Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Pharmacol Exp Ther ; 373(2): 220-229, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32094296

RESUMEN

PF06821497 has been identified as an orally available small-molecule enhancer of zeste homolog 2 inhibitor. The objectives of the present study were to characterize pharmacokinetic-pharmacodynamic-disease relationships of PF06821497 in xenograft mouse models with diffuse large B-cell lymphoma (Karpas422). An indirect-response model reasonably fit dose-dependent pharmacodynamic responses [histone H3 on lysine 27 (H3K27) me3 inhibition] with an unbound EC 50 of 76 nM, whereas a signal-transduction model sufficiently fit dose-dependent disease responses (tumor growth inhibition) with an unbound tumor stasis concentration (T sc ) of 168 nM. Thus, effective concentration for 70% of maximal effect (EC70) for H3K27me3 inhibition was roughly comparable to T sc , suggesting that 70% H3K27me3 inhibition could be required for tumor stasis. Consistently, an integrated pharmacokinetic-pharmacodynamic-disease model adequately describing tumor growth inhibition also suggested that ∼70% H3K27me3 inhibition was associated with tumor stasis. Based on these results, we would propose that an EC70 estimate for H3K27me3 inhibition corresponding to tumor stasis could be considered a minimum target efficacious concentration of PF06821497 in cancer patients. SIGNIFICANCE STATEMENT: Using a mathematical modeling approach, the quantitative relationships of an orally available anticancer small-molecule enhancer of zeste homolog 2 inhibitor, PF06821497, were characterized among pharmacokinetics, pharmacodynamic biomarker inhibition, and disease responses in nonclinical xenograft models with diffuse large B-cell lymphoma. The modeling results suggest that >70% histone H3 on lysine 27 (H3K27) me3 inhibition would be required for tumor stasis (i.e., 100% tumor growth inhibition). Accordingly, we would propose that an effective concentration for 70% of maximal effect estimate for H3K27me3 inhibition could be considered a minimum target efficacious concentration of PF06821497 in cancer patients.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Epigénesis Genética/efectos de los fármacos , Histonas/antagonistas & inhibidores , Isoquinolinas , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Piridinas , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Femenino , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacocinética , Isoquinolinas/farmacología , Ratones , Modelos Biológicos , Piridinas/administración & dosificación , Piridinas/farmacocinética , Piridinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Indian J Microbiol ; 60(3): 283-296, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32655196

RESUMEN

Human pathogens need to overcome an elaborate network of host defense mechanisms in order to establish their infection, colonization, proliferation and eventual dissemination. The interaction of pathogens with different effector molecules of the immune system results in their neutralization and elimination from the host. The complement system is one such integral component of innate immunity that is critically involved in the early recognition and elimination of the pathogen. Hence, under this immune pressure, all virulent pathogens capable of inducing active infections have evolved immune evasive strategies that primarily target the complement system, which plays an essential and central role for host defense. Recent reports on several bacterial pathogens have elucidated the molecular mechanisms underlying complement evasion, inhibition of opsonic phagocytosis and cell lysis. This review aims to comprehensively summarize the recent findings on the various strategies adopted by pathogenic bacteria to escape complement-mediated clearance.

3.
PLoS Genet ; 7(2): e1001286, 2011 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-21304883

RESUMEN

How epigenetic information is propagated during somatic cell divisions is still unclear but is absolutely critical for preserving gene expression patterns and cellular identity. Here we show an unanticipated mechanism for inheritance of DNA methylation patterns where the epigenetic mark not only recruits the catalyzing enzyme but also regulates the protein level, i.e. the enzymatic product (5-methylcytosine) determines the level of the methylase, thus forming a novel homeostatic inheritance system. Nucleosomes containing methylated DNA stabilize de novo DNA methyltransferases, DNMT3A/3B, allowing little free DNMT3A/3B enzymes to exist in the nucleus. Stabilization of DNMT3A/3B on nucleosomes in methylated regions further promotes propagation of DNA methylation. However, reduction of cellular DNA methylation levels creating more potential CpG substrates counter-intuitively results in a dramatic decrease of DNMT3A/3B proteins due to diminished nucleosome binding and subsequent degradation of the unstable free proteins. These data show an unexpected self-regulatory inheritance mechanism that not only ensures somatic propagation of methylated states by DNMT1 and DNMT3A/3B enzymes but also prevents aberrant de novo methylation by causing degradation of free DNMT3A/3B enzymes.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Epigénesis Genética , Nucleosomas/enzimología , 5-Metilcitosina/metabolismo , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN Metiltransferasa 3A , Estabilidad de Enzimas , Perfilación de la Expresión Génica , Silenciador del Gen , Vectores Genéticos , Células HCT116 , Humanos , ADN Metiltransferasa 3B
4.
Trends Pharmacol Sci ; 45(3): 243-254, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38383216

