scholarly journals A landscape of synergistic drug combinations in non-small-cell lung cancer

2021 ◽  
Author(s):  
Nishanth Ulhas Nair ◽  
Patricia Greninger ◽  
Adam Friedman ◽  
Arnaud Amzallag ◽  
Eliane Cortez ◽  
...  

Targeted therapeutics have advanced cancer treatment, but single agent activity remains limited by de novo and acquired resistance. Combining targeted drugs is broadly seen as a way to improve treatment outcome, motivating the ongoing search for efficacious combinations. To identify synergistic targeted therapy combinations and study the impact of tumor heterogeneity on combination outcome, we systematically tested over 5,000 two drug combinations at multiple doses across a collection of 81 non-small cancer cell lines. Both known and novel synergistic combinations were identified. Strikingly, very few combinations yield synergy across the majority of cell line models. Importantly, synergism mainly arises due to sensitization of single agent resistant models, rather than further sensitize already sensitive cell lines, frequently via dual targeting of a single or two highly interconnected pathways. This drug combinations resource, the largest of its kind should help delineate new synergistic regimens by facilitating the understanding of drug synergism in cancer.

Cells ◽  
2020 ◽  
Vol 9 (5) ◽  
pp. 1251
Author(s):  
Lukas M. Braun ◽  
Simon Lagies ◽  
Jessica Guenzle ◽  
Stefan Fichtner-Feigl ◽  
Uwe A. Wittel ◽  
...  

Pancreatic ductal adenocarcinoma (PDAC) correlates with high mortality and is about to become one of the major reasons for cancer-related mortality in the next decades. One reason for that high mortality is the limited availability of effective chemotherapy as well as the intrinsic or acquired resistance against it. Here, we report the impact of nab-paclitaxel on the cellular metabolome of PDAC cell lines. After establishment of nab-paclitaxel resistant cell lines, comparison of parental and resistant PDAC cell lines by metabolomics and biochemical assessments revealed altered metabolism, enhanced viability and reduced apoptosis. The results unveiled that acute nab-paclitaxel treatment affected primary metabolism to a minor extent. However, acquisition of resistance led to altered metabolites in both cell lines tested. Specifically, aspartic acid and carbamoyl-aspartic acid were differentially abundant, which might indicate an increased de novo pyrimidine synthesis. This pathway has already shown a similar behavior in other cancerous entities and thus might serve in the future as vulnerable target fighting resistance acquisition occurring in common malignancies.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3946-3946
Author(s):  
Justin Cidado ◽  
Scott Boiko ◽  
Theresa Proia ◽  
Haley Woods ◽  
Omid Tavana ◽  
...  

Abstract Apoptosis is controlled through the dynamic interactions of the Bcl2 protein family, and cancers have evolved mechanisms to hijack this pathway to evade apoptosis, often by upregulating anti-apoptotic proteins (e.g. Mcl1, Bcl2). This survival adaptation creates a dependency that could be exploited therapeutically, which is why considerable effort has been made to develop small molecule inhibitors of the anti-apoptotic Bcl2 family proteins. This class of drug was clinically validated with the approval of venetoclax, a selective Bcl2 inhibitor, for the treatment of CLL. Venetoclax is undergoing evaluation in numerous other clinical trials for predominantly hematologic malignancies. Despite impressive responses observed with venetoclax in CLL (ORR 79%), acquired resistance is beginning to emerge. Likewise, other hematologic indications are more intrinsically resistant to venetoclax, exhibiting much lower response rates in the respective Phase I clinical trials (AML = 38%, NHL = 44%, MM = 12%). Preclinical studies with venetoclax have reported increased levels of other anti-apoptotic proteins as a likely mechanism contributing to both de novo and acquired resistance. Therefore, combining cell death inducing agents that inhibit Mcl1 or BclxL could be a means of combating resistance. Inhibition of cyclin-dependent kinase 9 (CDK9), which regulates transcription elongation, has been reported to reduce protein levels of genes with short-lived transcripts and proteins, such as MCL1. AZD4573 is a novel and selective CDK9 inhibitor that shows potent single agent activity, inducing cell death in vitro and tumor regressions in vivo in a diverse set of hematologic cancers (Cidado et. al., AACR Annual Meeting 2018). AZD4573 is currently being evaluated in a Phase I clinical trial for patients with hematological malignancies (NCT03263637). This study evaluates whether combinations of AZD4573 with other cell death inducing agents could overcome de novo venetoclax monotherapy resistance. A panel of 12 AML and 6 DLBCL cell lines were treated for 6 h with venetoclax, AZD4573, and a selective BclxL probe compound (AZ'3202) either as single agents or in combinations and assayed for caspase activation. The Loewe model was used to calculate synergy scores to assess benefit over monotherapy, and combinations with scores >5 were deemed beneficial. Treatment with AZD4573+venetoclax and AZD4573+AZ'3202 resulted in beneficial combinations for 13/18 and 10/18 cell lines, respectively. On the other hand, venetoclax+AZ'3202 showed significant combination benefit in only two cell lines (NB4, SUDHL4), suggesting a primary dependency upon Mcl1 for most of these hematologic cancer cell lines. Interestingly, cell lines sensitive to single agent AZ'3202 (4 AML, 0 DLBCL) did not show any combination benefit when treated with AZD4573+venetoclax, highlighting the exquisite dependency of those four models upon BclxL and mutual exclusivity with Mcl1. Cell lines benefitting from the AZD4573+venetoclax combination tended to fall into one of two categories: having single agent activity to either agent that is enhanced by the combination or having no single agent activity but the combination shifts the cell line into a responder. SUDHL4 cells were sensitive to AZD4573 (caspase activation EC50 = 18 nM) but not venetoclax (EC50 = 476 nM) while OCI-AML3 was insensitive to both (EC50 > 30 µM). In vitro biomarker kinetic analysis revealed an increase in Mcl1 levels (~2-fold) after 3 h of venetoclax treatment that was abrogated upon combination treatment, providing a mechanistic rationale for the combination benefit. Furthermore, when tested in an OCI-AML3 tumor xenograft study in mice, AZD4573 or venetoclax monotherapy exhibited minimal tumor growth inhibition (44% and 16%, respectively) while the combination led to tumor regressions (64%) with minimal effect on body weight. In a SUDHL4 tumor xenograft study, venetoclax monotherapy displayed minimal tumor growth inhibition (25%), but intermittent dosing of AZD4573 exhibited 94% tumor growth inhibition. Still, combination therapy demonstrated a clear benefit as it led to complete tumor regressions with 6/8 mice remaining tumor-free until the end of study (150 days). Together, this work presents supporting evidence that combining cell death inducing agents would be effective at overcoming de novo or acquired resistance associated with monotherapy treatments. Disclosures Cidado: AstraZeneca: Employment, Equity Ownership. Boiko:AstraZeneca: Employment. Proia:AstraZeneca: Employment. Woods:AstraZeneca: Employment. Tavana:AstraZeneca: Employment. San Martin:AstraZeneca: Employment. Tron:AstraZeneca: Employment. Shao:AstraZeneca: Employment. Drew:AstraZeneca: Employment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2639-2639
Author(s):  
Silvia CW Ling ◽  
Angela M Nikolic ◽  
Ammira Al-Shabeeb ◽  
Edwin Lau ◽  
Phoebe Joy Ho ◽  
...  

Abstract Bortezomib is the first proteasome inhibitor in clinical use. It is particularly effective in myeloma compared with other cancers; as a single agent, its response rate in relapsed myeloma is about 40%. Nevertheless, a large proportion of patients are primarily resistant and acquired bortezomib resistance is also an emerging issue. The mechanism of bortezomib resistance in myeloma remains elusive. We have previously shown that bortezomib sensitivity correlates inversely with the levels of XBP-1 mRNA in vitro and in myeloma patients. XBP-1 is a major regulator of the unfolded protein response (UPR), which is essential for immunoglobulin folding and assembly, and plasma cell development. Previous studies suggested that immunoglobulin production sensitises myeloma cell lines to proteasome inhibitors (S Meister, et al., 2007, Cancer Res 67:1783). We hypothesize that dependence on XBP-1 and the UPR mediate sensitivity to bortezomib. The goal was to develop bortezomib resistant myeloma cell lines, study their phenotype, characterise the mechanism of resistance and, in particular, study the changes in the UPR. Bortezomib resistant sublines (9 in total) were derived from 4 myeloma cell lines (KMS-11, H929, U266 and OPM2), by long term exposure to bortezomib. The fold resistance varies from 3 to 12 and is stable for at least 4 passages without bortezomib. During the evolution of resistance, the myelomas all adopted adherent growth, independent of the morphology of the parent cell lines. When adherent versions of the parent cell lines were derived without exposure to bortezomib, they were, surprisingly, resistant to bortezomib. All bortezomib-resistant myeloma sublines downregulated XBP-1 mRNA levels and the reduction was correlated with the fold resistance. Moreover, splicing of XBP-1 mRNA to the active form was also reduced. The adherent versions of the parent cell lines that showed de novo resistance to bortezomib also had lower levels of XBP-1. Bortezomib normally induces XBP-1 expression and splicing acutely within 12–24 hours (AH Lee, et al., 2003. PNAS 100: 9946) but the resistant sublines were less responsive than the sensitive parents in this respect. Protein levels of other components of the UPR were evaluated by immunoblotting; BiP, phosphorylated-eIF2α, ATF6α and p58IPK were found to be unchanged in the resistant sublines. Cytoplasmic and secreted immunoglobulins were assayed by immunoblotting and ELISA respectively but no differences between the parent and resistant sublines were observed. In a small cohort of relapsed/refractory patients whose bone marrow biopsy was collected prior to bortezomib treatment, the quantity of intracytoplasmic light chain in the myeloma cells, as detected by flow cytometry, did not predict clinical response. The bortezomib-resistant myeloma sublines showed significant cross-resistance to doxorubicin and, to a lesser extent, vincristine, but none to melphalan. The cross-resistance is associated with adherent growth, as it was also observed in the adherent versions of the parent cell lines which were never exposed to bortezomib. Moreover, functional analysis of the multi-drug transporter activity, by drug accumulation assay, showed no difference between parent and resistant sub-lines. These results suggest that resistance to bortezomib is associated with downregulation of the IRE-XBP-1 pathway, but not necessarily other components of the UPR, consistent with observations by others concerning BiP (DT Rutkowski, et al., 2006. Plos Biology 4, e374). The association between adherence, bortezomib resistance and low XBP-1 requires further study.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 434-434 ◽  
Author(s):  
Jason Smith ◽  
Katherine J. Walsh ◽  
Cassandra L Jacobs ◽  
Qingquan Liu ◽  
Siyao Fan ◽  
...  

Abstract Abstract 434 Background Histone deacetylase inhibitors (HDACis) have demonstrated significant clinical activity in hematologic malignancies; however, single agent response rates have ranged between 20–50% with the duration of response often measured in months, suggesting that drug resistance is a major mode of failure. The pathways through which these agents work and the means by which tumors develop resistance to them are poorly understood. Combination therapy targeting multiple oncogenic pathways holds the promise to improve upon both the depth and durability of these responses. We investigated the mechanisms of inherent and acquired resistance to HDACis in a broad range of lymphomas. By detailing the molecular pathways implicated in resistance to HDACi, we sought to identify novel combinations of compounds that could overcome potential mechanisms that confer resistance. Methods and Results We tested two separate HDACis, LBH589 and SAHA in 51 cell lines representing a wide range of lymphomas including Burkitt lymphoma, diffuse large B cell lymphoma (DLBCL), mantle cell lymphoma, and Hodgkin lymphoma. Gene expression array data was generated for all these cell lines. We then identified genes that were significantly associated with resistance to both LBH589 and SAHA (p<.01) and applied hierarchical clustering to identify the functional significance of these genes. Histology was not predictive of sensitivity to either HDACi. These data were then analyzed using gene set enrichment to identify known molecular pathways associated with resistance. Activation of JAK/STAT signaling was found to be a major determinant of resistance among the cell lines that were relatively resistant to HDACi. (P<0.001, FDR <.25). To determine whether these genes that we found to be associated with resistance reflected potential mechanisms of acquired resistance to HDACi therapy, we separately engineered resistance to LBH589 and SAHA in three DLBCL cell lines (LY3, BJAB, Farage) through incremental dose escalation over a period of up to 6 months. Each of these three cell lines demonstrated sustained growth at drug concentrations that were at or above their original IC50. Each of these cell lines were then exposed to the other HDACi and tested for cross resistance. In each case, the cell lines demonstrated complete cross-resistance to the other drug. We then profiled the gene expression of these cell lines that had acquired resistance. Similar to our previous results, these cell lines demonstrated increased signaling through the JAK/STAT pathway, suggesting that mechanisms of inherent and acquired resistance are similar. We therefore reasoned that combining HDAC and JAK inhibition may overcome both inherent and acquired resistance. To investigate this hypothesis, we tested LBH589 and INCB018424, a JAK1/2 inhibitor, alone and in combination in the LY3, TMD-8, U2932, and BJAB cell lines. While INCB018424 demonstrated no single agent cytotoxicity, it yielded a high degree of synergy when combined with LBH589 with the combination index computed by the Chou-Talalay method ranging from .19 to .9. Conclusion HDACis show single agent activity in the treatment of a number of hematologic malignancies, however most patients develop resistance to these drugs after relatively short-lived remissions. Thus, the greatest promise of these drugs may lie in combination with other agents that target molecular pathways that underlie resistance to these drugs. Using gene expression profiling of a broad range of tumor types and sensitivity to HDACis we were able to identify activation of the JAK/STAT pathway as a common feature of inherent and acquired resistance to HDACis. We combined the JAK1/2 inhibitor INCB018424 with LBH589 and demonstrated a high degree of synergy. As the number of small molecule inhibitors with clinical activity increases, the need to identify rational preclinical combinations becomes greater. Pairing gene expression profiling and resistant cell lines is a promising approach to the selection of combinations likely to maximize clinical benefit while limiting toxicity. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1455-1455
Author(s):  
Ansu Abu Alex ◽  
Hamenth Kumar P ◽  
Saravanan Ganesan ◽  
Nithya Balasundaram ◽  
Kavitha M Lakshmi ◽  
...  

Abstract NK cells are primary effectors of the innate immune response against cells that have undergone malignant transformation. Several lines of evidence indicate that the expression level of NK ligands on leukemic cells affects the sensitivity of the leukemic cells to cytolytic activity by NK cells. Various agents have been evaluated for their ability to induce these ligands on leukemic cells to augment the NK cell mediated anti-leukemia effect. There is substantial evidence that has established the importance of the adaptive immune system in the treatment of acute promyelocytic leukemia (APL) (Rose Ann Padua et al. Nat Med 2003). While there is significant data which address the mechanisms of arsenic trioxide (ATO) on malignant promyelocytes, limited data is available of its effect on the innate and adaptive immune system. We undertook a series of experiments to address the impact of ATO on NK cell receptor and malignant promyelocyte ligand expression and its effect on NK cell mediated cytotoxicity. We also evaluated NK cell reconstitution in patients treated with ATO and the impact of KIR genotypes on relapse. We first evaluated the cytotoxic activity of NK92MI (NK cell line) against 5 different myeloid (K562, U937, HL60, UF1, NB4) and 2 lymphoid cell lines (Jurkat E6.1, SUP-B15) by CFSE/ 7AAD cytotoxicity assay. Target (T) cells (1x 105/100 µL/well) pre-treated with CFSE were co-cultured with effector NK cells (E) at a E:T ratio of 1:1, 2:1 and 5:1 for 5 hours at 37°C in 96 well plates. The percentage cytolytic activity of the NK cells was then calculated after adding 7AAD and acquired in FACS Calibur (Becton Dickinson, San Jose, CA, USA). Significant cytolytic activity was noted against K562 and NB4 cell lines. At the highest E:T ratio there was a median 22% cytolytic activity against NB4 (N=5). We observed that NB4 when treated overnight with 1µM ATO (>99% viability retained after this exposure) significantly increased the cytotoxic effect of NK92MI cell line at all the E:T ratios as shown in figure 1A (n=5; P=0.0023). No other cell line showed a similar increase in cytotoxic effect following exposure to ATO at these concentrations (data not shown). We next evaluated the effect of exposure of NB4 cells to ATO at 1µM for 6 hours on NK ligand expression by flowcytometry. As shown in figure 1B there was a significant increase in activating ligand MICA/B in NB4 cell lines (n=3; P=0.016) which was not seen in any of the other cell lines. Similar significant increased expression of Nectin-2 (DNAM-1 ligand) and HLA Class I was seen. Exposure of NK92MI to ATO for 6 hours at 1uM (non cytotoxic dose:IC50-3.8uM) resulted in increased expression of activating receptors NKG2D, NKP30 and KIR2DS4 (figure 1C) and inhibitory receptor NKG2A and decrease in inhibitory receptors KIR3DL1/DL2. There were no changes in the expression of NKP46, KIR2DL1, KIR2DL2 and DNAM1 receptors. We undertook a prospective study to evaluate the pattern of NK (CD56+CD3-) reconstitution in patients with newly diagnosed APL treated at our center with a single agent ATO regimen (Mathews et al. JCO 2011). The mean NK cell counts in patients were below the 2SD deviation level of the normal range even after completion of therapy (approximately a year)(figure 1D). All other subsets evaluated (CD4, CD8, CD3, CD19, CD56+CD3+, CD4CD45RO) had returned to levels within the normal range by the end of consolidation therapy (approximately 3 months from diagnosis). KIR genotyping was done on 55 patients with APL who received treatment with single agent ATO based regimen. The median follow up of this cohort was 20 months and 14 cases relapsed following initial therapy. The presence or absence of 17 KIR genes was done by PCR-SSP method (KIR Typing kit, Miltenyi Biotech Inc, CA). There was no association with any specific genotype or haplotype with risk of relapse. In summary we have noted that there is up regulation of receptors on NK cells and ligands on malignant promyelocytes following exposure to ATO that favors NK cell mediated cytotoxicity. In-vitro we have demonstrated a significant increase in NK cell mediated cytolytic activity against malignant promyelocytes exposed to ATO even at relatively low E:T ratios. This could be an important mechanism by which ATO induces durable remissions in patients with APL. The delayed NK cell recovery following treatment with ATO raises the possibility of using NK cell therapy to augment the effect of ATO in the treatment of patients.Figure 1Figure 1. Disclosures: No relevant conflicts of interest to declare.


2018 ◽  
Vol 16 (05) ◽  
pp. 1850017 ◽  
Author(s):  
Aman Sharma ◽  
Rinkle Rani

Combination drug therapy is considered a better treatment option for various diseases, such as cancer, HIV, hypertension, and infections as compared to targeted drug therapies. Combination or synergism helps to overcome drug resistance, reduction in drug toxicity and dosage. Considering the complexity and heterogeneity among cancer types, drug combination provides promising treatment strategy. Increase in drug combination data raises a challenge for developing a computational approach that can effectively predict drugs synergism. There is a need to model the combination drug screening data to predict new synergistic drug combinations for successful cancer treatment. In such a scenario, machine learning approaches can be used to alleviate the process of drugs synergy prediction. Experimental data from a single-agent or multi-agent drug screens provides feature data for model training. On the contrary, identification of effective drug combination using clinical trials is a time consuming and resource intensive task. This paper attempts to address the aforementioned challenges by developing a computational approach to effectively predict drug synergy. Single-drug efficacy is used for predicting drug synergism. Our approach obviates the need to understand the underlying drug mechanism to predict drug combination synergy. For this purpose, nine machine learning algorithms are trained. It is observed that the Random forest models, in comparison to other models, have shown significant performance. The [Formula: see text]-fold cross-validation is performed to evaluate the robustness of the best predictive model. The proposed approach is applied to mutant-BRAF melanoma and further validated using melanoma cell-lines from AstraZeneca-Sanger Drug Combination Prediction DREAM Challenge dataset.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 797-797 ◽  
Author(s):  
Jason Stephansky ◽  
Katsuhiro Togami ◽  
Mahmoud Ghandi ◽  
Joan Montero ◽  
Nick vonEgypt ◽  
...  

Abstract SL-401 is a novel targeted therapy comprised of recombinant interleukin 3 (IL3) fused to a truncated diphtheria toxin (DT) payload. SL-401 delivers DT to cells expressing the IL3 receptor (CD123). After internalization, DT catalyzes ADP ribosylation of eukaryotic elongation factor 2 (eEF2), blocking protein synthesis and killing target cells. SL-401 is currently in clinical trials for CD123+ cancers, including acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN). Other than CD123 expression, the determinants of response are largely unknown. Our goal was to study mechanisms of de novo and acquired resistance to inform combination strategies and enhance the efficacy of SL-401. In 16 AML and BPDCN patients enrolled in a phase 1-2 trial, we did not see decreased expression of CD123 by flow cytometry during or after SL-401. To study alternative resistance mechanisms, we generated 3-6 independent SL-401 resistant clones from each of 4 CD123+ AML (THP1, NOMO1, EOL1) or BPDCN (CAL1) cell lines. We treated cells with the LD95 (lethal dose to 95%), retreating upon recovery. All lines developed &gt;2-3 log resistance to SL-401 within 28 days. All resistant clones maintained CD123 surface expression, consistent with what we observed in patients. Using confocal microscopy, we saw that a fluorescently tagged SL-401 was internalized equally in resistant and parental cells. SL-401 resistant cells were also resistant to full length DT, suggesting that the mechanism of resistance involved DT rather than IL3 binding/internalization. We performed whole transcriptome RNA-sequencing and whole exome sequencing (WES) on parental and SL-401 resistant cells. There were no recurrent acquired DNA mutations. However, in RNA-seq the most downregulated gene in 6 independent clones from 2 lines was DPH1 (FC -7.5, FDR&lt;0.0001). DPH1 is the first enzyme in a cascade that converts histidine 715 on eEF2 to diphthamide, the direct target for ADP ribosylation by DT. Decreased expression of DPH1 was confirmed in the 6 resistant clones by qRT-PCR, and in 3 clones from an additional line. Across 33 cell lines and subclones there was an inverse linear correlation between DPH1 level and SL-401 IC50 (P=0.0005). To validate this finding in patients, we performed paired RNA-seq on CD45+CD123+ sorted blasts from 2 AMLs pre & post 2 cycles of SL-401. Both patients' AMLs had reduced DPH1 after exposure to SL-401 (mean -2.1 fold). To determine if loss of DPH1 was sufficient to confer de novo resistance to SL-401, we generated THP1, NOMO1, and CAL1 cells stably expressing the Cas9 nuclease and transduced them with 1 of 4 DPH1-targeting or 2 non-targeting CRISPR sgRNAs co-expressing GFP. Four days after infection at titers that achieve ~20% GFP+ cells, we treated with the LD95 of SL-401. We observed a survival advantage for DPH1 sgRNA-transduced cells (2.9 fold increase in GFP+ cells, P&lt;0.0001) and decreased apoptosis measured by AnnexinV positivity, compared to GFP- cells in the same cultures. By contrast, there was no survival advantage for cells transduced with control sgRNAs. These data suggest that DPH1 loss is sufficient to mediate SL-401 resistance in AML and BPDCN. Pseudomonas toxin (PT) also targets eEF2 on diphthamide, and PT resistance in a B-ALL cell line has been associated with DPH1 loss via reversible promoter CpG DNA methylation (Hu, Leuk Res 2013). Therefore, we tested the DNA methyltransferase inhibitor azacitidine in combination with SL-401 and observed synergistic cytotoxicity, in naïve (combination index (CI) = 0.45; &lt;1 indicates synergy) and SL-401 resistant (CI = 0.55) cells. Most strikingly, 4-week pulsatile treatment with non-lethal "epigenetic" doses of azacitidine (300 nM 2d on/2d off) fully reversed SL-401 resistance in 6 CAL1 and THP1 clones that were insensitive at baseline (Figure). Controls grown in vehicle or with weekly SL-401 challenge showed no reversion, suggesting that azacitidine had a specific sensitizing effect. Restoration of SL-401 sensitivity was accompanied by an increase in DPH1 expression compared to resistant clones. In summary, we found that DPH1 is a biomarker of SL-401 activity and acquired resistance, and resistance is reversible by azacitidine. Based on these data, we have initiated a multicenter phase 1 trial of the combination of SL-401 and azacitidine in patients with AML or MDS (NCT03113643), with correlative laboratory studies designed to explore these hypotheses. Disclosures vonEgypt: Stemline Therapeutics: Employment. Lindsay: Stemline Therapeutics: Employment. Brooks: Stemline Therapeutics: Employment, Equity Ownership, Patents & Royalties. Lane: Stemline Therapeutics: Research Funding; N-of-one: Consultancy.


2020 ◽  
Vol 48 (W1) ◽  
pp. W494-W501 ◽  
Author(s):  
Heewon Seo ◽  
Denis Tkachuk ◽  
Chantal Ho ◽  
Anthony Mammoliti ◽  
Aria Rezaie ◽  
...  

Abstract Drug-combination data portals have recently been introduced to mine huge amounts of pharmacological data with the aim of improving current chemotherapy strategies. However, these portals have only been investigated for isolated datasets, and molecular profiles of cancer cell lines are lacking. Here we developed a cloud-based pharmacogenomics portal called SYNERGxDB (http://SYNERGxDB.ca/) that integrates multiple high-throughput drug-combination studies with molecular and pharmacological profiles of a large panel of cancer cell lines. This portal enables the identification of synergistic drug combinations through harmonization and unified computational analysis. We integrated nine of the largest drug combination datasets from both academic groups and pharmaceutical companies, resulting in 22 507 unique drug combinations (1977 unique compounds) screened against 151 cancer cell lines. This data compendium includes metabolomics, gene expression, copy number and mutation profiles of the cancer cell lines. In addition, SYNERGxDB provides analytical tools to discover effective therapeutic combinations and predictive biomarkers across cancer, including specific types. Combining molecular and pharmacological profiles, we systematically explored the large space of univariate predictors of drug synergism. SYNERGxDB constitutes a comprehensive resource that opens new avenues of research for exploring the mechanism of action for drug synergy with the potential of identifying new treatment strategies for cancer patients.


2012 ◽  
Vol 18 (10 Supplement) ◽  
pp. B24-B24
Author(s):  
Raquel Aloyz ◽  
Caroline Rousseau ◽  
Zuanel Diaz ◽  
Adriana Aguilar-Mahecha ◽  
Mark Basik ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4803-4803
Author(s):  
Lorenzo Falchi ◽  
Monica Schippa ◽  
Debora Luzi ◽  
Rita Emili ◽  
Viola Festuccia ◽  
...  

Abstract Introduction Progressive improvement has been observed in CKR and survival of CML patients (pts) in response to interferon (IFN)a-based regimens, or imatinib. The purpose of this study is the evaluation of: rate of, time to and duration of CKR in accordance to first line therapy employed and Sokal score; impact on overall survival of CKR, and Sokal score, separately considered or combined together. Patients. 109Ph’+ and 5Ph’−, (BCR-ABL positive), CML pts were treated at diagnosis with allogeneic transplantation (3 pts), hydroxyurea (HU) (19 pts), INFa (51pts, G1), INFa associated with ARA-C (20 pts, G2), imatinib alone (18 pts, G3), or imatinib combined with INFa (3 pts, G4). INFa was employed as second line therapy in 12 pts initially treated with hydroxyurea (G5), while INFa/ARA-C combination or imatinib alone was given to 24 (G6) and 23 (G7) pts with de novo or acquired resistance or intolerance to INFa. Third line therapy, consisting of the combination of imatinib with IFNa, was employed in 11 (G8) pts with no CKR (5 pts) or in complete cytogenetic, but not in molecular remission (6 pts). Results. 40 of 94 Ph’+ evaluable non-allotransplanted pts obtained one or more (overall 47) CKRs to INFa-based regimens or imatinib. CKR rate, median time to CKR and response duration are shown in table 1. In the analysis according to Sokal score 82/94 pts, with complete prognostic data at diagnosis, were included. The percentage of responders was higher in the low compared to the non-low Sokal risk group (57% vs. 31%). Irrespective of the treatment, median duration value of the first CKR was also better in the former [18+mths(1–64)] than in the latter group [6mths(2–54)] with 16 vs.4 pts still in first or subsequent remission. Overall survival for CKRs was 68+mths(5–275) vs. 52mths(5–270) for CKRs with 35 vs. 6 pts still alive respectively. Overall survival according to Sokal score at diagnosis was 61+ mths(5–275) for low vs 53mths(5–212) for non-low risk patients. The impact on survival of CKR and Sokal risk were then analyzed simultaneously. The median survival of 27 CKRs and 20 not CKRs with low Sokal risk were 61+mths(5–275)and 63 mths(14–270) respectively as compared to 73+mths(11–212) of 11 CKRs and 36mths(5–139) of 24 not CKRs with unfavourable characteristics at diagnosis. The number of patients still alive in these 4 groups were 24/27, 3/20, 8/11, 3/24 respectively at the time when this analysis was performed. Conclusions. The present data not only confirm the effectiveness of imatinib-over the INFa-based regimens in inducing CKR, but also suggest that response to treatment may be better than Sokal risk in predicting patient survival. Rate,time to and duration of CKR according to treatment PT Group CKR rate(%) time to CKR( months) Duration of CKR (months) N° of pts in cCKR Median Range Median Range G1 17 16 (3–28) 6 (2–53) 0 G2 30 11 (3–24) 15,5 (2–33) 0 G3 85 6 (2–14) 10+ (1–44) 11 G4 100 4 (4–5) 48 (2–50) 1 G5 0 0 G6 9 9,5 (7–12) 40,5 (17–64) 1 G7 54 9 (4–38) 21,5+ (3–54) 6 G8 80 3 (2–4) 26+ (4–48) 9


Sign in / Sign up

Export Citation Format

Share Document