Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 41
Filtrer
2.
Cancers (Basel) ; 16(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39123362

RÉSUMÉ

HER2 (human epidermal growth factor receptor 2) is highly expressed in a variety of cancers, including breast, lung, gastric, and pancreatic cancers. Its amplification is linked to poor clinical outcomes. At the genetic level, HER2 is encoded by the ERBB2 gene (v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2), which is frequently mutated or amplified in cancers, thus spurring extensive research into HER2 modulation and inhibition as viable anti-cancer strategies. An impressive body of FDA-approved drugs, including anti-HER2 monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), and HER2-tyrosine kinase inhibitors (TKIs), have demonstrated success in enhancing overall survival (OS) and disease progression-free survival (PFS). Yet, drug resistance remains a persistent challenge and raises the risks of metastatic potential and tumor relapse. Research into alternative therapeutic options for HER2+ breast cancer therefore proves critical for adapting to this ever-evolving landscape. This review highlights current HER2-targeted therapies, discusses predictive biomarkers for drug resistance, and introduces promising emergent therapies-especially combination therapies-that are aimed at overcoming drug resistance in the context of HER2+ breast cancer.

3.
J Transl Med ; 22(1): 758, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138582

RÉSUMÉ

BACKGROUND: While melanomas commonly harbor losses of 9p21, on which CDKN2A resides, the presence of additional tumor suppressor elements at this locus is incompletely characterized. Here we assess the expression levels and functional role of microRNA-876-3p (miR-876), whose gene also maps to 9p21. METHODS: Expression of miR-876 was assessed in human tissues and cell lines using quantitative miRNA reverse transcriptase polymerase chain reaction (qRT-PCR). MIR876 copy number was determined in The Cancer Genome Atlas (TCGA) melanoma cohort. The consequences of regulation of miR-876 expression were assessed on melanoma cell colony formation, migration, invasion, apoptosis, cell cycle progression, and drug sensitivity in culture, and on in vivo tumor growth in a xenograft model. Genome-wide transcriptomic changes induced by miR-876 overexpression were determined using RNA sequencing (RNA-Seq). RESULTS: miR-876 expression was significantly decreased in primary melanoma samples when compared with nevi, and in human melanoma cell lines when compared with human melanocytes. Analysis of the TCGA cohort revealed deletions in MIR876 in > 50% of melanomas. miR-876 overexpression resulted in decreased melanoma cell colony formation, migration, and invasion, which was accompanied by cell cycle arrest and increased apoptosis. Intra-tumoral injections of miR-876 significantly suppressed melanoma growth in vivo. RNA-Seq analysis of miR-876-treated tumors revealed downregulation of several growth-promoting genes, along with upregulation of tumor suppressor genes, which was confirmed by qRT-PCR analysis. Computational analyses identified MAPK1 (or ERK2) as a possible target of miR-876 action. Overexpression of miR-876 significantly suppressed luciferase expression driven by the MAPK1/ERK2 3' UTR, and resulted in decreased ERK protein expression in melanoma cells. MAPK1/ERK2 cDNA overexpression rescued the effects of miR-876 on melanoma colony formation. miR-876 overexpression sensitized melanoma cells to treatment with the BRAF inhibitor vemurafenib. CONCLUSIONS: These studies identify miR-876 as a distinct tumor suppressor on 9p21 that is inactivated in melanoma and suggest miR-876 loss as an additional mechanism to activate ERK and the mitogen activated protein kinase (MAPK) pathway in melanoma. In addition, they suggest the therapeutic potential of combining miR-876 overexpression with BRAF inhibition as a rational therapeutic strategy for melanoma.


Sujet(s)
Chromosomes humains de la paire 9 , Régulation de l'expression des gènes tumoraux , Gènes suppresseurs de tumeur , Mélanome , microARN , Humains , Mélanome/génétique , Mélanome/anatomopathologie , microARN/génétique , microARN/métabolisme , Lignée cellulaire tumorale , Animaux , Chromosomes humains de la paire 9/génétique , Mouvement cellulaire/génétique , Apoptose/génétique , Extracellular Signal-Regulated MAP Kinases/métabolisme , Prolifération cellulaire/génétique , Séquence nucléotidique , Invasion tumorale
4.
Int J Mol Sci ; 25(10)2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38791398

RÉSUMÉ

Chimeric antigen receptor (CAR)-T cell immunotherapy represents a cutting-edge advancement in the landscape of cancer treatment. This innovative therapy has shown exceptional promise in targeting and eradicating malignant tumors, specifically leukemias and lymphomas. However, despite its groundbreaking successes, (CAR)-T cell therapy is not without its challenges. These challenges, particularly pronounced in the treatment of solid tumors, include but are not limited to, the selection of appropriate tumor antigens, managing therapy-related toxicity, overcoming T-cell exhaustion, and addressing the substantial financial costs associated with treatment. Nanomedicine, an interdisciplinary field that merges nanotechnology with medical science, offers novel strategies that could potentially address these limitations. Its application in cancer treatment has already led to significant advancements, including improved specificity in drug targeting, advancements in cancer diagnostics, enhanced imaging techniques, and strategies for long-term cancer prevention. The integration of nanomedicine with (CAR)-T cell therapy could revolutionize the treatment landscape by enhancing the delivery of genes in (CAR)-T cell engineering, reducing systemic toxicity, and alleviating the immunosuppressive effects within the tumor microenvironment. This review aims to explore how far (CAR)-T cell immunotherapy has come alone, and how nanomedicine could strengthen it into the future. Additionally, the review will examine strategies to limit the off-target effects and systemic toxicity associated with (CAR)-T cell therapy, potentially enhancing patient tolerance and treatment outcomes.


Sujet(s)
Immunothérapie adoptive , Tumeurs , Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/immunologie , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Nanotechnologie/méthodes , Nanomédecine/méthodes , Animaux , Microenvironnement tumoral/immunologie , Lymphocytes T/immunologie , Antigènes néoplasiques/immunologie
5.
Eur J Pharmacol ; 970: 176508, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38493913

RÉSUMÉ

Necroptosis is a pivotal contributor to the pathogenesis of various human diseases, including those affecting the nervous system, cardiovascular system, pulmonary system, and kidneys. Extensive investigations have elucidated the mechanisms and physiological ramifications of necroptosis. Among these, protein phosphorylation emerges as a paramount regulatory process, facilitating the activation or inhibition of specific proteins through the addition of phosphate groups to their corresponding amino acid residues. Currently, the targeting of kinases has gained recognition as a firmly established and efficacious therapeutic approach for diverse diseases, notably cancer. In this comprehensive review, we elucidate the intricate role of phosphorylation in governing key molecular players in the necroptotic pathway. Moreover, we provide an in-depth analysis of recent advancements in the development of kinase inhibitors aimed at modulating necroptosis. Lastly, we deliberate on the prospects and challenges associated with the utilization of kinase inhibitors to modulate necroptotic processes.


Sujet(s)
Tumeurs , Protein kinases , Humains , Phosphorylation , Protein kinases/métabolisme , Nécroptose , Tumeurs/traitement médicamenteux , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Apoptose
8.
Biomedicines ; 11(10)2023 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-37893069

RÉSUMÉ

This review highlights Receptor Expressed in Lymphoid Tissues (RELT), a Tumor Necrosis Factor Superfamily member, and its two paralogs, RELL1 and RELL2. Collectively, these three proteins are referred to as RELTfms and have gained much interest in recent years due to their association with cancer and other human diseases. A thorough knowledge of their physiological functions, including the ligand for RELT, is lacking, yet emerging evidence implicates RELTfms in a variety of processes including cytokine signaling and pathways that either promote cell death or survival. T cells from mice lacking RELT exhibit increased responses against tumors and increased inflammatory cytokine production, and multiple lines of evidence indicate that RELT may promote an immunosuppressive environment for tumors. The relationship of individual RELTfms in different cancers is not universal however, as evidence indicates that individual RELTfms may be risk factors in certain cancers yet appear to be protective in other cancers. RELTfms are important for a variety of additional processes related to human health including microbial pathogenesis, inflammation, behavior, reproduction, and development. All three proteins have been strongly conserved in all vertebrates, and this review aims to provide a clearer understanding of the current knowledge regarding these interesting proteins.

9.
Int J Mol Sci ; 25(1)2023 Dec 19.
Article de Anglais | MEDLINE | ID: mdl-38203175

RÉSUMÉ

The farnesoid-X receptor (FXR), a member of the nuclear hormone receptor superfamily, can be activated by bile acids (BAs). BAs binding to FXR activates BA signaling which is important for maintaining BA homeostasis. FXR is differentially expressed in human organs and exists in immune cells. The dysregulation of FXR is associated with a wide range of diseases including metabolic disorders, inflammatory diseases, immune disorders, and malignant neoplasm. Recent studies have demonstrated that FXR influences tumor cell progression and development through regulating oncogenic and tumor-suppressive pathways, and, moreover, it affects the tumor microenvironment (TME) by modulating TME components. These characteristics provide a new perspective on the FXR-targeted therapeutic strategy in cancer. In this review, we have summarized the recent research data on the functions of FXR in solid tumors and its influence on the TME, and discussed the mechanisms underlying the distinct function of FXR in various types of tumors. Additionally, the impacts on the TME by other BA receptors such as takeda G protein-coupled receptor 5 (TGR5), sphingosine-1-phosphate receptor 2 (S1PR2), and muscarinic receptors (CHRM2 and CHRM3), have been depicted. Finally, the effects of FXR agonists/antagonists in a combination therapy with PD1/PD-L1 immune checkpoint inhibitors and other anti-cancer drugs have been addressed.


Sujet(s)
Tumeurs , Humains , Tumeurs/traitement médicamenteux , Association thérapeutique , Acides et sels biliaires , Homéostasie , Inhibiteurs de points de contrôle immunitaires , Microenvironnement tumoral , Récepteur muscarinique de type M3
10.
PLoS One ; 17(5): e0269135, 2022.
Article de Anglais | MEDLINE | ID: mdl-35622821

RÉSUMÉ

Early detection of breast cancer plays a critical role in successful treatment that saves thousands of lives of patients every year. Despite massive clinical data have been collected and stored by healthcare organizations, only a small portion of the data has been used to support decision-making for treatments. In this study, we proposed an engineered up-sampling method (ENUS) for handling imbalanced data to improve predictive performance of machine learning models. Our experiment results showed that when the ratio of the minority to the majority class is less than 20%, training models with ENUS improved the balanced accuracy 3.74%, sensitivity 8.36% and F1 score 3.83%. Our study also identified that XGBoost Tree (XGBTree) using ENUS achieved the best performance with an average balanced accuracy of 97.47% (min = 93%, max = 100%), sensitivity of 97.88% (min = 89% and max = 100%), and F1 score of 96.20% (min = 89.5%, max = 100%) in the validation dataset. Furthermore, our ensemble algorithm identified Cell_Shape and Nuclei as the most important attributes in predicting breast cancer. The finding re-affirms the previous knowledge of the relationship between Cell_Shape, Nuclei, and the grades of breast cancer using a data-driven approach. Finally, our experiment showed that Random Forest and Neural Network models had the least training time. Our study provided a comprehensive comparison of a wide range of machine learning methods in predicting breast cancer risk. It can be used as a tool for healthcare practitioners to effectively detect and treat breast cancer.


Sujet(s)
Tumeurs du sein , Algorithmes , Tumeurs du sein/diagnostic , Femelle , Humains , Apprentissage machine ,
11.
Cancers (Basel) ; 14(6)2022 Mar 09.
Article de Anglais | MEDLINE | ID: mdl-35326556

RÉSUMÉ

Chimeric antigen receptors (CAR) T cells are T cells engineered to express membrane receptors with high specificity to recognize specific target antigens presented by cancer cells and are co-stimulated with intracellular signals to increase the T cell response. CAR-T cell therapy is emerging as a novel therapeutic approach to improve T cell specificity that will lead to advances in precision medicine. CAR-T cells have had impressive outcomes in hematological malignancies. However, there continue to be significant limitations of these therapeutic responses in targeting solid malignancies such as heterogeneous antigens in solid tumors, tumor immunosuppressive microenvironment, risk of on-target/off-tumor, infiltrating CAR-T cells, immunosuppressive checkpoint molecules, and cytokines. This review paper summarizes recent approaches and innovations through combination therapies of CAR-T cells and other immunotherapy or small molecule drugs to counter the above disadvantages to potentiate the activity of CAR-T cells.

12.
Int J Mol Sci ; 23(4)2022 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-35216405

RÉSUMÉ

In the United States, breast cancer is among the most frequently diagnosed cancers in women. Breast cancer is classified into four major subtypes: human epidermal growth factor receptor 2 (HER2), Luminal-A, Luminal-B, and Basal-like or triple-negative, based on histopathological criteria including the expression of hormone receptors (estrogen receptor and/or progesterone receptor) and/or HER2. Primary breast cancer treatments can include surgery, radiation therapy, systemic chemotherapy, endocrine therapy, and/or targeted therapy. Endocrine therapy has been shown to be effective in hormone receptor-positive breast cancers and is a common choice for adjuvant therapy. However, due to the aggressive nature of triple-negative breast cancer, targeted therapy is becoming a noteworthy area of research in the search for non-endocrine-targets in breast cancer. In addition to HER2-targeted therapy, other emerging therapies include immunotherapy and targeted therapy against critical checkpoints and/or pathways in cell growth. This review summarizes novel targeted breast cancer treatments and explores the possible implications of combination therapy.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Immunothérapie/méthodes , Thérapie moléculaire ciblée/méthodes , Récepteurs aux stéroïdes/métabolisme
13.
Int J Mol Sci ; 22(11)2021 May 25.
Article de Anglais | MEDLINE | ID: mdl-34070674

RÉSUMÉ

BRCA1 and BRCA2 are tumor suppressor genes with pivotal roles in the development of breast and ovarian cancers. These genes are essential for DNA double-strand break repair via homologous recombination (HR), which is a virtually error-free DNA repair mechanism. Following BRCA1 or BRCA2 mutations, HR is compromised, forcing cells to adopt alternative error-prone repair pathways that often result in tumorigenesis. Synthetic lethality refers to cell death caused by simultaneous perturbations of two genes while change of any one of them alone is nonlethal. Therefore, synthetic lethality can be instrumental in identifying new therapeutic targets for BRCA1/2 mutations. PARP is an established synthetic lethal partner of the BRCA genes. Its role is imperative in the single-strand break DNA repair system. Recently, Olaparib (a PARP inhibitor) was approved for treatment of BRCA1/2 breast and ovarian cancer as the first successful synthetic lethality-based therapy, showing considerable success in the development of effective targeted cancer therapeutics. Nevertheless, the possibility of drug resistance to targeted cancer therapy based on synthetic lethality necessitates the development of additional therapeutic options. This literature review addresses cancer predisposition genes, including BRCA1, BRCA2, and PALB2, synthetic lethality in the context of DNA repair machinery, as well as available treatment options.


Sujet(s)
Protéine BRCA1/génétique , Protéine BRCA2/génétique , Tumeurs du sein/génétique , Protéine du groupe de complémentation N de l'anémie de Fanconi/génétique , Prédisposition génétique à une maladie , Mutations synthétiques létales , Tumeurs du sein/thérapie , Réparation de l'ADN , Femelle , Humains
14.
J Cancer ; 12(4): 1212-1219, 2021.
Article de Anglais | MEDLINE | ID: mdl-33442419

RÉSUMÉ

Chimeric antigen receptor (CAR) T-cell therapy is an innovative form of immunotherapy wherein autologous T-cells are genetically modified to express chimeric receptors encoding an antigen-specific single-chain variable fragment and costimulatory molecules. Moreover, CAR T-cell therapy can only work successfully in patients who have an intact immune system. Therefore, patients receiving cytotoxic chemotherapy will be immunosuppressed making CAR-T therapy less effective. In adoptive CD8+ T-cell therapy (ACT), numerous tumor-specific, engineered T-cells are sourced from patients, expanded in vitro, and infused back expressing tumor-specific antigen receptors. The most successful ACT, anti-CD19 chimeric antigen receptor T-cell therapy directed against B-cell lymphoma, has proved to be efficacious. However, current efforts to utilize this approach for solid tumors, like breast cancer, have shown only modest improvement. Nevertheless, the potential efficacy of CAR-T therapy is promising in an era of immunological advances. By appropriately manipulating CAR T-cells to combat the immunosuppressive forces of the tumor microenvironment, significant eradication of the solid tumor may occur. This review discusses CAR T-cell therapy and its specificity and safety in adoptive cell transfers in breast cancer. We will highlight novel discoveries in CAR T-cell immunotherapy and the formidable barriers including suppression of T-cell function and localization at tumor sites.

15.
Biochem Biophys Rep ; 24: 100868, 2020 Dec.
Article de Anglais | MEDLINE | ID: mdl-33367115

RÉSUMÉ

Receptor Expressed in Lymphoid Tissues (RELT) is a human tumor necrosis factor receptor superfamily member (TNFRSF) that is expressed most prominently in cells and tissues of the hematopoietic system. RELL1 and RELL2 are two homologs that physically interact with RELT and co-localize with RELT at the plasma membrane. This study sought to further elucidate the function of RELT by identifying novel protein interactions with RELT family members. The transcription factor MyoD family inhibitor domain-containing (MDFIC) was identified in a yeast two-hybrid genetic screen using RELL1 as bait. MDFIC co-localizes with RELT family members at the plasma membrane; this co-localization was most prominently observed with RELL1 and RELL2. In vitro co-immunoprecipitation (Co-IP) was utilized to demonstrate that MDFIC physically interacts with RELT, RELL1, and RELL2. Co-IP using deletion mutants of MDFIC and RELT identified regions important for physical association between MDFIC and RELT family members and a computational analysis revealed that RELT family members are highly disordered proteins. Immunohistochemistry of normal human lymph nodes revealed RELT staining that was most prominent in macrophages. Interestingly, the level of RELT staining significantly increased progressively in low and high-grade B-cell lymphomas versus normal lymph nodes. RELT co-staining with CD20 was observed in B-cell lymphomas, indicating that RELT is expressed in malignant B cells. Collectively, these results further our understanding of RELT-associated signaling pathways, the protein structure of RELT family members, and provide preliminary evidence indicating an association of RELT with B-cell lymphomas.

16.
PLoS One ; 15(5): e0233672, 2020.
Article de Anglais | MEDLINE | ID: mdl-32469945

RÉSUMÉ

Agents that modulate pre-mRNA splicing are of interest in multiple therapeutic areas, including cancer. We report our recent screening results with the application of a cell-based Triple Exon Skipping Luciferase Reporter (TESLR) using a library that is composed of FDA approved drugs, clinical compounds, and mechanistically characterized tool compounds. Confirmatory assays showed that three clinical antitumor therapeutic candidates (milciclib, PF-3758309 and PF-562271) are potent splicing modulators and that these drugs are, in fact, nanomolar inhibitors of multiple kinases involved in the regulation the spliceosome. We also report the identification of new SF3B1 antagonists (sudemycinol C and E) and show that these antagonists can be used to develop a displacement assay for SF3B1 small molecule ligands. These results further support the broad potential for the development of agents that target the spliceosome for the treatment of cancer and other diseases, as well as new avenues for the discovery of new chemotherapeutic agents for a range of diseases.


Sujet(s)
Antinéoplasiques/pharmacologie , Tests de criblage d'agents antitumoraux/méthodes , Exons/effets des médicaments et des substances chimiques , Précurseurs des ARN/génétique , Épissage des ARN/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Humains , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Inhibiteurs de protéines kinases/pharmacologie
17.
J Chromatogr A ; 1577: 47-58, 2018 Nov 23.
Article de Anglais | MEDLINE | ID: mdl-30316615

RÉSUMÉ

This study combines a high-performance liquid chromatography-fluorescent detection method (HPLC-FLD) with in-situ cell imaging for the sensitive analysis of glutathione (GSH), cysteine (Cys) and homocysteine (Hcys), using BODIPY®507/545 IA as a labeling reagent. The analytical potential of BODIPY®507/545 IA in cell imaging was deeply explored, concerning fluorescent response, selectivity, cell-permeability, biotoxicity and so on. It is demonstrated that BODIPY®507/545 IA has good biocompatibility and the fluorescence intensity is enhanced remarkably after reacting with thiols. The best derivative condition was obtained in boric acid buffer (0.05 mmol/L, pH 9.5) at 45 °C for 15 min. For chromatographic method, two sensitive methods, HPLC-FLD and capillary electrophoresis-laser-induced fluorescence detection (CE-LIF) were both developed, validated, and compared. The detection limits for the thiols ranged from 5 to 10 nmol/L with HPLC-FLD and 0.5 nmol/L for the CE-LIF method. Finally, HPLC-FLD is adopted to quantify the thiols in HepG2 cell samples after cell imaging.


Sujet(s)
Chromatographie en phase liquide à haute performance , Homocystéine/analyse , Imagerie optique , Thiols/analyse , Composés du bore/composition chimique , Cystéine/analyse , Électrophorèse capillaire , Fluorescence , Colorants fluorescents/composition chimique , Glutathion/analyse , Cellules HepG2 , Humains , Limite de détection
19.
Bioorg Med Chem Lett ; 27(3): 406-412, 2017 02 01.
Article de Anglais | MEDLINE | ID: mdl-28049589

RÉSUMÉ

The splicing of pre-mRNA is a critical process in normal cells and is deregulated in cancer. Compounds that modulate this process have recently been shown to target a specific vulnerability in tumors. We have developed a novel cell-based assay that specifically activates luciferase in cells exposed to SF3B1 targeted compounds, such as sudemycin D6. This assay was used to screen a combined collection of approved drugs and bioactive compounds. This screening approach identified several active hits, the most potent of which were CGP-74514A and aminopurvalanol A, both have been reported to be cyclin-dependent kinases (CDKs) inhibitors. We found that these compounds, and their analogs, show significant cdc2-like kinase (CLK) inhibition and clear structure-activity relationships (SAR) at CLKs. We prepared a set of analogs and were able to 'dial out' the CDK activity and simultaneously developed CLK inhibitors with low nanomolar activity. Thus, we have demonstrated the utility of our exon-skipping assay and identified new molecules that exhibit potency and selectivity for CLK, as well as some structurally related dual CLK/CDK inhibitors.


Sujet(s)
Kinases cyclines-dépendantes/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/composition chimique , Amino-2 purine/analogues et dérivés , Amino-2 purine/composition chimique , Amino-2 purine/métabolisme , Adénine/analogues et dérivés , Adénine/composition chimique , Adénine/métabolisme , Sites de fixation , Kinases cyclines-dépendantes/génétique , Kinases cyclines-dépendantes/métabolisme , Exons , Gènes rapporteurs , Tests de criblage à haut débit , Humains , Concentration inhibitrice 50 , Luciferases/génétique , Simulation de dynamique moléculaire , Liaison aux protéines , Inhibiteurs de protéines kinases/métabolisme , Structure tertiaire des protéines , Épissage des ARN , Relation structure-activité
20.
J Med Chem ; 59(24): 11161-11170, 2016 12 22.
Article de Anglais | MEDLINE | ID: mdl-27936709

RÉSUMÉ

Here we describe a new approach for tumor targeting in which augmented concentrations of Fe(II) in cancer cells and/or the tumor microenvironment triggers drug release from an Fe(II)-reactive prodrug conjugate. The 1,2,4-trioxolane scaffold developed to enable this approach can in principle be applied to a broad range of cancer therapeutics and is illustrated here with Fe(II)-targeted forms of a microtubule toxin and a duocarmycin-class DNA-alkylating agent. We show that the intrinsic reactivity/toxicity of the duocarmycin analog is masked in the conjugated form and this greatly reduced toxicity in mice. This in turn permitted elevated dosing levels, leading to higher systemic exposure and a significantly improved response in tumor xenograft models. Overall our results suggest that Fe(II)-dependent drug delivery via trioxolane conjugates could have significant utility in expanding the therapeutic index of a range of clinical and preclinical stage cancer chemotherapeutics.


Sujet(s)
Antinéoplasiques/pharmacologie , Composés du fer II/pharmacologie , Indoles/pharmacologie , Promédicaments/pharmacologie , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Systèmes de délivrance de médicaments , Tests de criblage d'agents antitumoraux , Duocarmycines , Femelle , Composés du fer II/composition chimique , Humains , Indoles/synthèse chimique , Indoles/composition chimique , Tumeurs expérimentales de la mamelle/traitement médicamenteux , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris de lignée NOD , Souris nude , Souris SCID , Structure moléculaire , Promédicaments/synthèse chimique , Promédicaments/composition chimique , Pyrroles/synthèse chimique , Pyrroles/composition chimique , Pyrroles/pharmacologie , Relation structure-activité , Cellules cancéreuses en culture
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE