scholarly journals Combination treatment optimization using a pan-cancer pathway model

2021 ◽  
Vol 17 (12) ◽  
pp. e1009689
Author(s):  
Robin Schmucker ◽  
Gabriele Farina ◽  
James Faeder ◽  
Fabian Fröhlich ◽  
Ali Sinan Saglam ◽  
...  

The design of efficient combination therapies is a difficult key challenge in the treatment of complex diseases such as cancers. The large heterogeneity of cancers and the large number of available drugs renders exhaustive in vivo or even in vitro investigation of possible treatments impractical. In recent years, sophisticated mechanistic, ordinary differential equation-based pathways models that can predict treatment responses at a molecular level have been developed. However, surprisingly little effort has been put into leveraging these models to find novel therapies. In this paper we use for the first time, to our knowledge, a large-scale state-of-the-art pan-cancer signaling pathway model to identify candidates for novel combination therapies to treat individual cancer cell lines from various tissues (e.g., minimizing proliferation while keeping dosage low to avoid adverse side effects) and populations of heterogeneous cancer cell lines (e.g., minimizing the maximum or average proliferation across the cell lines while keeping dosage low). We also show how our method can be used to optimize the drug combinations used in sequential treatment plans—that is, optimized sequences of potentially different drug combinations—providing additional benefits. In order to solve the treatment optimization problems, we combine the Covariance Matrix Adaptation Evolution Strategy (CMA-ES) algorithm with a significantly more scalable sampling scheme for truncated Gaussian distributions, based on a Hamiltonian Monte-Carlo method. These optimization techniques are independent of the signaling pathway model, and can thus be adapted to find treatment candidates for other complex diseases than cancers as well, as long as a suitable predictive model is available.

2020 ◽  
Author(s):  
Robin Schmucker ◽  
Gabriele Farina ◽  
James Faeder ◽  
Fabian Fröhlich ◽  
Ali Sinan Saglam ◽  
...  

AbstractThe design of efficient combination therapies is a difficult key challenge in the treatment of complex diseases such as cancers. The large heterogeneity of cancers and the large number of available drugs renders exhaustive in vivo or even in vitro investigation of possible treatments impractical. In recent years, sophisti-cated mechanistic, ordinary differential equation-based pathways models that can predict treatment responses at a molecular level have been developed. However, surprisingly little effort has been put into leveraging these models to find novel therapies. In this paper we use for the first time, to our knowledge, a large-scale state-of-the-art pan-cancer signaling pathway model to identify potentially novel combination therapies to treat individual cancer cell lines from various tissues (e.g., minimizing proliferation while keeping dosage low to avoid adverse side effects) and populations of cancer cell lines (e.g., minimizing the maximum or average proliferation across the cell lines while keeping dosage low). We also show how our method can be used to optimize the mixtures and dosages used in sequential treatment plans—that is, optimized sequences of potentially different drug combinations—providing additional benefits. In order to solve the treatment optimization problems, we combine the Covariance Matrix Adaptation Evolution Strategy (CMA-ES) algorithm with a significantly more scalable sampling scheme for truncated Gaussian distributions, based on a Hamiltonian Monte-Carlo method. These optimization techniques are independent of the signaling pathway model, and can thus be used for other signaling pathway models also, provided that a suitable predictive model is available.


2021 ◽  
Vol 12 ◽  
Author(s):  
Jiajun He ◽  
Hongjian Ding ◽  
Huaqing Li ◽  
Zhiyu Pan ◽  
Qian Chen

While many anti-cancer modalities have shown potent efficacy in clinical practices, cancer prevention, timely detection, and effective treatment are still challenging. As a newly recognized iron-dependent cell death mechanism characterized by excessive generation of lipid peroxidation, ferroptosis is regarded as a potent weapon in clearing cancer cells. The cystine/glutamate antiporter solute carrier family 7 member 11 (SLC7A11) is the core target for ferroptosis regulation, the overexpression of which dictates downregulated sensitivity to ferroptosis in cancer cells. Hence, we elaborated the pan-cancer level bioinformatic study and systematically elucidated the role of intra-tumoral expression of SLC7A11 in the survival of cancer patients and potential immunotherapeutic response. Specifically, 25/27 (92.6%) cancers were featured with upregulated SLC7A11 expression, where SLC7A11 overexpression is a risk factor for worse overall survival in 8 cancers. We also validated SLC7A11 expression in multiple pancreatic cancer cell lines in vitro and found that it was upregulated in most pancreatic cancer cell lines (p < 0.05). Single-cell sequencing method revealed the SLC7A11 was majorly expressed in cancer cells and mononuclear cells. To further explore the function of SLC7A11 in cancer progression, we analyzed the influence on cell proliferation after the knockdown or knockout of SLC7A11 by either CRISPR or RNAi methods. Besides, the association between SLC7A11 and drug resistance was characterized using bioinformatic approaches as well. We also analyzed the association between the expression of SLC7A11 in multi-omics level and the intra-tumoral infiltration of immune cells based on cell annotation algorithms. Moreover, the relationship between SLC7A11 and the expression of MHC, immune stimulators, immune inhibitors as well as the response to immunotherapy was investigated. In addition, the SLC7A11 expression in colon adenocarcinoma, uterine corpus endometrial carcinoma, and stomach adenocarcinoma (STAD) is also positively associated with microsatellite instability and that in head and neck squamous cell carcinoma, STAD, and prostate adenocarcinoma is positively associated with neoantigen level, which further revealed the potential relationship between SLC7A11 and immunotherapeutic response.


2019 ◽  
Vol 15 (6) ◽  
pp. 399-405 ◽  
Author(s):  
Julia L. Fleck ◽  
Ana B. Pavel ◽  
Christos G. Cassandras

Sequences of genetic events were identified that may help explain common patterns of oncogenesis across 22 tumor types. The general effect of late-stage mutations on drug sensitivity and resistance mechanisms in cancer cell lines was evaluated.


Proceedings ◽  
2017 ◽  
Vol 1 (10) ◽  
pp. 1016 ◽  
Author(s):  
Melike Ozgul ◽  
Elgin Turkoz Uluer ◽  
Tuna Onal ◽  
Damla Akogullari ◽  
Kemal Ozbilgin ◽  
...  

2012 ◽  
Vol 2 (1) ◽  
pp. 59-68 ◽  
Author(s):  
Perumal Elumalai ◽  
Dharmalingam Nandagopal Gunadharini ◽  
Kalimuthu Senthilkumar ◽  
Sivanantham Banudevi ◽  
Ramachandran Arunkumar ◽  
...  

2020 ◽  
Author(s):  
Mingxue Yu ◽  
Wenli Xu ◽  
Yusheng Jie ◽  
Jiahui Pang ◽  
Siqi Huang ◽  
...  

Abstract Background:Hepatocellular carcinoma (HCC) is a common cancer and the leading cause is persistent hepatitis B virus infection. We aimed to identify some core genes and pathways for HBV-related HCC. Methods: Gene expression profiles of GSE62232, GSE121248, and GSE94660 were available from Gene Expression Omnibus. The GEO2R online tool and Venn diagram software were applied to analyze commonly differentially expressed genes. Then functional enrichment analysis using Gene Ontology (GO) and the Kyoto Encyclopedia of Gene and Genome (KEGG) as well as the protein-protein interaction (PPI) network was conducted. The overall survival rates and the expression levels were detected by Kaplan-Meier plotter and Gene Expression Profiling Interactive Analysis (GEPIA). Next, gene set enrichment analysis (GSEA) was performed to verify the KEGG pathway analysis. Furthermore, quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) was performed to validate the levels of gene expression in tumor tissues from HBV related HCC patients, HBV-related liver cell lines, and transfection si-p53 and knock-out p53 liver cancer cell lines. Finally, the prediction of the ceRNA network was constructed with R software. Results: Fifteen highly expressed genes associated with significantly worse prognoses were selected and CCNB1, CDK1, and RRM2 in the p53 signaling pathway were identified as core genes. GSEA results showed highly-expressed samples of three core genes were all enriched in the p53 signaling pathway in a validation dataset(P<0.0001). Expression of these three core genes were consistently higher in tumor tissue samples (P<0.0001) and liver cancer cell lines (P<0.05). However, transfection si-p53 and knock-out p53 liver cancer cell lines had lower expression (P<0.05). LncRNAs, including NEAT1, MALAT1, XIST, AC021078.1, and SNHG16, were identified by close interactions with core genes. Conclusions: CCNB1, CDK1, and RRM2 were enriched in the p53 signaling pathway and could be potential biomarkers and therapeutic targets for HBV-related HCC.


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.


Sign in / Sign up

Export Citation Format

Share Document