Connectivity Mapping of BCL6 Targeted Therapy Guides Rational Design of Potent and Specific Non-Chemotherapy Combinatorial Regimens in DLBCL.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 523-523
Author(s):  
Leandro Cerchietti ◽  
Maria E. Figueroa ◽  
David Meyers ◽  
Philip A. Cole ◽  
Kapil Bhalla ◽  
...  

Abstract DLBCL is the most common form of non-Hodgkin’s lymphoma. Combinations of untargeted chemotherapeutic agents cure between 40–60% of DLBCL patients. We are interested in the rational design of targeted combinatorial therapy for DLBCL using non-chemotherapy agents. Towards this goal we developed an inhibitor of the BCL6 transcriptional repressor, the most commonly involved oncogene in DLBCL. This BCL6 peptide inhibitor (BPI) causes de-repression of BCL6 target genes and kills DLBCL cells. Since single agent targeted therapy is unlikely to cure tumors, we hypothesized that identification of survival pathways triggered by BPI would facilitate rational design of combinatorial biological therapy for DLBCL. In order to identify such pathways we performed gene expression (GE) microarray studies in ten DLCBL cell lines treated with BPI vs. control. Six cell lines were BCL6 positive and four were BCL6 negative. Only the BCL6 positive cells yielded differences in gene expression. Among BPI induced genes was the p300 histone acetyl-transferase. The overlapping genes among the six cell lines were used to generate a BPI response signature. We used this signature to query the Broad Institute Connectivity Map, which contains the GE signature of 164 distinct small-molecule perturbagens. The top scoring classes of drugs were the histone deacetylase inhibitors (HDIs) and HSP90 inhibitors. Considering that BPI is chemically un-related to HDIs or HSP90 inhibitors and that BPI induces p300, we hypothesized that a major biological effect of BPI is to cause the acetylation of HSP90 (which inhibits Hsp90 pro-survival activity) and p53, (which enhances its pro-apoptotic activity). We verified that p300 is a direct BCL6 target gene by ChIP assays, that BPI induces p300 mRNA and protein by QPCR and western blot, and that p300 is silenced in most primary DLBCLs at both the mRNA and protein levels. Accordingly, BPI induced acetylation of Hsp90 and inhibited its function, as demonstrated by the decrease in the abundance of Hsp90 client proteins (AKT/PKB and c-raf). BPI also induced acetylation and functional activity of p53 in a p300-dependent manner (and also induces p53 expression). The importance of p300 was confirmed since a p300-dominant negative construct and the specific p300(HAT) inhibitor Lys-CoA-TAT could block BPI antilymphoma activity. Remarkably, we observed a dose-sequence dependent synergistic effect of BPI followed by Hsp90 inhibitors in killing DLBCL cells. Hsp90 is a relevant target in DLBCL since HSP90?/? protein was expressed in ∼90% of DLBCL patients (n=70). HDIs also increase acetylation of Hsp90 and p53. The HDI drugs SAHA, valproic acid and TSA all profoundly synergized with BPI to specifically eradicate BCL6 positive DLBCL cell lines. In conclusion, we discovered an unexpected mechanistic link between BCL6 and suppression of protein acetylation in lymphomagenesis. This information was harnessed for the rational design of synergistic targeted therapy with BCL6 inhibitors followed by Hsp90 or HDAC inhibitors to target cellular pathways induced by BPI. We anticipate that these drug combinations will result in more potent and less toxic therapeutic treatment of DLBCL, possibly with less or no added chemotherapy.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4503-4503
Author(s):  
Jingda Xu ◽  
R Eric Davis ◽  
Zhiqiang Wang ◽  
Jason R. Westin

Abstract Introduction: XPO1 (CRM1, Exportin 1) is the sole transporter of many tumor suppressor proteins (including MYC, BCL2, BCL6, BTK, IkB) and is elevated in non-Hodgkin Lymphoma. Selinexor (Sel, KPT-330) is an oral covalent inhibitor of XPO1, the first clinical molecule of the Selective Inhibitors of Nuclear Export drug class. The phase I clinical trial of Sel in hematologic malignancies showed promising early single-agent efficacy with modest toxicity in relapsed Diffuse Large B-cell Lymphoma (DLBCL, Gutierrez et al, ASCO 2014). DLBCL, the most common lymphoid malignancy, is currently cured in only 10% of relapsed patients, and consists of 2 subtypes based on putative cell of origin (COO): activated B-cell (ABC) and germinal center B-cell (GCB). We performed preclinical studies of Sel, modeling its single-agent efficacy in frontline and relapsed DLBCL and its potential synergy with other clinically relevant therapeutics. Methods: To model drug resistant DLBCL, resistant subpopulations of 12 patient-derived DLBCL cell lines were created by in vitro intermittent exposure to active congeners of cyclophosphamide, doxorubicin, and vincristine (ivCHOP), approximating clinical practice. To determine if CHOP-resistant DLBCL is also resistant to other agents, we determined single-agent dose response curves and IC50 values for both parental and ivCHOP resistant (CHOP-res) subclones of 4 of these lines at submission (HBL1 & TMD8 of ABC subtype, OCI-Ly7 & HT of GCB subtype, with 8 lines in progress) with Sel, chemotherapy (CT, ivCHOP, DHAP, and ICE), and targeted therapy (TT, ibrutinib, ABT-199, idelalisib, everolimus, MLN0128, alisertib, lenalidomide, bortezomib, I-BET151, and ONC201). Viability was assessed with CellTiter-Glo (Promega) after a 3 day cell culture. IC50 values were determined using GraphPad Prism. Based on these results, we evaluated the ability of Sel to synergize with other agents or restore sensitivity in CHOP-res with a combination “checkerboard” (orthogonal dose titration for each drug). The Combination Index (CI) for pairs at all concentrations was calculated with ComboSyn, with CI values <1 indicating synergy. Gene expression profiling with Illumina HT12v4 arrays will compare parental and CHOP-res of 12 DLBCL lines. Results: All CHOP-res lines of both COO types had higher IC50 for both ivCHOP (mean, 3.7x) and DHAP (4.5x) as compared to parental cells (Table 1). In contrast, the IC50 of Sel is unchanged between parental and CHOP-res, for both COO types. Other targeted agents displayed variable activity between parental and CHOP-res and between COO types, with the IC50 of ibrutinib being nearly 2 log lower in ABC lines. CI values showed that Sel was generally a strong synergizer (Table 1), especially with TT and in ABC lines. Sel had lower CI values with CT, but restored sensitivity to ivCHOP in HBL1 (Figure 1). Bortezomib and Sel were moderately antagonistic, although further tests are ongoing. Gene expression profiling, comparing parental vs. CHOP-res and Sel synergizing vs. non-synergizing lines, is ongoing. Conclusions: Our data suggest that Sel: 1) is equally active, and thus not cross-resistant, in cell lines made resistant to standard chemotherapeutics, 2) is a potent, broadly active synergizer with targeted therapy against lines modeling relapsed DLBCL, and 3) has greater synergy in ABC DLBCL, in which it may be able to reverse acquired resistance to frontline therapy. This behavior fits with the broad effects of XPO1 inhibition. The cross-resistance of CHOP-res lines to DHAP models clinical outcomes, and re-sensitization of CHOP-res lines with Sel suggests the potential for relapsed and frontline clinical trials. Further work with our model may discover more synergies of Sel, suggesting future clinical combinations and biomarkers associated with response. Table 1HBL1TMD8OCI-Ly7HTABCGCBIC50SR ΔSR ΔSR ΔSR ΔivCHOP2E-62.31E-75.51E-63.78E-63.2DHAP6E-73.65E-85.52E-73.71E-75.2Selinexor5E-80.66E-81.57E-80.94E-71.3Ibrutinib8E-81.02E-70.43E-60.92E-61.0ABT-1992E-60.53E-61.23E-60.38E-940.3Bortezomib4E-100.61E-101.03E-102.84E-100.9MLN01282E-71.22E-89.44E-85.42E-73.4CI with selinexorivCHOP 0.27 0.27 1.26 3.24DHAP 0.65 0.65 2.23 0.49Ibrutinib 0.06 0.06 0.02 0.95ABT-199 0.47 0.47 0.26 0.89Bortezomib 3.23 3.23 3.53 10Dexamethasone 0.19 0.19 0.39 2.09MLN0128 0.11 0.35 0.47 0.09 ivCHOP sensitive=S, Resistant=R, Δ fold change from S to R Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5695-5695
Author(s):  
Amit Kumar Mitra ◽  
Sanjoy Dey ◽  
Andrew Hangsleben ◽  
Michael Steinbach ◽  
Vipin Kumar ◽  
...  

Abstract Multiple myeloma (MM) is the second-most common hematopoietic malignancy in the United States accounting for 1% of all cancers and 10% of all hematologic malignancies. Despite recent improvements in treatment strategies including the emergence of proteasome inhibitors (PIs) as effective chemotherapeutic agents, MM still remains difficult to cure with median survival rate of around 7 years, primarily due to wide inter-individual variation in response to treatment. We believe such heterogeneity in response to PIs is governed by the underlying molecular characteristics of the tumor including alterations in gene expression profile (GEP). In the current study, we used a panel of Human Myeloma Cell Lines (HMCLs) representing the gamut of biological and genetic heterogeneity in MM to evaluate the gene expression signatures associated with response to the second-generation PI Ixazomib and produced a predictive score (PI score) for Ix response. HMCLs (n=45) were treated with increasing concentrations of Ixazomib used as single agent and half maximal inhibitory concentration (IC50) values were determined using cell viability equation. Gene expression profiling data was obtained as publicly available data from the Keats lab website at TGen (http://www.keatslab.org/myeloma-cell-lines). Genes with high expression value and high standard deviation beyond the median values were pre-filtered and log expression values were normalized by subtracting mean expression of individual genes across all the samples and the housekeeping genes (GAPDH). Subsequently, analysis of correlation between Ix IC50 data and GEP data and the False Discovery Rate (FDR) based on 1000 random permutations were performed to identify true patterns of genes that are highly predictive of Ix response and to look for the top genes that could discriminate between the top sensitive and top resistant cell lines. Gene clusters were identified that correlated with response and will be presented. Our results will demonstrate in vitro modeling of response using GEP approaches that may provide predictive scoring algorithms of a defined set of genes that will be useful in clinical evaluation of drug choice in treating individual patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3689-3689
Author(s):  
James M Bogenberger ◽  
Nanna Hansen ◽  
Devora Delman ◽  
Ruben A. Mesa ◽  
Raoul Tibes

Abstract Background: Histone deacetylase inhibitors (HDIs) have shown clinical activity in myeloid malignancies, albeit insufficient to justify single-agent therapy. Thus, we have sought targets/therapies that synergistically enhance the activity of HDIs for combination therapy. We conducted RNA-interference (RNAi) drug modifier screens with the HDI SAHA, and have previously reported convergence of multiple screen hits on different points of the p38-SAPK/JNK signaling pathway. However, in subsequent pharmacological studies, well-characterized p38α/β inhibitors SB2012190 and LY2228820, and JNK inhibitor SP600125 did not modulate SAHA anti-leukemic activity. Nonetheless, a putative MK2 substrate-selective p38 inhibitor known as CMPD1 (Biochemistry 2004 Sep 21; 43(37):11658-71) was found to potently synergize with SAHA in all AML cell lines tested (N=8) in a dose-dependent manner. Further, using ex vivo cultures of primary myeloid malignancies (N=14), CMPD1 showed strong, selective synergy with SAHA in malignant CD34+ isolated cells, as compared to little or no synergy in CD34-depleted cells. Furthermore, CMPD1 specifically synergized with SAHA and a similar HDI panobinostat, but was found to interact antagonistically with cytarabine, and only additively with azacitidine. Herein, we present additional data from our investigation to better understand the mechanism of CMPD1 synergy with HDIs for future clinical translation. Results: Concurrent cell cycle and apoptosis measurements by flow cytometry show that single-agent CMPD1 results in a potent G2/M arrest that is resolved over time without a significant induction of apoptosis, whereas the combination of SAHA + CMPD1 results in a similar or increased level of G2/M arrest, which conversely culminates in a significant induction of apoptosis. The synergistic dose of single-agent SAHA used in these flow cytometry studies did not cause any significant cell cycle changes, and did not induce significant apoptosis alone in the AML cell lines studied. CMPD1 is known to be a MK2 substrate-selective p38 inhibitor; however, we find that CMPD1 potently increases the phosphorylation of MK2 at doses strongly synergistic with SAHA. This is not a universal activity of p38 inhibitors in our in vitro AML models, as the p38 inhibitor SB202190 does not increase MK2 phosphorylation alone or in combination with SAHA. We further hypothesized that CMPD1 synergizes with SAHA through a transcription-based mechanism, thus we measured mRNA expression by next-generation sequencing after SAHA and CMPD1 treatment alone, and in combination, in an AML cell line TF-1. We find that SAHA + CMPD1 treatment induces a synergistic increase in transcript levels of: i) several chemokines such as IL-8, CXCL1, CXCL2, and CXCL3, ii) inhibitor of DNA binding proteins ID2 and ID3, and iii) cell cycle regulator p21. Transcript levels of the Hippo pathway transcription factor YAP1 were also found to be increased by CMPD1 treatment in TF-1. We confirm an increase of total YAP1 protein levels by western blot. We also find an increase of YAP1 phosphorylation at serine 127 in CMPD1 or CMPD1 + SAHA treated samples in several AML cell lines, yet we find that Ser-127 phosphorylated YAP1 is located in the nucleus. Consistent with YAP1 Ser-127 phosphorylation, we find that upstream signaling kinase LATS1 is activated/phosphorylated at serine 909 in AML cell lines after treatment with CMPD1 or SAHA + CMPD1. We also confirm a synergistic increase in p21 protein levels upon SAHA + CMPD1 treatment by western blot in several AML cell lines. Changes in the protein levels of ID2 and ID3 were confirmed in TF-1 by western blot; however, ID protein levels appear to be uniquely affected by CMPD1 or the combination in a cell line-dependent manner. Conclusion: CMPD1 selectively synergizes with histone deacetylases inhibitors in myeloid malignancies. The synergistic activity of this combination highlights the potential for selectively targeting malignant CD34+ cells. The phosphorylation of LATS1 observed is consistent with i) the known roles for LATS1 in cell cycle regulation, ii) the G2/M arrest observed with CMPD1, and iii) the downstream phosphorylation of YAP1 observed. These findings linking CMPD1 with LATS1/YAP1 await further study and characterization. Although CMPD1 is not a clinically-relevant compound, CMPD1 may be used as a scaffold or tool compound to enable future development and research. Disclosures Mesa: Pfizer: Research Funding; Novartis Pharmaceuticals Corporation: Consultancy; Genentech: Research Funding; NS Pharma: Research Funding; Promedior: Research Funding; Gilead: Research Funding; CTI Biopharma: Research Funding; Incyte Corporation: Research Funding.


Animals ◽  
2021 ◽  
Vol 11 (2) ◽  
pp. 502
Author(s):  
Filipe Almeida ◽  
Andreia Gameiro ◽  
Jorge Correia ◽  
Fernando Ferreira

Feline mammary carcinoma (FMC) is the third most common type of neoplasia in cats, sharing similar epidemiological features with human breast cancer. In humans, histone deacetylases (HDACs) play an important role in the regulation of gene expression, with HDAC inhibitors (HDACis) disrupting gene expression and leading to cell death. In parallel, microtubules inhibitors (MTIs) interfere with the polymerization of microtubules, leading to cell cycle arrest and apoptosis. Although HDACis and MTIs are used in human cancer patients, in cats, data is scarce. In this study, we evaluated the antitumor properties of six HDACis (CI-994, panobinostat, SAHA, SBHA, scriptaid, and trichostatin A) and four MTIs (colchicine, nocodazole, paclitaxel, and vinblastine) using three FMC cell lines (CAT-MT, FMCp, and FMCm), and compared with the human breast cancer cell line (SK-BR-3). HDACis and MTIs exhibited dose-dependent antitumor effects in FMC cell lines, and for all inhibitors, the IC50 values were determined, with one feline cell line showing reduced susceptibility (FMCm). Immunoblot analysis confirmed an increase in the acetylation status of core histone protein HDAC3 and flow cytometry showed that HDACis and MTIs lead to cellular apoptosis. Overall, our study uncovers HDACis and MTIs as promising anti-cancer agents to treat FMCs.


Biomedicines ◽  
2022 ◽  
Vol 10 (1) ◽  
pp. 149
Author(s):  
David J. Wooten ◽  
Indu Sinha ◽  
Raghu Sinha

Survival rate for pancreatic cancer remains poor and newer treatments are urgently required. Selenium, an essential trace element, offers protection against several cancer types and has not been explored much against pancreatic cancer specifically in combination with known chemotherapeutic agents. The present study was designed to investigate selenium and Gemcitabine at varying doses alone and in combination in established pancreatic cancer cell lines growing in 2D as well as 3D platforms. Comparison of multi-dimensional synergy of combinations’ (MuSyc) model and highest single agent (HSA) model provided quantitative insights into how much better the combination performed than either compound tested alone in a 2D versus 3D growth of pancreatic cancer cell lines. The outcomes of the study further showed promise in combining selenium and Gemcitabine when evaluated for apoptosis, proliferation, and ENT1 protein expression, specifically in BxPC-3 pancreatic cancer cells in vitro.


2020 ◽  
Author(s):  
Fang Ren ◽  
Gong Zhang ◽  
Caiyu Li ◽  
Gailing Li ◽  
Yuan Cao ◽  
...  

Abstract Background: Hesperetin, an active compound found in citrus fruits, possesses antiproliferative effects toward several types of cancer cell lines, including cervical cancer. In this study, we explore the antitumor effects of Hesperetin on the human cervical cancer human papilloma virus (HPV)-positive (CaSki and HeLa) and HPV-negative (C-33A) cell lines and further elucidated the underlying mechanisms of this action. Methods: Cell viability and proliferation was measured by the MTT assay and 5-ethynyl-2′-deoxyuridine (EdU) incorporation assay, respectively. dUTP-fluorescein nick end-labeling (TUNEL) staining, Annexin V‑fluorescein isothiocyanate (FITC)/propidium iodide (PI) staining and flow cytometry was used to assess the degree of apoptosis. JC-1 staining assay was used to evaluate the change of mitochondrial membrane potential (ΔΨm) and Western blot assays were used to determine apoptosis-related factors at protein level. Results: Hesperetin (100, 200 and 400 μM) exhibited a significant exclusive inhibitory effect against the growth of HPV-infected CaSki and HeLa cancer cells via induction of apoptosis in a concentration-dependent manner, while it was almost not active in HPV-negative C-33A cancer cells and normal cervix epithelial H8 cells. Moreover, this antitumor effect executed by Hesperetin was associated with disruption of ΔΨm, the release of cytochrome c from mitochondria, activation of pro-apoptotic proteins (Bax, cleaved caspase-3 and caspase-9) and inhibition of anti-apoptotic proteins (Bcl-2). During this process, cleaved caspase-8 remained unchanged. In addition, Hesperetin led to a downregulation of E6 oncoprotein expression and upregulation of tumor suppressor protein p53 level. Conclusions: Collectively, these results implicated that Hesperetin can induce apoptosis of HPV‑positive cervical cancer cells via a mitochondria-mediated intrinsic signaling pathway, together with the repression of E6 and enhancement of p53 protein level, indicating Hesperetin may be considered as a potential candidate for the development of innovative anti-HPV cervical cancer agents.


2019 ◽  
Vol 8 (3) ◽  
pp. 89-101
Author(s):  
Jayse Alves ◽  
Georgia Muccillo Dexheimer ◽  
Laura Reckzigel ◽  
Marcia Goettert ◽  
Vanderlei Biolchi ◽  
...  

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e19521-e19521
Author(s):  
Bartlomiej Przychodzen

e19521 Background: Histone deacetylase inhibitors (HDACi) are small molecules that increase acetylation of lysine residues by blocking histone deactylases. These anticancer agents affect epigenetic and non-epigenetic gene expression resulting in cell cycle arrest of cancer cells. Furthermore HDACi can enhance its anti-tumor effects via the pharmacologic modulation of macrophage. Some HDACi’s such as Trichostatin A (TSA) can also affected the tumor immune microenvironment by suppressing the activity of infiltrating macrophages and inhibiting myeloid-derived suppressor cell recruiement (Li et al.,). Methods: We conducted a high throughput screen comparing gene expression profiles in known hematological cell lines to identify transcriptional signatures associated with TSA sensitivity obtained from GDSC. Results: We selected genes that showed at least 2fold expression difference and were statistically significant (p < 0.05). We identified 49 genes that were upregulated and 85 that were downregulated. The most significant results include multiple genes known to be correlated with the B-cell maturation process. We found that CD24 a small GPI linked glycoprotein expressed at the surface of most B lymphocyte precursors, neutrophils, epithelial cells and frequently found to be highly expressed in various hematological and solid neoplasms, was up/downregulatred by XX. Interestingly, CD24 plays a role in the activation and differentiation of the cells as bone marrow samples lacking CD24 resulted in decreased numbers of both pre-B and immature B-cell populations. We also found that IKZF2, a transcription factor regulating lymphocyte development and queiesence and which is frequently deleted in hypodiploid B-ALLs. This result could revelent as other reports suggest a role of IKZF2 as a tumor suppressor with a central role regulating the balance of self-renewal and differentiation in leukemic stem cells. Conclusions: Our study identified transcriptional profiles which suggest that TSA sensitivity could be related to B cell maturation. Further experiments warrant replication of these findings which could prove useful in creating optimal, TSA-based treatments acting either as potent single agents or in combination enhancing anti-tumor effects of immunotherapies.


Blood ◽  
2011 ◽  
Vol 117 (10) ◽  
pp. 2910-2917 ◽  
Author(s):  
Daniela Buglio ◽  
Noor M. Khaskhely ◽  
Kui Shin Voo ◽  
Hector Martinez-Valdez ◽  
Yong-Jun Liu ◽  
...  

AbstractIn Hodgkin lymphoma (HL), the malignant cells are surrounded by a large number of reactive infiltrating inflammatory cells, including OX40-expressing T cells and interleukin 10 (IL-10)–producing regulatory T (T-reg) cells. These T-reg cells can suppress the immune response and thus contribute to the maintenance of immune tolerance and to insufficient antitumor response. The engagement of OX40L with the OX40 receptor is essential for the generation of antigen-specific memory T cells and for the induction of host antitumor immunity. In the present study, we investigated whether histone deacetylase inhibitors (HDACis) may induce a favorable antitumor immune response by regulating the expression of OX40L in HL. We found that HDACis up-regulated OX40L surface expression in HL cell lines in a dose-dependent manner. Small interfering RNAs (siRNAs) that selectively inhibited HDAC11 expression, significantly up-regulated OX40L and induced apoptosis in HL cell lines, and silencing HDAC11 transcripts increased the production of tumor necrosis-α (TNF-α) and IL-17 in the supernatants of HL cells. Furthermore, HDACI-induced OX40L inhibited the generation of IL-10–producing type 1 T-reg cells. These results demonstrate for the first time that HDAC11 plays an essential role in regulating OX40L expression. Pharmacologic inhibition of HDAC11 may produce a favorable antitumor immune response in patients with HL.


Toxics ◽  
2019 ◽  
Vol 7 (3) ◽  
pp. 43 ◽  
Author(s):  
Edna Ribeiro ◽  
Mariana Delgadinho ◽  
Miguel Brito

The worldwide production of synthetic chemicals, including endocrine disruptor chemicals (EDCs), such as Bisphenol A (BPA) has increased significantly in the last two decades. Human exposure to BPA, particularly through ingestion, is continuous and ubiquitous. Although, considered a weak environmental estrogen, BPA can induce divergent biological responses through several signaling pathways, including carcinogenesis in hormone-responsive organs. However, and despite the continuous increase of tumor cell-resistance to therapeutic drugs, such as doxorubicin (DOX), information regarding BPA drug interactions is still scarce, although its potential role in chemo-resistance has been suggested. This study aims to assess the potential interactions between environmentally relevant levels of BPA and DOX at a therapeutic dosage on Hep-2 and MRC-5 cell lines transciptome. Transcriptional effects in key-player genes for cancer biology, namely c-fos, p21, and bcl-xl, were evaluated through qRT-PCR. The cellular response was analyzed after exposure to BPA, DOX, or co-exposure to both chemicals. Transcriptional analysis showed that BPA exposure induces upregulation of bcl-xl and endorses an antagonistic non-monotonic response on DOX transcriptional effects. Moreover, the BPA interaction with DOX on c-fos and p21 expression emphasize its cellular specificity and divergent effects. Overall, Hep-2 was more susceptible to BPA effects in a dose-dependent manner while MRC-5 transcriptional levels endorsed a non-monotonic response. Our data indicate that BPA environmental exposure may influence chemotherapy outcomes, which emphasize the urgency for a better understanding of BPA interactions with chemotherapeutic agents, in the context of risk assessment.


Sign in / Sign up

Export Citation Format

Share Document