RESUMEN

Lysine acetyltransferases (KATs) are a family of epigenetic enzymes involved in the regulation of gene expression; they represent a promising class of emerging drug targets. The frequent molecular dysregulation of these enzymes, as well as their mechanistic links to biological functions that are crucial to cancer, have led to exploration around the development of small-molecule inhibitors against KATs. Despite early challenges, recent advances have led to the development of potent and selective enzymatic and bromodomain (BRD) KAT inhibitors. In this review we discuss the discovery and development of new KAT inhibitors and their application as oncology therapeutics. Additionally, new chemically induced proximity approaches are presented, offering opportunities for unique target selectivity profiles and tissue-specific targeting of KATs. Emerging clinical data for CREB binding protein (CREBBP)/EP300 BRD inhibitors and KAT6 catalytic inhibitors indicate the promise of this target class in cancer therapeutics.


Asunto(s)
Lisina Acetiltransferasas , Neoplasias , Humanos , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Lisina Acetiltransferasas/química , Lisina Acetiltransferasas/genética , Lisina Acetiltransferasas/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Neoplasias/tratamiento farmacológico
5.
PLoS Genet ; 6(4): e1000917, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20421991

RESUMEN

It was recently shown that a large portion of the human transcriptome can originate from within repetitive elements, leading to ectopic expression of protein-coding genes. However the mechanism of transcriptional activation of repetitive elements has not been definitively elucidated. For the first time, we directly demonstrate that hypomethylation of retrotransposons can cause altered gene expression in humans. We also reveal that active LINE-1s switch from a tetranucleosome to dinucleosome structure, acquiring H2A.Z- and nucleosome-free regions upstream of TSSs, previously shown only at active single-copy genes. Hypomethylation of a specific LINE-1 promoter was also found to induce an alternate transcript of the MET oncogene in bladder tumors and across the entire urothelium of tumor-bearing bladders. These data show that, in addition to contributing to chromosomal instability, hypomethylation of LINE-1s can alter the functional transcriptome and plays a role not only in human disease but also in disease predisposition.


Asunto(s)
Elementos de Nucleótido Esparcido Largo/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-met/genética , Receptores de Factores de Crecimiento/genética , Neoplasias de la Vejiga Urinaria/genética , Adulto , Anciano , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Metilación de ADN/genética , Humanos , Persona de Mediana Edad , Oncogenes , Activación Transcripcional
6.
Cancer Lett ; 552: 215981, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36341997

RESUMEN

Inhibitors of dihydroorotate dehydrogenase (DHODH), a key enzyme for de novo synthesis of pyrimidine nucleotides, have failed in clinical trials for various cancers despite robust efficacy in preclinical animal models. To probe for druggable mediators of DHODH inhibitor resistance, we performed a combination screen with a small molecule library against pancreatic cancer cell lines that are highly resistant to the DHODH inhibitor brequinar (BQ). The screen revealed that CNX-774, a preclinical Bruton tyrosine kinase (BTK) inhibitor, sensitizes resistant cell lines to BQ. Mechanistic studies showed that this effect is independent of BTK and instead results from inhibition of equilibrative nucleoside transporter 1 (ENT1) by CNX-774. We show that ENT1 mediates BQ resistance by taking up extracellular uridine, which is salvaged to generate pyrimidine nucleotides in a DHODH-independent manner. In BQ-resistant cell lines, BQ monotherapy slowed proliferation and caused modest pyrimidine nucleotide depletion, whereas combination treatment with BQ and CNX-774 led to profound cell viability loss and pyrimidine starvation. We also identify N-acetylneuraminic acid accumulation as a potential marker of the therapeutic efficacy of DHODH inhibitors. In an aggressive, immunocompetent pancreatic cancer mouse model, combined targeting of DHODH and ENT1 dramatically suppressed tumor growth and prolonged mouse survival. Overall, our study defines CNX-774 as a previously uncharacterized ENT1 inhibitor and provides strong proof of concept support for dual targeting of DHODH and ENT1 in pancreatic cancer.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Neoplasias Pancreáticas , Ratones , Animales , Dihidroorotato Deshidrogenasa , Tranportador Equilibrativo 1 de Nucleósido/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Pirimidinas/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Nucleótidos de Pirimidina , Neoplasias Pancreáticas
7.
Cell Chem Biol ; 30(10): 1191-1210.e20, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37557181

RESUMEN

KAT6A, and its paralog KAT6B, are histone lysine acetyltransferases (HAT) that acetylate histone H3K23 and exert an oncogenic role in several tumor types including breast cancer where KAT6A is frequently amplified/overexpressed. However, pharmacologic targeting of KAT6A to achieve therapeutic benefit has been a challenge. Here we describe identification of a highly potent, selective, and orally bioavailable KAT6A/KAT6B inhibitor CTx-648 (PF-9363), derived from a benzisoxazole series, which demonstrates anti-tumor activity in correlation with H3K23Ac inhibition in KAT6A over-expressing breast cancer. Transcriptional and epigenetic profiling studies show reduced RNA Pol II binding and downregulation of genes involved in estrogen signaling, cell cycle, Myc and stem cell pathways associated with CTx-648 anti-tumor activity in ER-positive (ER+) breast cancer. CTx-648 treatment leads to potent tumor growth inhibition in ER+ breast cancer in vivo models, including models refractory to endocrine therapy, highlighting the potential for targeting KAT6A in ER+ breast cancer.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/genética , Histonas/metabolismo , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Transducción de Señal , Línea Celular Tumoral
8.
Front Immunol ; 13: 1075662, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36713362

RESUMEN

Introduction: Bacillus anthracis is the causative agent for the lethal disease anthrax, primarily affecting animals and humans in close contact with an infected host. The pathogenicity of B. anthracis is attributed to the secreted exotoxins and their outer capsule. The host cell-binding exotoxin component "protective antigen" (PA) is reported to be a potent vaccine candidate. The aim of our study is to produce several PA constructs and analyze their vaccine potential. Methods: We have designed the various subunit, PA-based recombinant proteins, i.e., full-length Protective antigen (PA-FL), C-terminal 63 kDa fragment (PA63), Protective antigen domain 1-domain 4 chimeras (PA-D1-4) and protective antigen domain 4 (PA-D4) and analyzed their vaccine potential with different human-compatible adjuvants in the mouse model. We have optimized the process and successfully expressed our recombinant antigens as soluble proteins, except full-length PA. All the recombinant antigen formulations with three different adjuvants i.e., Addavax, Alhydrogel, and Montanide ISA 720, were immunized in different mouse groups. The vaccine efficacy of the formulations was analyzed by mouse serum antigen-specific antibody titer, toxin neutralization assay, and survival analysis of mouse groups challenged with a lethal dose of B. anthracis virulent spores. Results: We have demonstrated that the PA-FL addavax and PA63 addavax formulations were most effective in protecting spore-challenged mice and serum from the mice immunized with PAFL addavax, PA-FL alhydrogel, PA63 addavax, and PA63 alhydrogel formulations were equivalently efficient in neutralizing the anthrax lethal toxin. The higher levels of serum Th1, Th2, and Th17 cytokines in PA-FL addavax immunized mice correspond to the enhanced protection provided by the formulation in challenged mice. Discussion: We have demonstrated that the PA-FL addavax and PA63 addavax formulations exhibit equivalent efficiency as vaccine formulation both in a mouse model of anthrax and mammalian cell lines. However, PA63 is a smaller antigen than PA-FL and more importantly, PA63 is expressed as a soluble protein in E. coli, which imparts a translational advantage to PA63-based formulation. Thus, the outcome of our study has significant implications for the development of protective antigen-based vaccine formulations for human use against the lethal disease anthrax.


Asunto(s)
Vacunas contra el Carbunco , Carbunco , Bacillus anthracis , Animales , Ratones , Humanos , Carbunco/prevención & control , Hidróxido de Aluminio , Escherichia coli , Adyuvantes Inmunológicos , Exotoxinas , Mamíferos
9.
Mol Oncol ; 16(6): 1309-1328, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34669238

RESUMEN

Small-cell lung cancer (SCLC) is a heterogeneous disease, consisting of intratumoral and intertumoral neuroendocrine (ASCL1 and/or NEUROD1), mesenchymal-like, and YAP-driven transcriptional states. Lysine-specific demethylase 1 (LSD1; also known as KDM1A) inhibitors have recently been progressed to clinical trials in SCLC based on a promising preclinical antitumor activity. A potential clinical limitation of LSD1 inhibitors is the heterogeneous drug responses that have been observed in SCLC cell lines and patient-derived models. Based on these observations, we studied molecular and transcriptional signatures that predict patient response to this class of drug. Employing SCLC patient-derived transcriptional signatures, we define that SCLC cell lines sensitive to LSD1 inhibitors are enriched in neuroendocrine transcriptional markers, whereas cell lines enriched in a mesenchymal-like transcriptional program demonstrate intrinsic resistance to LSD1 inhibitors. We have identified a reversible, adaptive resistance mechanism to LSD1 inhibitors through epigenetic reprogramming to a TEAD4-driven mesenchymal-like state. Our data suggest that only a segment of SCLC patients, with a defined neuroendocrine differentiation state, will likely benefit from LSD1 inhibitors. It provides novel evidence for the selection of a TEAD4-driven mesenchymal-like subpopulation resistant to LSD1 inhibitors in SCLC patients that may require effective drug combinations to sustain effective clinical responses.


Asunto(s)
Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Proteínas de Unión al ADN/genética , Resistencia a Medicamentos , Histona Demetilasas , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas Musculares , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Factores de Transcripción de Dominio TEA , Factores de Transcripción/genética
10.
Carcinogenesis ; 31(1): 27-36, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19752007

RESUMEN

Epigenetic mechanisms are essential for normal development and maintenance of tissue-specific gene expression patterns in mammals. Disruption of epigenetic processes can lead to altered gene function and malignant cellular transformation. Global changes in the epigenetic landscape are a hallmark of cancer. The initiation and progression of cancer, traditionally seen as a genetic disease, is now realized to involve epigenetic abnormalities along with genetic alterations. Recent advancements in the rapidly evolving field of cancer epigenetics have shown extensive reprogramming of every component of the epigenetic machinery in cancer including DNA methylation, histone modifications, nucleosome positioning and non-coding RNAs, specifically microRNA expression. The reversible nature of epigenetic aberrations has led to the emergence of the promising field of epigenetic therapy, which is already making progress with the recent FDA approval of three epigenetic drugs for cancer treatment. In this review, we discuss the current understanding of alterations in the epigenetic landscape that occur in cancer compared with normal cells, the roles of these changes in cancer initiation and progression, including the cancer stem cell model, and the potential use of this knowledge in designing more effective treatment strategies.


Asunto(s)
Epigénesis Genética , Neoplasias/genética , Humanos , MicroARNs/genética , Neoplasias/patología , Células Madre Neoplásicas
11.
Nucleic Acids Res ; 36(14): 4689-98, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18628296

RESUMEN

Alterations in cytosine-5 DNA methylation are frequently observed in most types of human cancer. Although assays utilizing PCR amplification of bisulfite-converted DNA are widely employed to analyze these DNA methylation alterations, they are generally limited in throughput capacity, detection sensitivity, and or resolution. Digital PCR, in which a DNA sample is analyzed in distributive fashion over multiple reaction chambers, allows for enumeration of discrete template DNA molecules, as well as sequestration of non-specific primer annealing templates into negative chambers, thereby increasing the signal-to-noise ratio in positive chambers. Here, we have applied digital PCR technology to bisulfite-converted DNA for single-molecule high-resolution DNA methylation analysis and for increased sensitivity DNA methylation detection. We developed digital bisulfite genomic DNA sequencing to efficiently determine single-basepair DNA methylation patterns on single-molecule DNA templates without an interim cloning step. We also developed digital MethyLight, which surpasses traditional MethyLight in detection sensitivity and quantitative accuracy for low quantities of DNA. Using digital MethyLight, we identified single-molecule, cancer-specific DNA hypermethylation events in the CpG islands of RUNX3, CLDN5 and FOXE1 present in plasma samples from breast cancer patients.


Asunto(s)
Metilación de ADN , ADN de Neoplasias/sangre , Genómica/métodos , Reacción en Cadena de la Polimerasa/métodos , Sulfitos/química , Neoplasias de la Mama/genética , Islas de CpG , ADN de Neoplasias/metabolismo , Humanos , Técnicas Analíticas Microfluídicas
12.
Front Immunol ; 11: 462, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32296419

RESUMEN

Bacillus anthracis poly-γ-D-glutamic acid (PGA) capsule is an essential virulent factor that helps the bacterial pathogen to escape host immunity. Like other encapsulated bacterial species, the B. anthracis capsule may also inhibit complement-mediated clearance and ensure bacterial survival in the host. Previous reports suggest that B. anthracis spore proteins inhibit complement activation. However, the mechanism through which the B. anthracis capsule imparts a survival advantage to the active bacteria has not been demonstrated till date. Thus, to evaluate the role of the PGA capsule in evading host immunity, we have undertaken the present head-to-head comparative study of the phagocytosis and complement activation of non-encapsulated and encapsulated B. anthracis strains. The encapsulated virulent strain exhibited resistance toward complement-dependent and complement-independent bacterial phagocytosis by human macrophages. The non-encapsulated Sterne strain was highly susceptible to phagocytosis by THP-1 macrophages, after incubation with normal human serum (NHS), heat-inactivated serum, and serum-free media, thus indicating that the capsule inhibited both complement-dependent and complement-independent opsonic phagocytosis. An increased binding of C3b and its subsequent activation to C3c and C3dg, which functionally act as potent opsonins, were observed with the non-encapsulated Sterne strain compared with the encapsulated strain. Other known mediators of complement fixation, IgG, C-reactive protein (CRP), and serum amyloid P component (SAP), also bound more prominently with the non-encapsulated Sterne strain. Studies with complement pathway-specific, component-deficient serum demonstrated that the classical pathway was primarily involved in mediating C3b binding on the non-encapsulated bacteria. Both strains equally bound the complement regulatory proteins C4BP and factor H. Importantly, we demonstrated that the negative charge of the PGA capsule was responsible for the differential binding of the complement proteins between the non-encapsulated and encapsulated strains. At lower pH closer to the isoelectric point of PGA, the neutralization of the negative charge was associated with an increased binding of C3b and IgG with the encapsulated B. anthracis strain. Overall, our data have demonstrated that the B. anthracis capsule inhibits complement fixation and opsonization resulting in reduced phagocytosis by macrophages, thus allowing the bacterial pathogen to evade host immunity.


Asunto(s)
Carbunco/inmunología , Bacillus anthracis/fisiología , Macrófagos/inmunología , Ácido Poliglutámico/análogos & derivados , Antígenos Bacterianos/inmunología , Bacillus anthracis/patogenicidad , Cápsulas Bacterianas/inmunología , Cápsulas Bacterianas/metabolismo , Activación de Complemento , Complemento C3b/metabolismo , Humanos , Evasión Inmune , Proteínas Opsoninas/metabolismo , Fagocitosis , Ácido Poliglutámico/metabolismo , Unión Proteica , Células THP-1 , Virulencia
13.
Future Med Chem ; 12(15): 1415-1430, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32723083

RESUMEN

Immune dysfunction in the tumor microenvironment occurs through epigenetic changes in both tumor cells and immune cells that alter transcriptional programs driving cell fate and cell function. Oncogenic activation of the histone methyltransferase EZH2 mediates gene expression changes, governing tumor immunogenicity as well as differentiation, survival and activation states of immune lineages. Emerging preclinical studies have highlighted the potential for EZH2 inhibitors to reverse epigenetic immune suppression in tumors and combine with immune checkpoint therapies. However, EZH2 activity is essential for the development of lymphoid cells, performing critical immune effector functions within tumors. In this review, we highlight the complexity of EZH2 function in immune regulation which may impact the implementation of combination with immunotherapy agents in clinic.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/inmunología , Inmunoterapia , Neoplasias/terapia , Microambiente Tumoral/inmunología , Proteína Potenciadora del Homólogo Zeste 2/genética , Epigénesis Genética/genética , Epigénesis Genética/inmunología , Humanos , Neoplasias/inmunología
14.
Adv Ther ; 37(7): 3059-3082, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32445185

RESUMEN

Epigenetic processes are essential for normal development and the maintenance of tissue-specific gene expression in mammals. Changes in gene expression and malignant cellular transformation can result from disruption of epigenetic mechanisms, and global disruption in the epigenetic landscape is a key feature of cancer. The study of epigenetics in cancer has revealed that human cancer cells harbor both genetic alterations and epigenetic abnormalities that interplay at all stages of cancer development. Unlike genetic mutations, epigenetic aberrations are potentially reversible through epigenetic therapy, providing a therapeutically relevant treatment option. Histone methyltransferase inhibitors are emerging as an epigenetic therapy approach with great promise in the field of clinical oncology. The recent accelerated approval of the enhancer of zeste homolog 2 (EZH2; also known as histone-lysine N-methyltransferase EZH2) inhibitor tazemetostat for metastatic or locally advanced epithelioid sarcoma marks the first approval of such a compound for the treatment of cancer. Many other histone methyltransferase inhibitors are currently in development, some of which are being tested in clinical studies. This review focuses on histone methyltransferase inhibitors, highlighting their potential in the treatment of cancer. We also discuss the role for such epigenetic drugs in overcoming epigenetically driven drug resistance mechanisms, and their value in combination with other therapeutic approaches such as immunotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Proteína Potenciadora del Homólogo Zeste 2/efectos de los fármacos , Histona Metiltransferasas/metabolismo , Oncología Médica/normas , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Piridonas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Epigénesis Genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Guías de Práctica Clínica como Asunto
15.
Cancer Treat Rev ; 76: 1-9, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30913454

RESUMEN

Although recent advances in the treatment of castration-resistant prostate cancer (CRPC) have significantly improved patient outcomes, advanced prostate cancer is still associated with substantial morbidity and mortality, particularly in patients who develop resistance after multiple lines of therapy. Various cell signaling, DNA repair, and epigenetic enzymatic pathways are being targeted with small-molecule inhibitors in order to identify treatment strategies for patients with CRPC. In this review, we discuss novel targets and agents, studied preclinically and now being validated in clinical trials, including poly ADP-ribose polymerase (PARP), enhancer of zeste homologue 2 (EZH2), hedgehog pathway, MDM2/p53, and tyrosine kinase inhibitors. Further, we outline current approaches for novel prostate cancer vaccines such as DCVAC/PCa, PROSTVAC-V/F, MVI-816, CV9104, and PF-06753512. This wide spectrum of potential treatment strategies holds promise for additional improvements in the treatment of patients with CRPC, as these novel agents are aimed at targets known to be associated with growth and malignant progression of prostate cancer. If primary study endpoints are met, findings from ongoing phase III trials of well-tolerated and active combinations may provide new effective treatment options for advanced prostate cancer and thereby contribute to enhanced disease control in CRPC patients.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/terapia , Animales , Vacunas contra el Cáncer/administración & dosificación , Humanos , Masculino , Terapia Molecular Dirigida/métodos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Ensayos Clínicos Controlados Aleatorios como Asunto
16.
J Med Chem ; 61(3): 650-665, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29211475

RESUMEN

A new series of lactam-derived EZH2 inhibitors was designed via ligand-based and physicochemical-property-based strategies to address metabolic stability and thermodynamic solubility issues associated with previous lead compound 1. The new inhibitors incorporated an sp3 hybridized carbon atom at the 7-position of the lactam moiety present in lead compound 1 as a replacement for a dimethylisoxazole group. This transformation enabled optimization of the physicochemical properties and potency compared to compound 1. Analysis of relationships between calculated log D (clogD) values and in vitro metabolic stability and permeability parameters identified a clogD range that afforded an increased probability of achieving favorable ADME data in a single molecule. Compound 23a exhibited the best overlap of potency and pharmaceutical properties as well as robust tumor growth inhibition in vivo and was therefore advanced as a development candidate (PF-06821497). A crystal structure of 23a in complex with the three-protein PRC2 complex enabled understanding of the key structural features required for optimal binding.


Asunto(s)
Diseño de Fármacos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Isoquinolinas/farmacología , Isoquinolinas/farmacocinética , Administración Oral , Disponibilidad Biológica , Línea Celular Tumoral , Humanos , Isoquinolinas/administración & dosificación , Isoquinolinas/química , Modelos Moleculares , Conformación Molecular
17.
Comput Soc Netw ; 3(1): 7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-29355190

RESUMEN

BACKGROUND: A great variety of artificial and natural systems can be abstracted into a set of entities interacting with each other. Such abstractions can very well represent the underlying dynamics of the system when modeled as the network of vertices coupled by edges. Prediction of dynamics in these structures based on topological attribute or dependency relations is an important task. Link Prediction in such complex networks is regarded useful in almost all types of networks as it can be used to extract missing information, identify spurious interactions, and evaluate network evolving mechanisms. Various similarity and likelihood-based indices have been employed to infer different topological and relation-based information to form a link prediction algorithm. These algorithms, however, are too specific to the domain and do not encapsulate the generic nature of the real-world information. In most natural and engineered systems, the entities are linked with multiple types of associations and relations which play a factor in the dynamics of the network. It forms multiple subsystems or multiple layers of networked information. These networks are regarded as Multiplex Networks. METHODS: This work presents an approach for link prediction in Multiplex networks where the associations are learned from the multiple layers of networks for link prediction purposes. Most of the real-world networks are represented as weighted networks. Weight prediction coupled with Link Prediction can be of great use. Link scores are received using various similarity measures and used to predict weights. This work further proposes and testifies a strategy for weight prediction. RESULTS AND CONCLUSIONS: This work successfully proposes an algorithm for Weight Prediction using Link similarity measures on multiplex networks. The predicted weights show very less deviation from their actual weights. In comparison to other indices, the proposed method has a far low error rate and outperforms them concerning the metric performance NRMSE.

18.
Cancer Res ; 75(18): 3865-3878, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26139243

RESUMEN

The SWI/SNF multisubunit complex modulates chromatin structure through the activity of two mutually exclusive catalytic subunits, SMARCA2 and SMARCA4, which both contain a bromodomain and an ATPase domain. Using RNAi, cancer-specific vulnerabilities have been identified in SWI/SNF-mutant tumors, including SMARCA4-deficient lung cancer; however, the contribution of conserved, druggable protein domains to this anticancer phenotype is unknown. Here, we functionally deconstruct the SMARCA2/4 paralog dependence of cancer cells using bioinformatics, genetic, and pharmacologic tools. We evaluate a selective SMARCA2/4 bromodomain inhibitor (PFI-3) and characterize its activity in chromatin-binding and cell-functional assays focusing on cells with altered SWI/SNF complex (e.g., lung, synovial sarcoma, leukemia, and rhabdoid tumors). We demonstrate that PFI-3 is a potent, cell-permeable probe capable of displacing ectopically expressed, GFP-tagged SMARCA2-bromodomain from chromatin, yet contrary to target knockdown, the inhibitor fails to display an antiproliferative phenotype. Mechanistically, the lack of pharmacologic efficacy is reconciled by the failure of bromodomain inhibition to displace endogenous, full-length SMARCA2 from chromatin as determined by in situ cell extraction, chromatin immunoprecipitation, and target gene expression studies. Furthermore, using inducible RNAi and cDNA complementation (bromodomain- and ATPase-dead constructs), we unequivocally identify the ATPase domain, and not the bromodomain of SMARCA2, as the relevant therapeutic target with the catalytic activity suppressing defined transcriptional programs. Taken together, our complementary genetic and pharmacologic studies exemplify a general strategy for multidomain protein drug-target validation and in case of SMARCA2/4 highlight the potential for drugging the more challenging helicase/ATPase domain to deliver on the promise of synthetic-lethality therapy.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Proteínas Cromosómicas no Histona/deficiencia , ADN Helicasas/antagonistas & inhibidores , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Piridinas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/deficiencia , Unión Competitiva , Catálisis , Línea Celular Tumoral , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/genética , ADN Helicasas/química , ADN Helicasas/deficiencia , ADN Complementario/genética , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Humanos , Neoplasias Pulmonares/patología , Análisis por Micromatrices , Neoplasias/genética , Proteínas Nucleares/química , Proteínas Nucleares/deficiencia , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Tumor Rabdoide/genética , Tumor Rabdoide/patología , Sarcoma Sinovial/genética , Sarcoma Sinovial/patología , Factores de Transcripción/química , Factores de Transcripción/genética
19.
Epigenetics Chromatin ; 5(1): 3, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22284370

RESUMEN

BACKGROUND: DNA methylation, histone modifications and nucleosome occupancy act in concert for regulation of gene expression patterns in mammalian cells. Recently, G9a, a H3K9 methyltransferase, has been shown to play a role in establishment of DNA methylation at embryonic gene targets in ES cells through recruitment of de novo DNMT3A/3B enzymes. However, whether G9a plays a similar role in maintenance of DNA methylation in somatic cells is still unclear. RESULTS: Here we show that G9a is not essential for maintenance of DNA methylation in somatic cells. Knockdown of G9a has no measurable effect on DNA methylation levels at G9a-target loci. DNMT3A/3B remain stably anchored to nucleosomes containing methylated DNA even in the absence of G9a, ensuring faithful propagation of methylated states in cooperation with DNMT1 through somatic divisions. Moreover, G9a also associates with nucleosomes in a DNMT3A/3B and DNA methylation-independent manner. However, G9a knockdown synergizes with pharmacologic inhibition of DNMTs resulting in increased hypomethylation and inhibition of cell proliferation. CONCLUSIONS: Taken together, these data suggest that G9a is not involved in maintenance of DNA methylation in somatic cells but might play a role in re-initiation of de novo methylation after treatment with hypomethylating drugs, thus serving as a potential target for combinatorial treatments strategies involving DNMTs inhibitors.

20.
Cancer Cell ; 21(5): 655-667, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22624715

RESUMEN

Cancer cells typically exhibit aberrant DNA methylation patterns that can drive malignant transformation. Whether cancer cells are dependent on these abnormal epigenetic modifications remains elusive. We used experimental and bioinformatic approaches to unveil genomic regions that require DNA methylation for survival of cancer cells. First, we surveyed the residual DNA methylation profiles in cancer cells with highly impaired DNA methyltransferases. Then, we clustered these profiles according to their DNA methylation status in primary normal and tumor tissues. Finally, we used gene expression meta-analysis to identify regions that are dependent on DNA methylation-mediated gene silencing. We further showed experimentally that these genes must be silenced by DNA methylation for cancer cell survival, suggesting these are key epigenetic events associated with tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Metilación de ADN , Epigénesis Genética , Neoplasias/genética , Muerte Celular , Supervivencia Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Análisis por Conglomerados , Biología Computacional , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HCT116 , Humanos , Neoplasias/enzimología , Neoplasias/patología , Fenotipo , Interferencia de ARN , Reproducibilidad de los Resultados , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA