inhibitors. EC2629 treatment of nude mice bearing FR-positive KB human xenografts led to cures in 100% of the test animals with very low dose levels (300 nmol/kg) following a convenient once a week schedule. The observed activity was not accompanied by any noticeable weight loss (up to 20 weeks post end of dosing). Complete responses were also observed against FR-positive paclitaxel (KB-PR) and cisplatin (KB-CR) resistant models. When evaluated against FR-positive patient derived xenograft (PDX) models of ovarian (ST070), endometrial (ST040) and triple negative breast cancers (ST502, ST738), EC2629 showed significantly greater anti-tumor activity compared to their corresponding standard of care treatments. Taken together, these studies thus demonstrated that EC2629, with its distinct DNA reacting mechanism, may be useful in treating FR-positive tumors, including those that are classified as drug resistant.


Assuntos
Antineoplásicos/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , DNA/química , Neoplasias do Endométrio/tratamento farmacológico , Receptores de Folato com Âncoras de GPI/química , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Alquilantes/química , Animais , Bovinos , Cisplatino/administração & dosagem , Cães , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Feminino , Ácido Fólico/análogos & derivados , Ácido Fólico/farmacologia , Humanos , Concentração Inibidora 50 , Células KB , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Paclitaxel/administração & dosagem , Ratos , Alcaloides de Vinca/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Chem Res Toxicol ; 33(9): 2219-2224, 2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32388971

RESUMO

The cellular outcomes of chemical exposure are as much about the cellular response to the chemical as it is an effect of the chemical. We are growing in our understanding of the genotoxic interaction between chemistry and biology. For example, recent data has revealed the biological basis for mutation induction curves for a methylating chemical, which has been shown to be dependent on the repair capacity of the cells. However, this is just one end point in the toxicity pathway from chemical exposure to cell death. Much remains to be known in order for us to predict how cells will respond to a certain dose. Methylating agents, a subset of alkylating agents, are of particular interest, because of the variety of adverse genetic end points that can result, not only at increasing doses, but also over time. For instance, methylating agents are mutagenic, their potency, for this end point, is determined by the cellular repair capacity of an enzyme called methylguanine DNA-methyltransferase (MGMT) and its ability to repair the induceed methyl adducts. However, methyl adducts can become clastogenic. Erroneous biological processing will convert mutagenic adducts to clastogenic events in the form of double strand breaks (DSBs). How the cell responds to DSBs is via a cascade of protein kinases, which is called the DNA damage response (DDR), which will determine if the damage is repaired effectively, via homologous recombination, or with errors, via nonhomologous end joining, or whether the cell dies via apoptosis or enters senescence. The fate of cells may be determined by the extent of damage and the resulting strength of DDR signaling. Therefore, thresholds of damage may exist that determine cell fate. Such thresholds would be dependent on each of the repair and response mechanisms that these methyl adducts stimulate. The molecular mechanism of how methyl adducts kill cells is still to be fully resolved. If we are able to quantify each of these thresholds of damage for a given cell, then we can ascertain, of the many adducts that are induced, what proportion of them are mutagenic, what proportion are clastogenic, and how many of these clastogenic events are toxic. This review examines the possibility of dose and damage thresholds for methylating agents, from the perspective of the underlying evolutionary mechanisms that may be accountable.


Assuntos
Alquilantes/efeitos adversos , Inibidores Enzimáticos/efeitos adversos , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Alquilantes/química , Animais , Inibidores Enzimáticos/química , Humanos , Metilação/efeitos dos fármacos , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo
12.
Curr Comput Aided Drug Des ; 16(2): 190-195, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31389315

RESUMO

BACKGROUND: Reckoning molecular topological indices of drug structures gives the data about the underlying topology of these drug structures. Novel anticancer drugs have been leading by researchers to produce ideal drugs. MATERIALS AND METHODS: Pharmacological properties of these new drug agents explored by utilizing simulation strategies. Topological indices additionally have been utilized to research pharmacological properties of some drug structures. Novel alkylating agents based anticancer drug candidates and ve-degree molecular topological indices have been introduced recently. RESULTS AND CONCLUSION: In this study we calculate ve-degree atom-bond connectivity, harmonic, geometric-arithmetic and sum-connectivity molecular topological indices for the newly defined alkylating agents based dual-target anticancer drug candidates.


Assuntos
Alquilantes/química , Antineoplásicos/química , Desenho de Fármacos , Relação Quantitativa Estrutura-Atividade
13.
Chem Res Toxicol ; 33(2): 625-633, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-31841318

RESUMO

DNA methylating agents are abundant in the environment and are sometimes used in cancer chemotherapy. They react with DNA to form methyl-DNA adducts and byproduct lesions that can be both toxic and mutagenic. Foremost among the mutagenic lesions is O6-methylguanine (m6G), which base pairs with thymine during replication to cause GC → AT mutations. The gpt delta C57BL/6J mouse strain of Nohmi et al. (Mol. Mutagen 1996, 28, 465-70) reliably produces mutational spectra of many DNA damaging agents. In this work, mouse embryo fibroblasts (MEFs) were made from gpt delta C57BL/6J mice and evaluated as a screening tool to determine the qualitative and quantitative features of mutagenesis by N-methyl-N-nitrosourea (MNU), a direct-acting DNA alkylator that serves as a model for environmental N-nitrosamines, such as N-nitrosodimethylamine and therapeutic agents such as Temozolomide. The DNA repair protein MGMT (O6-methylguanine DNA methyltransferase) protects against environmental mutagenesis by DNA methylating agents and, by removing m6G, limits the therapeutic potential of Temozolomide in cancer therapy. The gpt delta MEFs were treated with MNU to establish dose-dependent toxicity. In parallel, MNU mutagenicity was determined in the presence and absence of the MGMT inhibitor AA-CW236 (4-(2-(5-(chloromethyl)-4-(4-(trifluoromethoxy)phenyl)-1H-1,2,3-triazol-1-yl)ethyl)-3,5-dimethylisoxazole). With and without the inhibitor, the principal mutagenic event of MNU was GC → AT, but more mutations were observed when the inhibitor was present. Evidence that the mutagenic lesion was m6G was based on mass spectral data collected using O6-methyl-d3-guanine as an internal standard; m6G levels were higher in AA-CW236 treated MEFs by an amount proportional to the higher mutation frequency seen in the same cells. This work establishes gpt delta MEFs as a versatile tool for probing mutagenesis by environmental and therapeutic agents and as a cell culture model in which chemical genetics can be used to determine the impact of DNA repair on biological responses to DNA damaging agents.


Assuntos
Alquilantes/farmacologia , Metilases de Modificação do DNA/antagonistas & inibidores , Enzimas Reparadoras do DNA/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Metilnitrosoureia/farmacologia , Mutagênese/efeitos dos fármacos , Proteínas Supressoras de Tumor/antagonistas & inibidores , Alquilantes/química , Animais , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Inibidores Enzimáticos/química , Fibroblastos/metabolismo , Metilnitrosoureia/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Supressoras de Tumor/metabolismo
14.
Chem Res Toxicol ; 32(12): 2517-2525, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31726825

RESUMO

Nitrogen mustards have long been used in cancer chemotherapy, and their cytotoxicity has traditionally been attributed to the formation of DNA interstrand cross-links and DNA monoalkylation. Recent studies have shown that exposure to nitrogen mustards also induces the formation of DNA-protein cross-links (DPCs) via bridging between N7 of a deoxyguanosine residue in the DNA and the side chain of a Cys residue in the protein. However, the formation of nitrogen mustard-induced DNA-histone cross-links has never been observed. Herein, we demonstrate that treating reconstituted nucleosome core particles (NCPs) with the nitrogen mustard mechlorethamine results in the formation of DNA-histone cross-links in addition to DNA monoalkylation and interstrand cross-link formation. The yields of these three types of DNA lesions in the NCPs decreased in the following order: DNA monoalkylation ≫ DNA interstrand cross-links > DNA-histone cross-links. Mechanistic studies involving tailless histones and competitive inhibition by a polyamine demonstrated that Lys residues in the N- and C-terminal tails of the histones were the predominant sites involved in DNA-histone cross-link formation. Given that NCPs are the fundamental repeating units of chromatin in eukaryotes, our findings suggest that nitrogen mustard-induced formation of DNA-histone cross-links may occur in living cells and that DPC formation may contribute to the cytotoxicity of nitrogen mustards.


Assuntos
Alquilantes/química , Reagentes de Ligações Cruzadas/química , DNA/efeitos dos fármacos , Histonas/efeitos dos fármacos , Mecloretamina/química , Nucleossomos/efeitos dos fármacos , Sequência de Aminoácidos , Animais , DNA/química , Histonas/química , Masculino , Modelos Químicos , Nucleossomos/química , Salmão , Espermatozoides/química
15.
Chem Commun (Camb) ; 55(87): 13140-13143, 2019 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-31617528

RESUMO

In this work, we depleted glutathione (GSH) by releasing SO2 with internal stimulus GSH itself, and also selectively marked the cancer cells followed by release of anticancer drug using another orthogonal stimulus i.e., two-photon (TP) NIR light by a single naphthalene based chromophore (TP absorbance 77 GM and uncaging cross-section 21 GM). We demonstrated the improved therapeutic efficacy of chlorambucil by the stepwise dual stimuli approach and dual surveillance of both the drug uncaging process in real-time using in vitro studies.


Assuntos
Alquilantes/farmacologia , Antineoplásicos Alquilantes/farmacologia , Clorambucila/farmacologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Naftalenos/farmacologia , Fótons , Alquilantes/química , Antineoplásicos Alquilantes/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Clorambucila/química , Liberação Controlada de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Glutationa/metabolismo , Humanos , Raios Infravermelhos , Estrutura Molecular , Naftalenos/química , Imagem Óptica , Dióxido de Enxofre/metabolismo
16.
ACS Sens ; 4(7): 1791-1797, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31299153

RESUMO

A pyrene-containing salicylic acid derivative (4) was found to be low in fluorescence, but its derivative pyrene-containing methyl salicylate (3) was found to be highly fluorescent in aqueous solution. This derivative has been tested in solution and found to be superior in the fluorogenic assay of pharmaceutical compounds, detection of chemical warfare agents, a preliminary toxicology test, mutagenicity of medicinal compounds, and other chemical analyses, including trimethylsilyl diazomethane; alkyl bromides and iodides; a sulfur mustard mimic 2-chloroethyl ethyl sulfide; and anticancer drugs, busulfan and pipobroman. The salicylic acid derivative (4) was applied as a fluorogenic probe for the detection of alkylating agents by esterification and generating fluorescence at 475 nm in solutions at low concentrations.


Assuntos
Alquilantes/análise , Corantes Fluorescentes/química , Pirenos/química , Salicilatos/química , Alquilantes/química , Antineoplásicos/análise , Antineoplásicos/química , Bussulfano/análise , Bussulfano/química , Substâncias para a Guerra Química/análise , Substâncias para a Guerra Química/química , Corantes Fluorescentes/síntese química , Gás de Mostarda/análogos & derivados , Gás de Mostarda/análise , Gás de Mostarda/química , Pipobromano/análise , Pipobromano/química , Pirenos/síntese química , Salicilatos/síntese química , Espectrometria de Fluorescência , Temozolomida/análise , Temozolomida/química
17.
Bioorg Med Chem Lett ; 29(17): 2455-2458, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31350125

RESUMO

Antibody-drug conjugates (ADCs) incorporating potent indolinobenzodiazepine (IGN) DNA alkylators as the cytotoxic payload are currently undergoing clinical evaluation. The optimized design of these payloads consists of an unsymmetrical dimer possessing both an imine and an amine effectively eliminating DNA crosslinking and demonstrating improved tolerability in mice. Here we present an alternate approach to generating DNA alkylating ADCs by linking the IGN monomer with a biaryl system which has a high DNA binding affinity to potentially enhance tolerability. These BIA ADCs were found to be highly cytotoxic in vitro and demonstrated potent antitumor activity in vivo.


Assuntos
Alquilantes/química , Desenho de Fármacos , Imunoconjugados/química , Animais , Anticorpos Monoclonais/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Humanos , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Camundongos , Camundongos SCID , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Relação Estrutura-Atividade , Transplante Heterólogo
18.
Cancer Sci ; 110(10): 3296-3305, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31348600

RESUMO

Tissue factor (TF) is known to be overexpressed in various cancers including pancreatic cancer. The upregulation of TF expression has been observed not only in tumor cells, but also in tumor stromal cells. Because of the potential of TF as a delivery target, several studies investigated the effectiveness of Ab-drug conjugates (ADCs) against TF for cancer therapy. However, it is still unclear whether anti-TF ADC can exert toxicity against both tumor cells and tumor stromal cells. Here, we prepared ADC using a rat anti-mouse TF mAb (clone.1157) and 2 types of in vivo murine pancreatic cancer models, one s.c. and other orthotopic with an abundant tumor stroma. We also compared the feasibility of bis-alkylating conjugation (bisAlk) with that of conventional maleimide-based conjugation (MC). In the s.c. models, anti-TF ADC showed greater antitumor effects than control ADC. The results also indicated that the bisAlk linker might be more suitable than the MC linker for cancer treatments. In the orthotopic model, anti-TF ADC showed greater in vivo efficacy and more extended survival time control ADC. Treatment with anti-TF ADC (20 mg/kg, three times a week) did not affect mouse body weight changes in any in vivo experiment. Furthermore, immunofluorescence staining indicated that anti-TF ADC delivered agents not only to TF-positive tumor cells, but also to TF-positive tumor vascular endothelial cells and other tumor stromal cells. We conclude that anti-TF ADC should be a selective and potent drug for pancreatic cancer therapy.


Assuntos
Alquilantes/química , Antineoplásicos Imunológicos/administração & dosagem , Imunoconjugados/administração & dosagem , Maleimidas/química , Neoplasias Pancreáticas/tratamento farmacológico , Tromboplastina/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Esquema de Medicação , Feminino , Humanos , Imunoconjugados/química , Imunoconjugados/farmacologia , Camundongos , Camundongos Transgênicos , Neoplasias Pancreáticas/metabolismo , Ratos , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Anticancer Agents Med Chem ; 19(9): 1080-1102, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30834842

RESUMO

BACKGROUND & OBJECTIVE: Nitrogen mustard derivatives form one of the major classes of anti-cancer agents in USFDA approved drugs list. These are polyfunctional alkylating agents which are distinguished by a unique mechanism of adduct formation with DNA involving cross-linking between guanine N-7 of one strand of DNA with the other. The generated cross-linking is irreversible and leads to cell apoptosis. Hence it is of great interest to explore this class of anticancer alkylating agents. METHODS: An exhaustive list of reviews, research articles, patents, books, patient information leaflets, and orange book is presented and the contents related to nitrogen mustard anti-cancer agents have been reviewed. Attempts are made to present synthesis schemes in a simplified manner. The mechanism of action of the drugs and their side effects are also systematically elaborated. RESULTS: This review provides a platform for understanding all aspects of such drugs right from synthesis to their mechanism of action and side effects, and lists USFDA approved ANDA players among alkylating anticancer agents in the current market. CONCLUSION: Perusing this article, generic scientists will be able to access literature information in this domain easily to gain insight into the nitrogen mustard alkylating agents for further ANDA development. It will help the scientific and research community to continue their pursuit for the design of newer and novel heterocyclic alkylating agents of this class in the coming future.


Assuntos
Alquilantes/farmacologia , Antineoplásicos Alquilantes/farmacologia , Neoplasias/tratamento farmacológico , Compostos de Mostarda Nitrogenada/farmacologia , Alquilantes/síntese química , Alquilantes/química , Antineoplásicos Alquilantes/síntese química , Antineoplásicos Alquilantes/química , Proliferação de Células/efeitos dos fármacos , Humanos , Neoplasias/patologia , Compostos de Mostarda Nitrogenada/síntese química , Compostos de Mostarda Nitrogenada/química , Estados Unidos , United States Food and Drug Administration
20.
J Am Chem Soc ; 141(10): 4257-4263, 2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30601664

RESUMO

The runt-related transcription factor (RUNX) family has been associated with cancer development. The binding of RUNX family members to specific DNA sequences is hypothesized to promote the expression of downstream genes and cause cancer proliferation. On the basis of this proposed mechanism of cancer growth, we developed conjugate 1, which inhibits the binding of RUNX to its target DNA. Conjugate 1 is a DNA-alkylating pyrrole-imidazole (PI) polyamide conjugate containing chlorambucil as an anticancer agent. Conjugate 1 was reported to have a marked anticancer effect in mouse models of acute myeloid leukemia. Although the effectiveness of 1 has been demonstrated in vivo, the detailed mechanism by which it alkylates DNA is unknown. Here, we chemically elucidated the molecular characteristics of conjugate 1 to confirm its potential as a RUNX-inhibiting drug. We also generated an alternative conjugate 2, which targets the same DNA sequence, by replacing one pyrrole with ß-alanine. Comparison of the characteristics of conjugates 1 and 2 suggested that reaction selectivity and binding affinity to the RUNX-binding sequence were improved by the introduction of ß-alanine. These findings indicate the possibility of DNA-alkylating PI polyamides as candidates for cancer chemotherapeutics.


Assuntos
Alquilantes/farmacologia , Antineoplásicos/farmacologia , DNA/química , Nylons/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Alquilantes/química , Alquilação , Antineoplásicos/química , Linhagem Celular Tumoral , Clorambucila/análogos & derivados , Clorambucila/farmacologia , DNA/metabolismo , Humanos , Imidazóis/química , Imidazóis/farmacologia , Nylons/química , Ligação Proteica/efeitos dos fármacos , Pirróis/química , Pirróis/farmacologia , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA