View Article

Abstract

Because of the wide range of tumor types, elevated toxicity, restricted effectiveness, and emergence of therapy tolerance, the problem of how to successfully discover new drugs for cancer patients remains unanswered. The majority of medications on the market today work by modifying several different cellular targets in order to produce their biological effects. A fresh framework known as polypharmacology, which is concerned with looking for several target medications to interfere with networks linked to disease, was created. The advancements in network biology have significantly improved our comprehension of the complex route connections in cancer. Hopkins pioneered network pharmacology, which improves the high rate of clinical success with fewer adverse effects. Network pharmacology may be deemed a suitable technique for drug discovery for complicated disorders like cancer when combined with comprehensive strategies grounded in polypharmacology and networking dynamics. Approaches like "-omics" are used in network pharmacology to detect differences at the fundamental molecular and cellular levels in cancer and other diseases in reaction to the particular medication and/or pathology. A deeper understanding of the causes of medication failure in terms of toxicity, side effects, and clinical efficacy may also be possible using networking pharmacology. Pharmacology networks are able to help avoid the problems associated with cancer treatment and expedite the development of novel chemotherapy drugs.

Keywords

Network Pharmacology, Systems biology, Polypharmacology, Drug discovery

Introduction

As a neoplasmic disease, cancer causes aberrant uncontrolled cellular division and the growth of cellular masses.According to the World Cancer Report, 1.16 million people were expected to new cases with 2.26 million deaths and 784,800 deaths over a five-year period prevalent incidents in India. The eight categories for cancer treatment, as specified by the National Cancer Institute, include immunotherapy, medications, surgery, targeted therapy, chemotherapy, and stem cell transplantation.1 Acquired drug resistance, which can arise from both targeted and cytotoxic chemotherapy treatments, is the result of certain medicines that have produced drug resistance following an initial positive response. Moreover, functional proteins and many gene connections have a role in cancer.Because of the wide range of tumor types, elevated toxicity, restricted effectiveness, and emergence of therapy tolerance, the problem of how to successfully discover new drugs for cancer patients remains unanswered.2 It is the inability for a "one gene, one drug, one disease" concept that leads for this anticancer treatment problem.3,4 As a result, scientific research is actively developing innovative, powerful, and nontoxic remedies.5

Many scientific and technical advancements have affected pharmaceutical research during the previous few decades. The amount of new medications licensed or brought to clinical use during that time sharply decreased, despite enormous investments in medical research and development. It is often acknowledged that drug development attrition can be ascribed to elements like resistance to medicines, significant population heterogeneity, and toxicological results or therapeutic safeguarding for potential drugs in early-stage studies.6 The majority of medications on the market today work by modifying several different cellular targets in order to produce their biological effects. Polypharmacology is an emerging perspective, that emphasizes on finding multiple target drugs to break down networks linked to disease, was created as a result of the serendipitous consequences of pharmaceutical effect on many targets and conceptual examination of biologic connections.7,8 It has been demonstrated that single target medication interference is ineffective in treating complicated chronic conditions include neurological diseases, HIV and AIDS, cardiac ailments, and malignancy.9-11 On the other hand, medications with numerous targets may be most effective and least harmful.12 Therefore, signaling network analysis is a very appealing approach due to its ability to look into intricate relationships.13,14

The advancement of computational methods has led to the suggestion that disease networks created using network biology can be effective tools for excluding pharmacological targets.

Hopkins pioneered network pharmacology, which improves the high rate of clinical success with fewer adverse effects. This method has been responsible for about 40% of the current drug discoveries.3 Over the past few years, network-based techniques like network pharmacology and network medicine have grown significantly.7,10,15 A new area of study called network pharmacology offers a valuable resource for identifying fresh targets for thoughtful drug discovery.16 Systems polypharmacology techniques are frequently needed in drug discovery to address issues such developing resistance or ineffectiveness against single targeted molecules.17   The development of practical network interpreting and medication target estimation using open database sources are made easier by network pharmacy. By contrasting how a medication interacts with its target model, these methods aid in the investigation of the fundamental mechanisms by which medications impact the networks of biology.18 Natural plant products are generally acknowledged as a promising revolutionary antitumor drug discovery using a novel lead resource. The primary source of medications was thought to be a supplementary chemical compounds obtained from plants in ancient healthcare systems, including those in India across the globe because of their greater structural diversity, low toxicity and complexity of the body's metabolism instead of synthetic medications.19 Due to their availability in nature, these bioactive natural chemicals have the potential to block prospective targets, lower the cost of developing novel drugs, and offer the possibility of combination therapy.20

Network Biology to Network Pharmacology

The development of extremely powerful big data analysis technology has created new opportunities for the discovery of more fascinating and potent medicinal and diagnostic treatments.21,22 Knowing the extent to which the proteins obstruct the intricate regulatory apparatus's ability to function is essential.23 As a result, network biology formed, that maintains broad rules governing biological networks offer a novel theoretical framework that eventually modifies our comprehension of the biology of illnesses. In the 21st century, a number of methods for building regulatory networks were put forth that investigated the association between illness phenotypes and genotypes using computer tools, mainly data mining.24 Network biology discoveries has demonstrated that focusing just one protein is ineffective for mitigating complex diseases.14,25 It led to the understanding of polypharmacology by drug developers, who had previously believed that it was an inadequate strategy which required to be eliminated in order to create a workable many target medication.26,27 Extremely specialized single-target medications have dramatically changed to multi-targeted drugs using the advent of networking pharmacology as an entirely autonomous technique. The following century saw the convergence of polypharmacology and system biology in a number of health-related fields.28-30

Transitioning from Polypharmacology to Network Pharmacology

A synergistic approach that can inhibit the action of harmful proteins is highly preferred as it can target entire connections of biomolecules underlying the illness condition, as different components that are functional in plants for medicinal purposes lack robust relationships with the compliance network's effector proteins.31,32 Combining network biology and polypharmacology can help comprehend the pharmacological activity of herbal remedies and expand our understanding of druggable targets.7,33,34 The outcomes of synergy and polypharmacology are preparing their way to uncover novel medications in the upcoming big data time period. The field of drug discovery is expanded by polypharmacology.35-37 Strong binding affinities serve as the defining characteristic of the ordered connections made between molecules. The development of licensable medications without adverse effects has been made possible by the integration of polypharmacology with advances in structural biology and chemoinformatics.38,39

In silico analysis and virtual pharmacology may be helpful new techniques for studying herbal medicine. Primarily, one should take into account the underlying pharmacology. Certainly, before these substances are used in in vitro trials, advances will result from more comprehensive research on the availability of natural substances. Additionally, studying the pharmacodynamics of natural substances is essential to comprehending network pharmacology of medicinal plants and pharmacological combinations.18,40,41

Is network pharmacology a good fit for drug development in cancer?

Changes in multiple network of cells lead to the complex disease known as malignancy, and it is thought that sophisticated treatment strategies are needed.42,43 One of the major problems for the future is to identify medications that interact with many pathways or identify potential therapeutic combinations with the goal of reducing neoplastic disorders by understanding human cell signaling networks and the way they change in different types of tumor.44 Remarkably minimal functional consequences result from changing or eliminating a single protein, even in cancer. In order to achieve maximum efficacy, biological network interventions should be both numerous and highly selective in order to prevent harmful side effects on healthy organs.45 The advancements in network biology have caused a significant increase in our understanding of multifaceted route connections in cancer. This could facilitate a deeper comprehension of signature datasets that together unravel intricate drug action processes.46

Network pharmacology may be deemed a suitable technique for drug discovery for complicated disorders like cancer when combined with comprehensive methods grounded in polypharmacology and network biology. As a result, pharmacology of networks is becoming more established in the field of cancer care.

Molecular categorization of cancer subtypes

The knowledge, diagnosis, and treatment of cancer depend heavily on the concept of cancer subtyping. The molecular classification of cancer subgroups that are not histologically identifiable is aided by network pharmacology. Recently, the Cancer Genome Atlas Research Network (TCGA) finished a thorough molecular study of many malignancies; bladder urothelial adenocarcinoma47, gastric adenocarcinoma48, renal cell carcinoma (KIRC)49, endometrial cancer (UCEC)50, acute myelogenous leukemia (AML)51, breast cancer (BRCA)52, lung squamous cell carcinoma (LUSC)53, colon (COAD) and rectal (READ) adenocarcinomas54, serous ovarian carcinoma (OV)55 and glioblastoma multiforme (GBM)56. This analysis has demonstrated that founded on genetic modifications that align along common routes, three to four subgroups can be distinguished within every form of single-tissue carcinoma.57 A dense multinomial logit framework for extremely dimensional multinomial categorization with network constraints was created by Tian et al. and can be used to predict cancer subtypes.58 Graph-based logistic regression Zhang et al. created Laplacian regularization, an efficient approach identifying important pathways and modules connected to disease subtypes.59

Frameworks of Network Pharmacology

Network pharmacology emerged as a powerful tool for systematically shedding light on complex biological relationships. Approaches like "-omics" are used in network pharmacology to detect differences at the fundamental molecular and cellular levels in cancer and other diseases in reaction to the particular medication and/or pathology. The developed set of variables helps to generate networks from the level of genomics to the level of metabolomic with the goal to classify biological events in disease conditions. The abundance of multi-omics datasets for tumor creates novel pathways for merging data, promising a deeper comprehension of cancer and physiologically and therapeutically significant tumor classification.60 The "-omics" technologies have a few benefits and drawbacks: Using "-omics" technologies, it is possible to effectively treat complex diseases like cancer that have complex genetic causes. In a single experiment, numerous genetic material, protein molecules, and other substances can be studied simultaneously. This enables the measurement of not only complete biological networks and well-known signaling pathways, but also novel processes and routes that are yet to be documented in a specific testing context. Without a doubt, it is the main advantage of modern technologies. One more important benefit is that it is possible to quantify the changes in cells brought about by intricate herbal blends. The reductionistic techniques of western science and medicine frequently fall short of providing a satisfactory investigation of the holistic approach of herbal treatment. Innovative network pharmacy techniques might provide fresh ways to holistic medicine.61 However, there are some drawbacks that need to be taken into account. Identifying causal mechanisms from background noise can be challenging when dealing with large volumes of generated data. Further validation tests are essential to validate the outcomes derived by "-omics" technology. Historically, network pharmacology has not conducted many time and concentration kinetics experiments because to the significant expenses involved. This carries the risk of missing the best drug concentrations and time periods to assess a pharmacological impact. Price reductions and other technological advancements could potentially mitigate this drawback. Another drawback is that less is known about many genes, making it challenging to understand how they manifest themselves in a functional setting.61

Genomics

Network pharmacology makes extensive use of genomics for tumor classification. For this goal, a novel and intriguing data source is somatic alteration screening. Hofree et al. grouped the somatic alteration patterns of the three main kinds of cancer (lung adeno carcinoma, ovarian and uterine carcinoma) listed in TCGA into substantial cancer subgroups that are biologically meaningful.62 Using the TCGA data on ovarian cancer, the AMARETTO algorithm was used. This not only found known driver gene mutations but also discovered novel cancer driver genes of relevance.60 Single nucleotide polymorphisms (SNP) bioinformatic analysis could help with the management of adverse medication responses in a specific individual populations along with speed up the process of developing new treatment plans.63,64 It is commonly known that SNPs are important for drug transporters and enzymes that metabolize drugs. One openly available resource on the internet is the Pharmacogenetics and Pharmacogenomics Knowledge Base (PharmGKB) that provides details on how genetic variability and medication response interact.65 A gene identification technique called GATHER was created to examine genome patterns. Its fundamental component is a statistical basis that measures the importance of functional relationships.66

Epigenomics

To investigate gene-environment interactions and treatment success, using a systems biology perspective employing novel demonstrating approaches seems vital given the multitude of the mechanisms of biology and the effects of epigenetic modifications upon the expression of tumor phenotypes. A wealth of data containing genes that are prognostically relevant have been produced by the examination of epigenetic modifications.48,67 Seven genes were shown to be conserved throughout all stages of lung adenocarcinoma by epigenetic subnetwork analysis, which was carried out using a systems biology technique to characterize epigenetic genes stage-by-stage.68 According to reports, the methylation status of the genes HOXD3, ARHGDIB, and AGAP2 affects cytoskeletal architecture and cell signaling in squamous cell lung cancer. Additionally, the patients' prognosis was predicted by the methylation state of these genes.69 Gnad et al. identified abnormal epigenetic regulation genomes in tumor using heterogeneous gene translation and connection networking studies. They discovered that EZH2 was the most noticeably overexpressed epigenetic regulator in tumor and was categorized as an oncogene.70 Toxicology also involves epigenomics. For example, certain epigenetic modifications brought about by the mycotoxin ochratoxin are linked to the toxin's carcinogenic potential.71 By identifying epigenetically dysregulated functional hotspots, a systems-based methodology has identified one important cancer inhibitor in endometrial carcinoma is heart and neural crest derivatives expressed 2 (HAND2).72

Transcriptomics

The research on silencing RNA (siRNA), messenger RNA (mRNA) and RNA that does not code using RNA sequencing, hybridization of microarrays, and other techniques have significantly advanced our knowledge of how chemotherapy works and how cancer progresses.58,73

Lapatinib-regulated putative miRNAs were found using prediction techniques including TargetScan, miRanda, and PITA. miRNA/mRNA pairings, which are essential for the mobility and viability of cells, were identified using the MiRNA and Genes Integrated Analysis (MAGIA) method.74 Furthermore, systems techniques have identified additional targets from the TCGA database that are drugable.10,75 In squamous cell lung cancer, two new miRNA species were found by the Bayesian network analysis of the KEGG lung tumor components: let-7a and miR-31. These miRNA species are tightly linked to the tumor suppressor gene CDKN2A and the IGF1R/RAS/MAPK pathway.69 Age-associated epigenetic alterations aggregated to cause the inhibition of expression of cancer growth factor-B and is a contributing factor to ovarian carcinogenesis, based on ovarian tumor transcriptomic profiling that is systems-based.76

Proteomics

Proteomic technologies represent a novel approach to illness detection and therapy response analysis. For reliable estimation of ligand-receptor and drug-target reactions, proteome and transcriptome data are frequently examined simultaneously. The use of linear and non-linear optimization models allows the relationship between biologic elements in networks to be closely examined for pertinent and important pathways.77,78 In these analyses, the regression model and correlation coefficient selection are crucial. The correlation among abundance of proteins and transcription is clarified by the ZIP line regression paradigm more thoroughly than the Poisson regression model. It also explains why a large proportion of proteins have zero abundance because of technological constraints.79 For additional analysis of proteins, biological mechanisms, and molecule activity, such as those carried out by DAVID, SWISS Uniprot (http://www.uniprot.org/faq/45) or PANTHER (Protein Analysis Through Evolutionary Relationships) assesses properties of the protein.80,81 Proteomics facilitates patient classification for customized care. The analysis of secreted proteins in clusters without supervision is the conceptual idea. This makes it possible to group patients according to a certain proteome profile that is associated with the stage of the tumor and/or the way the medications are working.82,83 Phospho-proteomics, which enables the determination of the various phosphorylation states of proteins, is another proteome application.84 Drug actions and signaling network dynamics are predicted by optimization techniques like the ordinary differential equation, but they do not provide treatment with several targets.85 Iadevaia et al. discovered medication combinations with optimal therapeutic outcomes by predicting how focusing on specific proteins affects the network as a whole using particle swarm optimization (PSO).86 Proteomic technologies, including pharmacoproteomics, are at the forefront of personalized medicine because gene transcript sequencing is ineffective in revealing signal transduction pathways and networks of interactions between proteins. Pharmacoproteomic techniques can be used to effectively treat metastatic malignancies in their advanced stages by predicting effective combinations of several targeted inhibitors. In regard to altered pathways of signaling in metastases located at various locations throughout the patient, these medications may be selected.87

Metabolomics

The study of how the genome controls the periodic production of cellular metabolites is known as metabolomics. The genome, namely the c-MYC gene, regulates cancer's imbalanced metabolic processes, including the Warburg effect of glycolysis.88 Thus, new directions for network analysis are made possible by cancer's genome-wide metabolomic models (GSMM).89,90 Strong hints regarding the interactions between metabolic medications are provided by this, enabling the recommendation of anti-cancer medication combinations with the fewest negative effects on healthy human cells.91 For the scientific community to effectively exploit metabolomics data, they should be readily accessible via public metabolomic databases. Hur et al. have developed an integrated transcriptome dataset and public metabolomics library to facilitate the creation of theories regarding potential ways in which chemical substances derived from organic sources work. AtMetExpress, SetupX, Data Resources Of Plant Metabolomics (DROP met), The Medicinal Plant Metabolomics Resource (MPMR), and Plant Metabolomics are further instances of metabolic databases.92 Although there is excitement about the technical potential of metabolomics, it is important to recognize the limitations of this technology.89 Systems biological analysis in Recon1 GSMM using the E flux method revealed that the relevant genes' up- and down-regulation was not well correlated with alterations in thirteen different tumor cell lines' metabolic processes.93 For metabolomics data to be used as instruments supporting cancer diagnosis and treatment, they must be carefully interpreted.

Network pharmacology's therapeutic implications

The inability to successfully transition single-targeted drug development from pre-clinical studies to clinical settings increases questions about the appropriateness of this strategy to single target drug discovery. It is difficult even to employ medications that act on specific targets that are part of strong biological networks since these targets are not fully understood or validated in patients.9 Because of this, networking pharmacology is becoming more and more important and receiving a lot of interest within the field of current pharmaceuticals development.94

Pleiotropic natural product-based strategies that target several proteins and pathways in networks linked to cancer could be promising. Throughout the world, traditional herbal treatments are crucial to maintaining health. Natural remedies are regarded as a viable source of novel effective compounds for developing medicines due to their low toxicity, biological activity and structural variety, attracting interest from all over the world to this field. One disease, one medicine, and one target is becoming less common in favor of "one disease, one drug, multiple targets."95,96

The study of network pharmacology examines where and how to target in order to reduce disease traits like cancer growth. This helps develop treatments that don't cause side effects or drug resistance. The advantages of networking pharmacology are amplifying within medicine development, particularly in polypharmacology and repurposing drugs.97 The use of polypharmacology techniques aids in the discovery of substances with numerous modes of action and the ability to act concurrently through many targets.98 Compounds with polypharmacological qualities could result in the discovery of innovative medicinal applications for already-approved medications. This procedure is commonly referred to as "drug repurposing."99,100 By combining network biology and polypharmacology, it is possible to investigate interactions between ligands and targets, that increases the range of targets that can be influenced by drugs and facilitates drug screening and development.7,101  Structure-based medication creation will be made easier by network-based methods, predict adverse drug side effects, and anticipate how pharmaceuticals will bind to biomolecules and signaling pathways.102

Compared to ideas for creating single-specific medications over individual targets connected to tumor, network pharmacology offers fresh directions towards the creation of drugs which may be far more encouraging.7,103  Drug resistance is often associated with such treatments, despite the fact that this concept gained attention due to the possibility of having medications with less negative effects on normal tissues and a higher specificity towards cancer. Such a drug's ineffectiveness could be quickly caused by mutations in the appropriate target protein. On the other hand, if mutations occur in one of the targets, the efficacy of medications that target many targets remains unaffected. The majority of natural compounds hit numerous targets instead of just one in order to exhibit their bioactivity. As multitarget specific chemicals increase an organism's chances of survival in the fight for existence, it is possible that selective pressure during the evolution of life encouraged the formation of these compounds.

Network pharmacology could offer special chances for methodical target identification and ways to target them using natural molecules that are multi-target specific.104 In complex protein networks, highly linked nodes are more susceptible than other nodes to pharmacological blockage of the entire network.105 Drugs, however, cannot inhibit every protein node in a network. In a specific network system, only 15% among the entire protein connections are believed to be druggable. Several approaches can be taken into consideration in order to produce logical phytotherapies based on network information:

  1. If the bioactive chemical elements of a plant or herbal mixture are known, then they can be taken into consideration. This strategy is primarily experience-based and is based on their application in traditional treatments. Herbal formulations resemble multidrug combination therapy including synthetic medications and polypharmacology in several aspects.106
  2. Using selective polypharmacological techniques, single drugs and phytochemicals, respectively, has the potential to accomplish multi-target intended treatment.107,108
  3. Synthetic lethality is emphasized by recent ideas in network pharmacology.109 If linked to a cancer network, non-essential proteins in normal cells may become therapeutically relevant.110 Their concurrent suppression or elimination may lead to enhanced or even possibly complementary eradication of cancer cells.  Even when significant cancer-related targets are impacted, numerous single-gene or protein molecule failures show little to not any impact on the development of cancer.111

In cancer therapy, what makes perfect sense in an organism's normal physiology presents significant treatment challenges. Using phytochemicals or complex herbal mixes that target numerous targets in cancer networks, polypharmacology could be a conceptual answer to this challenge instead of focusing on eliminating individual disease-causing proteins. Green tea polyphenolics have several bioactivities against different diseases, including cancer, according to a network pharmacology approach. There were 200 human targets in total. The pathways of the pleiotropic effects of green tea polyphenolics on irritation, malignancy, diabetes, neurological disorders, heart disease, and muscle disorders were demonstrated in this study.112 Recently, the molecular effects of herbs on the human body were studied using an innovative unified infrastructure of Herbal Medicine Systems Pharmacology (HMSP). This platform facilitates processing of scientific information for the relationship between species and disorders associated with herbs, target fishing, medication target development, and ADME prediction.113

The use of network pharmacology techniques will support customized medicine treatment plans. Through TCGA's genome-wide profiling of different cancer types, signals that could potentially forecast the clinical result following adjuvant chemotherapy were found. Thus, to control metastatic colorectal cancer, doctors can tailor medication therapy according to the unique genome for every individual's cancer thanks to data from network pharmacology.114,115 Based on the genetic profiling of 482 cell lines and 1019 tumor samples, for a total of 4104 potential metabolic products have been anticipated. To confirm that these metabolites are suitable as biomarkers for therapy monitoring, more research is needed.116 Drug resistance may be overcome in part by using predictive biomarkers. Consequently, small compound antagonists of tyrosine kinase regarding non-small cellular pulmonary cancer possess a license to be used in clinical settings, marking a substantial advancement in customized healthcare.117,118  Targeted medication resistance can be predicted, clinically detected, and treated via metabolic profiling. Myeloid cell metabolism profiling has been used to study imatinib resistance, which may help find reliable biomarkers for the treatment of drug-resistant malignancies.119 The prognosis prediction of cancer patients following patient survival analysis is a significant area of study in the field of cancer research. Network pharmacology could be an effective method for forecasting patient survival durations. Through the examination of meta-dimensional "-omics" information from TCGA, a new integrated approach is used to calculate filtered survivability rates among patients with breast tumor.120 In addition, a network-based analytic tool was created using TCGA data to identify gene-gene interactions connected to the clinical outcome of patients with ovarian cancer.121

Revealing insights from network pharmacology

One of network pharmacology's greatest strengths is its ability to monitor pharmacological activities in a very comprehensive way, which presents the possibility of finding new pathways. However, there is a chance that many genes that are not causally connected to a drug's action will also surface as a result of the discovery of many up- or down-regulated genes. Differentiating this "background noise" from the signals that are mechanistically significant is the art.122

Network pharmacology is a potent instrument that can provide testable ideas regarding the ways in which two or more pharmaceuticals interact synergistically. An effective technique for determining natural preparations' antagonism and synergistic actions in the mammalian body could be to analyze RNA microarray data of cells and compare the quantity of the genes that have been disrupted by botanical infusions using its specific medicinal composition. In human T98G neuroglial cells, we investigated monitoring gene activity both within the presence as well as absence of the herbal compound ADAPT-232 and its components. Tyrosol, eleutheroside E, salidroside, schizandrin B, and triandrin are among the bioactive natural compounds found in ADAPT-232 which is a herbal blend consisting of Rhodiola rosea root, Schisandra chinensis berry, and Eleutherococcus senticosus root.123 It's interesting to note that the patterns of gene activity were impacted by the mixture of these individual phytochemicals. A collaborative connection between the chemicals throughout the blend was indicated by the activation of specific genes in combinations that the single drugs did not reveal to be unregulated. However, when the compounds were combined, there was an antagonistic interaction that resulted in the down-regulation of some genes, which was not observed when the compounds were treated separately. These findings suggest that the chemicals in a herbal mixture form a new entity with distinct pharmacological properties, such as enhanced pharmacological selectivity and reduced unwanted side effects, rather than just the sum of their parts.

The Preparation from Rhodiola rosea SHR-5 and some of its constituent parts (triandrin, tyrosol, and salidroside)124, in addition to the authentic extracts of Radix Eleutherococci (ESE), Herba Andrographidis (APE), along with their set composition Kan Jang (KJ), were shown to have comparable experiences.125 Employing datasets with Genes that were markedly up- and down-regulated were examined, and complex system studies of the consequences were performed, and predictions were made on the effects on cellular activities and illnesses.

Microarray investigations, however, do not offer definitive evidence that Mammalian physiologic responses following consumption of natural mixtures or its component parts are, in fact, caused by the genes activated by these substances.

These analyses, however, can offer insightful information about potential genes as well as future research directions and potential practical applications.

Conclusions and perspectives

The majority of single-targeted candidate medications rarely make it to the final stage of clinical trials, even with significant investments made in preclinical research. A small number of preclinical lead compounds—roughly ten thousand in number—have made their way into clinical practice over the last ten years. Single focused drug discovery was radically redesigned using network pharmacology. It might transform drug research and discovery in the future. A deeper understanding of the causes of medication failure in terms of toxicity, side effects, and clinical efficacy may also be possible with the use of network pharmacology. Numerous biomarkers for distinct cancer types are found through the molecular characterisation of cancer. Biomarkers shared by a variety of tumor types are very helpful as new diagnostic instruments.These biomarkers can be used in everyday therapy and diagnosis through the use of network-based algorithms. Martinez-Ledesma et al., for example, employed finding a multi-cancer indicator within 12 kinds of tumors that predicts individual survival results using a network-based clinical association (NCA) method.126 Approaches in network pharmacology could significantly reduce common medical procedure errors, for example receptive variability in general population and illness. Global populations exhibit heterogeneity, and genetic variations in pharmacologically significant genes that differ between geographic regions are noteworthy.127 Furthermore, The presence or absence of viable treatment goals characterizes tumor heterogeneity.128 There are presently very few therapy options available for diverse populations with diverse cancers.In order to challenge conventional wisdom on medication development and increase our understanding of how drugs work, it is imperative that new network pharmacology techniques be developed. When network pharmacology techniques are applied properly, they can help avoid the problems associated with cancer medication treatment and expedite the development of novel antitumor medications.

REFERENCES

  1. S. R. Chamberlin, A. Blucher, G. Wu et al., “Natural product target network reveals potential for cancer combination therapies,” Frontiers in Pharmacology, vol. 10, p. 557, 2019.
  2. V. K. Bhardwaj and R. Purohit, “A lesson for the maestro of the replication fork: targeting the protein-binding interface of proliferating cell nuclear antigen for anticancer therapy,” Journal of Cellular Biochemistry, vol. 123, no. 6, pp. 1091–1102, 2022.
  3. R. Chen, Z. Guan, X. Zhong, W. Zhang, and Y. Zhang, “Network pharmacology prediction: the possible mechanisms of cinobufotalin against osteosarcoma,” Computational and Mathematical Methods in Medicine, vol. 2022, Article ID 3197402, 9 pages, 2022.
  4. D. Shi, F. Khan, and R. Abagyan, “Extended multitarget pharmacology of anticancer drugs,” Journal of Chemical Information and Modeling, vol. 59, no. 6, 2019.
  5. M. N. Gupta, A. Alam, and S. E. Hasnain, “Protein promiscuity in drug discovery drug-repurposing and antibiotic resistance,” Biochimie, vol. 175, pp. 50–57, 2020.
  6. J.W. Scannell, A. Blanckley, H. Boldon, and B. Warrington. Diagnosing the decline in pharmaceutical R&D efficiency. Nat Rev Drug Discov. 11:191?200 (2012).
  7. A.L. Hopkins. Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol. 4:682?690 (2008).
  8. L. Xie, L. Xie, S.L. Kinnings, and P.E. Bourne. Novel computational approaches to polypharmacology as a means to define responses to individual drugs. Annu Rev Pharmacol Toxicol. 52:361?379 (2012).
  9. A.S. Azmi and R.M. Mohammad. Rectifying cancer drug discovery through network pharmacology. Future Med Chem. 6:529?539 (2014).
  10. A.L. Barabasi, N. Gulbahce, and J. Loscalzo. Network medicine: a network?based approach to human disease. Nat Rev Genet. 12:56?68 (2011).
  11. L.M. Espinoza?Fonseca. The benefits of the multi?target approach in drug design and discovery. Bioorg Med Chem. 14:896?897 (2006).
  12. S.K. Mencherand L.G. Wang. Promiscuous drugs compared to selective drugs (promiscuity can be a virtue). BMC Clin Pharmacol. 5:3 (2005).
  13. N. Chandraand J. Padiadpu. Network approaches to drug discovery. Expert Opin Drug Discov. 8:7?20 (2013).
  14. A.S. Azmi, Z. Wang, P.A. Philip, R.M. Mohammad, and F.H. Sarkar. Proof of concept: network and systems biology approaches aid in the discovery of potent anticancer drug combinations. Mol Cancer Ther. 9:3137?3144 (2010).
  15. K.I. Goh, M.E. Cusick, D. Valle, B. Childs, M. Vidal, and A.L. Barabasi. The human disease network. Proc Natl Acad Sci U S A. 104:8685?8690 (2007).
  16. M. Rask?Andersen, M.S. Almen, and H.B. Schioth. Trends in the exploitation of novel drug targets. Nat Rev Drug Discov. 10:579?590 (2011).
  17. M. Kibble, N. Saarinen, J. Tang, K. Wennerberg, S. Makela, and T. Aittokallio. Network pharmacology applications to map the unexplored target space and therapeutic potential of natural products. Natural product reports. 32:1249?1266 (2015).
  18. G.B. Zhang, Q.Y. Li, Q.L. Chen, and S.B. Su. Network pharmacology: a new approach for chinese herbal medicine research. Evid Based Complement Alternat Med. 2013:621423 (2013).
  19. W. Kooti, K. Servatyari, M. Behzadifar et al., “Efective medicinal plant in cancer treatment, part 2: Review study,” Journal of evidence-based complementary and alternative medicine, vol. 22, no. 4, pp. 982–995, 2017.
  20. A. N. Alamgir, Terapeutic Use of Medicinal Plants and their Extracts, Springer International Publishing AG, New York, NY, USA, 2017.
  21. Noor, F.; Saleem, M.H.; Aslam, M.F.; Ahmad, A.; Aslam, S. Construction of miRNA-mRNA network for the identification of key biological markers and their associated pathways in IgA nephropathy by employing the integrated bioinformatics analysis. Saudi J. Biol. Sci. 2021, 28, 4938–4945.
  22. Noor, F.; Saleem, M.H.; Aslam, M.F.; Ahmad, A.; Aslam, S. Construction of miRNA-mRNA network for the identification of key biological markers and their associated pathways in IgA nephropathy by employing the integrated bioinformatics analysis. Saudi J. Biol. Sci. 2021, 28, 4938–4945.
  23. Bergendahl, L.T.; Gerasimavicius, L.; Miles, J.; Macdonald, L.; Wells, J.N.; Welburn, J.P.; Marsh, J.A. The role of protein complexes in human genetic disease. Protein Sci. 2019, 28, 1400–1411.
  24. Wang, X.; Gulbahce, N.; Yu, H. Network-based methods for human disease gene prediction. Brief. Funct. Genom. 2011, 10, 280–293.
  25. Schrattenholz, A.; Soskic, V. What does systems biology mean for drug development? Cur. Med. Chem. 2008, 15, 1520–1528.
  26. Anighoro, A.; Bajorath, J.; Rastelli, G. Polypharmacology: Challenges and opportunities in drug discovery: Miniperspective. J. Med. Chem. 2014, 57, 7874–7887.
  27. Peters, J.-U. Polypharmacology–foe or friend? J. Med. Chem. 2013, 56, 8955–8971.
  28. S Azmi, A. Adopting network pharmacology for cancer drug discovery. Cur. Drug Discov. Technol. 2013, 10, 95–105.
  29. St ?epnicki, P.; Kondej, M.; Kosz?a, O.; Zuk, J.; Kaczor, A.A. Multi-targeted drug design strategies for the treatment of schizophrenia. ? Expert Opin. Drug Discov. 2021, 16, 101–114.
  30. Achenbach, J.; Tiikkainen, P.; Franke, L.; Proschak, E. Computational tools for polypharmacology and repurposing. Futur. Med. Chem 2011, 3, 961–968.
  31. Fitzgerald, J.B.; Schoeberl, B.; Nielsen, U.B.; Sorger, P.K. Systems biology and combination therapy in the quest for clinical efficacy. Nat. Chem. Biol. 2006, 2, 458–466.
  32. Ji, H.F.; Li, X.J.; Zhang, H.Y. Natural products and drug discovery: Can thousands of years of ancient medical knowledge lead us to new and powerful drug combinations in the fight against cancer and dementia? EMBO Rep. 2009, 10, 194–200.
  33. Hopkins, A.L. Network pharmacology. Nat. Biotechnol. 2007, 25, 1110–1111.
  34. Medina-Franco, J.L.; Giulianotti, M.A.; Welmaker, G.S.; Houghten, R.A. Shifting from the single to the multitarget paradigm in drug discovery. Drug Discov. Today 2013, 18, 495–501.
  35. Chaudhari, R.; Tan, Z.; Huang, B.; Zhang, S. Computational polypharmacology: A new paradigm for drug discovery. Expert Opin. Drug Discov. 2017, 12, 279–291.
  36. Reddy, A.S.; Zhang, S. Polypharmacology: Drug discovery for the future. Expert Rev. Clin. Pharmacol. 2013, 6, 41–47.
  37. Cichonska, A.; Rousu, J.; Aittokallio, T. Identification of drug candidates and repurposing opportunities through compound–target interaction networks. Expert Opin. Drug Discov. 2015, 10, 1333–1345.
  38. Karuppasamy, R.; Veerappapillai, S.; Maiti, S.; Shin, W.-H.; Kihara, D. Current progress and future perspectives of polypharmacology: From the view of non-small cell lung cancer. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2021; pp. 84–91.
  39. Duarte, Y.; Márquez-Miranda, V.; Miossec, M.J.; González-Nilo, F. Integration of target discovery, drug discovery and drug delivery: A review on computational strategies. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2019, 11, e1554.
  40. Ekins, S.; Mestres, J.; Testa, B. In silico pharmacology for drug discovery: Methods for virtual ligand screening and profiling. Br. J. Pharmacol. 2007, 152, 9–20.
  41. Ekins, S.; Mestres, J.; Testa, B. In silico pharmacology for drug discovery: Applications to targets and beyond. Br. J. Pharmacol. 2007, 152, 21–37.
  42. R.A. Pache, A. Ceol, and P. Aloy. NetAligner??a network alignment server to compare complexes, pathways and whole interactomes. Nucleic Acids Res. 40:W157?161 (2012).
  43. A. Anighoro, J. Bajorath, and G. Rastelli. Polypharmacology: challenges and opportunities in drug discovery. J Med Chem. 57:7874?7887 (2014).
  44. M. Rask?Andersen, J. Zhang, D. Fabbro, and H.B. Schioth. Advances in kinase targeting: current clinical use and clinical trials. Trends Pharmacol Sci. 35:604?620 (2014).
  45. A.S. Azmi. Network pharmacology for cancer drug discovery: are we there yet? Future Med Chem. 4:939?941 (2012).
  46. E.E. Schadt. Molecular networks as sensors and drivers of common human diseases. Nature. 461:218?223 (2009).
  47. N. Cancer Genome Atlas Research. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 507:315?322 (2014).
  48. N. Cancer Genome Atlas Research. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202?209 (2014).
  49. C.F. Davis, C.J. Ricketts, M. Wang, L. Yang, A.D. Cherniack, H. Shen, C. Buhay, H. Kang, S.C. Kim, C.C. Fahey, K.E. Hacker, G. Bhanot, D.A. Gordenin, A. Chu, P.H. Gunaratne, M. Biehl, S. Seth, B.A. Kaipparettu, C.A. Bristow, L.A. Donehower, E.M. Wallen, A.B. Smith, S.K. Tickoo, P. Tamboli, V. Reuter, L.S. Schmidt, J.J. Hsieh, T.K. Choueiri, A.A. Hakimi, N. Cancer Genome Atlas Research, L. Chin, M. Meyerson, R. Kucherlapati, W.Y. Park, A.G. Robertson, P.W. Laird, E.P. Henske, D.J. Kwiatkowski, P.J. Park, M. Morgan, B. Shuch, D. Muzny, D.A. Wheeler, W.M. Linehan, R.A. Gibbs, W.K. Rathmell, and C.J. Creighton. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell. 26:319?330 (2014).
  50. N. Cancer Genome Atlas Research, C. Kandoth, N. Schultz, A.D. Cherniack, R. Akbani, Y. Liu, H. Shen, A.G. Robertson, I. Pashtan, R. Shen, C.C. Benz, C. Yau, P.W. Laird, L. Ding, W. Zhang, G.B. Mills, R. Kucherlapati, E.R. Mardis, and D.A. Levine. Integrated genomic characterization of endometrial carcinoma. Nature. 497:67?73 (2013).
  51. N. Cancer Genome Atlas Research. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 368:2059?2074 (2013).
  52. N. Cancer Genome Atlas. Comprehensive molecular portraits of human breast tumours. Nature. 490:61?70 (2012).
  53. N. Cancer Genome Atlas Research. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 489:519?525 (2012).
  54. N. Cancer Genome Atlas. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330?337 (2012).
  55. N. Cancer Genome Atlas Research. Integrated genomic analyses of ovarian carcinoma. Nature. 474:609?615 (2011).
  56. C.W. Brennan, R.G. Verhaak, A. McKenna, B. Campos, H. Noushmehr, S.R. Salama, S. Zheng, D. Chakravarty, J.Z. Sanborn, S.H. Berman, R. Beroukhim, B. Bernard, C.J. Wu, G. Genovese, I. Shmulevich, J. Barnholtz?Sloan, L. Zou, R. Vegesna, S.A. Shukla, G. Ciriello, W.K. Yung, W. Zhang, C. Sougnez, T. Mikkelsen, K. Aldape, D.D. Bigner, E.G. Van Meir, M. Prados, A. Sloan, K.L. Black, J. Eschbacher, G. Finocchiaro, W. Friedman, D.W. Andrews, A. Guha, M. Iacocca, B.P. O'Neill, G. Foltz, J. Myers, D.J. Weisenberger, R. Penny, R. Kucherlapati, C.M. Perou, D.N. Hayes, R. Gibbs, M. Marra, G.B. Mills, E. Lander, P. Spellman, R. Wilson, C. Sander, J. Weinstein, M. Meyerson, S. Gabriel, P.W. Laird, D. Haussler, G. Getz, L. Chin, and T.R. Network. The somatic genomic landscape of glioblastoma. Cell. 155:462?477 (2013).
  57. K.A. Hoadley, C. Yau, D.M. Wolf, A.D. Cherniack, D. Tamborero, S. Ng, M.D. Leiserson, B. Niu, M.D. McLellan, V. Uzunangelov, J. Zhang, C. Kandoth, R. Akbani, H. Shen, L. Omberg, A. Chu, A.A. Margolin, L.J. Van't Veer, N. Lopez?Bigas, P.W. Laird, B.J. Raphael, L. Ding, A.G. Robertson, L.A. Byers, G.B. Mills, J.N. Weinstein, C. Van Waes, Z. Chen, E.A. Collisson, N. Cancer Genome Atlas Research, C.C. Benz, C.M. Perou, and J.M. Stuart. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell. 158:929?944 (2014).
  58. X. Tian, X. Wang, and J. Chen. Network?constrained group lasso for high?dimensional multinomial classification with application to cancer subtype prediction. Cancer Inform. 13:25? 33 (2014).
  59. W. Zhang, Y.W. Wan, G.I. Allen, K. Pang, M.L. Anderson, and Z. Liu. Molecular pathway identification using biological network?regularized logistic models. BMC Genomics. 14 Suppl 8:S7 (2013).
  60. O. Gevaert, V. Villalobos, B.I. Sikic, and S.K. Plevritis. Identification of ovarian cancer driver genes by using module network integration of multi?omics data. Interface Focus. 3:20130013 (2013).
  61. Poornima, Paramasivan, et al. "Network pharmacology of cancer: From understanding of complex interactomes to the design of multi-target specific therapeutics from nature." Pharmacological Research 111 (2016): 290-302.
  62. M. Hofree, J.P. Shen, H. Carter, A. Gross, and T. Ideker. Network?based stratification of tumor mutations. Nat Methods. 10:1108?1115 (2013).
  63. S. Marsh. Cancer pharmacogenetics. Methods Mol Biol. 448:437?446 (2008).
  64. T. Efferth and M. Volm. Pharmacogenetics for individualized cancer chemotherapy. Pharmacol Ther. 107:155?176 (2005).
  65. K.M. Giacomini, C.M. Brett, R.B. Altman, N.L. Benowitz, M.E. Dolan, D.A. Flockhart, J.A. Johnson, D.F. Hayes, T. Klein, R.M. Krauss, D.L. Kroetz, H.L. McLeod, A.T. Nguyen, M.J. Ratain, M.V. Relling, V. Reus, D.M. Roden, C.A. Schaefer, A.R. Shuldiner, T. Skaar, K. Tantisira, R.F. Tyndale, L. Wang, R.M. Weinshilboum, S.T. Weiss, I. Zineh, and N. Pharmacogenetics Research. The pharmacogenetics research network: from SNP discovery to clinical drug response. Clin Pharmacol Ther. 81:328?345 (2007).
  66.  J.T. Changand J.R. Nevins. GATHER: a systems approach to interpreting genomic signatures. Bioinformatics. 22:2926?2933 (2006).
  67. D.J. Weisenberger. Characterizing DNA methylation alterations from The Cancer Genome Atlas. J Clin Invest. 124:17?23 (2014).
  68. M.P. Pradhan, A. Desai, and M.J. Palakal. Systems biology approach to stage?wise characterization of epigenetic genes in lung adenocarcinoma. BMC Syst Biol. 7:141 (2013).
  69. T. Huang, J. Yang, and Y.D. Cai. Novel candidate key drivers in the integrative network of genes, microRNAs, methylations, and copy number variations in squamous cell lung carcinoma. Biomed Res Int. 2015:358125 (2015).
  70. F. Gnad, S. Doll, G. Manning, D. Arnott, and Z. Zhang. Bioinformatics analysis of thousands of TCGA tumors to determine the involvement of epigenetic regulators in human cancer. BMC Genomics. 16 Suppl 8:S5 (2015).
  71. M. Marin?Kuan, C. Cavin, T. Delatour, and B. Schilter. Ochratoxin A carcinogenicity involves a complex network of epigenetic mechanisms. Toxicon. 52:195?202 (2008).
  72. A. Jones, A.E. Teschendorff, Q. Li, J.D. Hayward, A. Kannan, T. Mould, J. West, M. Zikan, D. Cibula, H. Fiegl, S.H. Lee, E. Wik, R. Hadwin, R. Arora, C. Lemech, H. Turunen, P. Pakarinen, I.J. Jacobs, H.B. Salvesen, M.K. Bagchi, I.C. Bagchi, and M. Widschwendter. Role of DNA methylation and epigenetic silencing of HAND2 in endometrial cancer development. PLoS medicine. 10:e1001551 (2013).
  73. J.N. Fisher, M. Terao, M. Fratelli, M. Kurosaki, G. Paroni, A. Zanetti, M. Gianni, M. Bolis, M. Lupi, A. Tsykin, G.J. Goodall, and E. Garattini. MicroRNA networks regulated by all?trans retinoic acid and Lapatinib control the growth, survival and motility of breast cancer cells. Oncotarget. 6:13176?13200 (2015).
  74. S.A. Knaack, A.F. Siahpirani, and S. Roy. A pan?cancer modular regulatory network analysis to identify common and cancer?specific network components. Cancer Inform. 13:69?84 (2014).
  75. T. Mashima, M. Ushijima, M. Matsuura, S. Tsukahara, K. Kunimasa, A. Furuno, S. Saito, M. Kitamura, T. Soma?Nagae, H. Seimiya, S. Dan, T. Yamori, and A. Tomida. Comprehensive transcriptomic analysis of molecularly targeted drugs in cancer for target pathway evaluation. Cancer Sci. 106:909?920 (2015).
  76. N. Matsumura, Z. Huang, S. Mori, T. Baba, S. Fujii, I. Konishi, E.S. Iversen, A. Berchuck, and S.K. Murphy. Epigenetic suppression of the TGF?beta pathway revealed by transcriptome profiling in ovarian cancer. Genome research. 21:74?82 (2011).
  77. J. Wang, G. Wu, L. Chen, and W. Zhang. Integrated Analysis of Transcriptomic and Proteomic Datasets Reveals Information on Protein Expressivity and Factors Affecting Translational Efficiency. Methods Mol Biol (2015).
  78. S. Haiderand R. Pal. Integrated analysis of transcriptomic and proteomic data. Curr Genomics. 14:91?110 (2013).
  79. H. Naya, J.I. Urioste, Y.M. Chang, M. Rodrigues?Motta, R. Kremer, and D. Gianola. A comparison between Poisson and zero?inflated Poisson regression models with an application to number of black spots in Corriedale sheep. Genetics, selection, evolution : GSE. 40:379?394 (2008).
  80. P.D. Thomas, A. Kejariwal, M.J. Campbell, H. Mi, K. Diemer, N. Guo, I. Ladunga, B. Ulitsky? Lazareva, A. Muruganujan, S. Rabkin, J.A. Vandergriff, and O. Doremieux. PANTHER: a browsable database of gene products organized by biological function, using curated protein family and subfamily classification. Nucleic acids research. 31:334?341 (2003).
  81. W. Huang da, B.T. Sherman, and R.A. Lempicki. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols. 4:44?57 (2009).
  82. R. Akbani, P.K. Ng, H.M. Werner, M. Shahmoradgoli, F. Zhang, Z. Ju, W. Liu, J.Y. Yang, K. Yoshihara, J. Li, S. Ling, E.G. Seviour, P.T. Ram, J.D. Minna, L. Diao, P. Tong, J.V. Heymach, S.M. Hill, F. Dondelinger, N. Stadler, L.A. Byers, F. Meric?Bernstam, J.N. Weinstein, B.M. Broom, R.G. Verhaak, H. Liang, S. Mukherjee, Y. Lu, and G.B. Mills. A pan?cancer proteomic perspective on The Cancer Genome Atlas. Nat Commun. 5:3887 (2014).
  83. R. Akbani, P.K. Ng, H.M. Werner, M. Shahmoradgoli, F. Zhang, Z. Ju, W. Liu, J.Y. Yang, K. Yoshihara, J. Li, S. Ling, E.G. Seviour, P.T. Ram, J.D. Minna, L. Diao, P. Tong, J.V. Heymach, S.M. Hill, F. Dondelinger, N. Stadler, L.A. Byers, F. Meric?Bernstam, J.N. Weinstein, B.M. Broom, R.G. Verhaak, H. Liang, S. Mukherjee, Y. Lu, and G.B. Mills. Corrigendum: A pan?cancer proteomic perspective on The Cancer Genome Atlas. Nat Commun. 6:4852 (2015).
  84. J. Reimand, O. Wagih, and G.D. Bader. The mutational landscape of phosphorylation signaling in cancer. Sci Rep. 3:2651 (2013).
  85. N. Barkaiand S. Leibler. Robustness in simple biochemical networks. Nature. 387:913?917 (1997).
  86. S. Iadevaia, Y. Lu, F.C. Morales, G.B. Mills, and P.T. Ram. Identification of optimal drug combinations targeting cellular networks: integrating phospho?proteomics and computational network analysis. Cancer Res. 70:6704?6714 (2010).
  87. J.D. Wulfkuhle, K.H. Edmiston, L.A. Liotta, and E.F. Petricoin, 3rd. Technology insight: pharmacoproteomics for cancer??promises of patient?tailored medicine using protein microarrays. Nature clinical practice Oncology. 3:256?268 (2006).
  88. C.V. Dang, K.A. O'Donnell, K.I. Zeller, T. Nguyen, R.C. Osthus, and F. Li. The c?Myc target gene network. Semin Cancer Biol. 16:253?264 (2006).
  89. K. Yizhak, B. Chaneton, E. Gottlieb, and E. Ruppin. Modeling cancer metabolism on a genome scale. Mol Syst Biol. 11:817 (2015).
  90. N. Pornputtapong, I. Nookaew, and J. Nielsen. Human metabolic atlas: an online resource for human metabolism. Database (Oxford). 2015:bav068 (2015).
  91. L. Li, X. Zhou, W.K. Ching, and P. Wang. Predicting enzyme targets for cancer drugs by profiling human metabolic reactions in NCI?60 cell lines. BMC bioinformatics. 11:501 (2010).
  92. M. Hur, A.A. Campbell, M. Almeida?de?Macedo, L. Li, N. Ransom, A. Jose, M. Crispin, B.J. Nikolau, and E.S. Wurtele. A global approach to analysis and interpretation of metabolic data for plant natural product discovery. Natural product reports. 30:565?583 (2013).
  93. Y. Asgari, Z. Zabihinpour, A. Salehzadeh?Yazdi, F. Schreiber, and A. Masoudi?Nejad. Alterations in cancer cell metabolism: the Warburg effect and metabolic adaptation. Genomics. 105:275?281 (2015).
  94. J. Gertsch. Botanical drugs, synergy, and network pharmacology: forth and back to intelligent mixtures. Planta Med. 77:1086?1098 (2011).
  95. X. Xu. New concepts and approaches for drug discovery based on traditional Chinese medicine. Drug Discov Today Technol. 3:247?253 (2006).
  96. F. Cheung. TCM: Made in China. Nature. 480:S82?83 (2011).
  97. J. Tangand T. Aittokallio. Network pharmacology strategies toward multi?target anticancer therapies: from computational models to experimental design principles. Curr Pharm Des. 20:23?36 (2014).
  98. A.D. Boranand R. Iyengar. Systems approaches to polypharmacology and drug discovery. Curr Opin Drug Discov Devel. 13:297?309 (2010).
  99. C. Chakraborty, C.G. Doss, L. Chen, and H. Zhu. Evaluating protein?protein interaction (PPI) networks for diseases pathway, target discovery, and drug?design using 'in silico pharmacology'. Curr Protein Pept Sci. 15:561?571 (2014).
  100. S. Ekins, J. Mestres, and B. Testa. In silico pharmacology for drug discovery: methods for virtual ligand screening and profiling. Br J Pharmacol. 152:9?20 (2007).
  101. J.K. Morrow, L. Tian, and S. Zhang. Molecular networks in drug discovery. Crit Rev Biomed Eng. 38:143?156 (2010).
  102. H.B. Engin, A. Gursoy, R. Nussinov, and O. Keskin. Network?based strategies can help mono? and poly?pharmacology drug discovery: a systems biology view. Curr Pharm Des. 20:1201?1207 (2014).
  103. A.S. Azmi AS. Adopting network pharmacology for cancer drug discovery. Curr Drug Discov Technol. 10:95?105 (2013).
  104. T. Korcsmáros, M.S. Szalay, C. Böde, I.A. Kovács, and P. Csermely. How to design multi?target drugs. Expert Opin Drug Discov. 2:799?808 (2007).
  105. H. Jeong, S.P. Mason, A.L. Barabási, and Z.N. Oltvai. Lethality and centrality in protein networks. Nature. 411:41?2 (2001).
  106. C. Hao da and P.G. Xiao. Network pharmacology: a Rosetta Stone for traditional Chinese medicine. Drug Dev Res. 75:299?312 (2014).
  107. R. Morphy, C. Kay and Z. Rankovic. From magic bullets to designed multiple ligands. Drug Discov Today. 9:641?51 (2004).
  108. A.L. Hopkins, J.S. Mason, J.P. Overington. Can we rationally design promiscuous drugs? Curr Opin Struct Biol. 16:127?36 (2006).
  109. R.A. Jackson and E.S. Chen. Synthetic lethal approaches for assessing combinatorial efficacy of chemotherapeutic drugs. Pharmacol Ther. pii: S0163?7258(16)00015?2 (2016).
  110. Z. Wang, J. Li, R. Dang, L. Liang, and J. Lin. PhIN: A Protein Pharmacology Interaction Network Database. CPT Pharmacometrics Syst Pharmacol. 4:e00025 (2015).
  111. H. Kitano Towards a theory of biological robustness. Mol Syst Biol. 3:137 (2007).
  112. S. Zhang, L. Shan, Q. Li, X. Wang, S. Li, Y. Zhang, J. Fu, X. Liu, H. Li, and W. Zhang. Systematic Analysis of the Multiple Bioactivities of Green Tea through a Network Pharmacology Approach. Evid Based Complement Alternat Med. 2014:512081 (2014).
  113. F. Luo, J. Gu, L. Chen, and X. Xu. Systems pharmacology strategies for anticancer drug discovery based on natural products. Mol Biosyst. 10:1912?1917 (2014).
  114. A. Wongand B.B. Ma. Personalizing therapy for colorectal cancer. Clin Gastroenterol Hepatol. 12:139?144 (2014).
  115. L.K. Teh, S. Hamzah, H. Hashim, Z. Bannur, Z.A. Zakaria, Z. Hasbullani, J.K. Shia, H. Fijeraid, A. Md Nor, M. Zailani, P. Ramasamy, H. Ngow, S. Sood, and M.Z. Salleh. Potential of dihydropyrimidine dehydrogenase genotypes in personalizing 5?fluorouracil therapy among colorectal cancer patients. Ther Drug Monit. 35:624?630 (2013).
  116. B.A. Aksoy, E. Demir, O. Babur, W. Wang, X. Jing, N. Schultz, and C. Sander. Prediction of individualized therapeutic vulnerabilities in cancer from genomic profiles. Bioinformatics. 30:2051?2059 (2014).
  117. D. Hanahan. Rethinking the war on cancer. Lancet. 383:558?563 (2014).
  118. R. Roskoski, Jr. The ErbB/HER family of protein?tyrosine kinases and cancer. Pharmacological research. 79:34?74 (2014).
  119. N. Serkovaand L.G. Boros. Detection of resistance to imatinib by metabolic profiling: clinical and drug development implications. Am J Pharmacogenomics. 5:293?302 (2005).
  120. D. Kim, R. Li, S.M. Dudek, and M.D. Ritchie. Predicting censored survival data based on the interactions between meta?dimensional omics data in breast cancer. J Biomed Inform. 56:220? 228 (2015).
  121. H.H. Jeong, S. Leem, K. Wee, and K.A. Sohn. Integrative network analysis for survival?associated gene?gene interactions across multiple genomic profiles in ovarian cancer. J Ovarian Res. 8:42 (2015).
  122. B. Wiench, T. Eichhorn, M. Paulsen, and T. Efferth. Shikonin directly targets mitochondria and causes mitochondrial dysfunction in cancer cells. Evid Based Complement Alternat Med. 2012:726025 (2012).
  123. A. Panossian, R. Hamm, O. Kadioglu, G. Wikman, and T. Efferth. Synergy and antagonism of active constituents of ADAPT?232 on transcriptional level of metabolic regulation of isolated neuroglial cells. Front Neurosci. 7:16 (2013).
  124. A. Panossian, R. Hamm, G. Wikman, and T. Efferth. Mechanism of action of Rhodiola, salidroside, tyrosol and triandrin in isolated neuroglial cells: an interactive pathway analysis of the downstream effects using RNA microarray data. Phytomedicine 21:1325?48 (2014).  
  125. A. Panossian, E.J. Seo, G. Wikman, and T. Efferth. Synergy assessment of fixed combinations of Herba Andrographidis and Radix Eleutherococci extracts by transcriptome?wide microarray profiling. Phytomedicine 22:981?92 (2015).  
  126. E. Martinez?Ledesma, R.G. Verhaak, and V. Trevino. Identification of a multi?cancer gene expression biomarker for cancer clinical outcomes using a network?based algorithm. Sci Rep. 5:11966 (2015).
  127. G. Suarez?Kurtz, D.D. Vargens, A.B. Santoro, M.H. Hutz, M.E. de Moraes, S.D. Pena, A. Ribeiro? dos?Santos, M.A. Romano?Silva, and C.J. Struchiner. Global pharmacogenomics: distribution of CYP3A5 polymorphisms and phenotypes in the Brazilian population. PLoS One. 9:e83472 (2014).
  128. M. Radovich, S.E. Clare, R. Atale, I. Pardo, B.A. Hancock, J.P. Solzak, N. Kassem, T. Mathieson, A.M. Storniolo, C. Rufenbarger, H.A. Lillemoe, R.J. Blosser, M.R. Choi, C.A. Sauder, D. Doxey, J.E. Henry, E.E. Hilligoss, O. Sakarya, F.C. Hyland, M. Hickenbotham, J. Zhu, J. Glasscock, S. Badve, M. Ivan, Y. Liu, G.W. Sledge, and B.P. Schneider. Characterizing the heterogeneity of triple?negative breast cancers using microdissected normal ductal epithelium and RNA?sequencing. Breast cancer research and treatment. 143:57?68 (2014)

Reference

  1. S. R. Chamberlin, A. Blucher, G. Wu et al., “Natural product target network reveals potential for cancer combination therapies,” Frontiers in Pharmacology, vol. 10, p. 557, 2019.
  2. V. K. Bhardwaj and R. Purohit, “A lesson for the maestro of the replication fork: targeting the protein-binding interface of proliferating cell nuclear antigen for anticancer therapy,” Journal of Cellular Biochemistry, vol. 123, no. 6, pp. 1091–1102, 2022.
  3. R. Chen, Z. Guan, X. Zhong, W. Zhang, and Y. Zhang, “Network pharmacology prediction: the possible mechanisms of cinobufotalin against osteosarcoma,” Computational and Mathematical Methods in Medicine, vol. 2022, Article ID 3197402, 9 pages, 2022.
  4. D. Shi, F. Khan, and R. Abagyan, “Extended multitarget pharmacology of anticancer drugs,” Journal of Chemical Information and Modeling, vol. 59, no. 6, 2019.
  5. M. N. Gupta, A. Alam, and S. E. Hasnain, “Protein promiscuity in drug discovery drug-repurposing and antibiotic resistance,” Biochimie, vol. 175, pp. 50–57, 2020.
  6. J.W. Scannell, A. Blanckley, H. Boldon, and B. Warrington. Diagnosing the decline in pharmaceutical R&D efficiency. Nat Rev Drug Discov. 11:191?200 (2012).
  7. A.L. Hopkins. Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol. 4:682?690 (2008).
  8. L. Xie, L. Xie, S.L. Kinnings, and P.E. Bourne. Novel computational approaches to polypharmacology as a means to define responses to individual drugs. Annu Rev Pharmacol Toxicol. 52:361?379 (2012).
  9. A.S. Azmi and R.M. Mohammad. Rectifying cancer drug discovery through network pharmacology. Future Med Chem. 6:529?539 (2014).
  10. A.L. Barabasi, N. Gulbahce, and J. Loscalzo. Network medicine: a network?based approach to human disease. Nat Rev Genet. 12:56?68 (2011).
  11. L.M. Espinoza?Fonseca. The benefits of the multi?target approach in drug design and discovery. Bioorg Med Chem. 14:896?897 (2006).
  12. S.K. Mencherand L.G. Wang. Promiscuous drugs compared to selective drugs (promiscuity can be a virtue). BMC Clin Pharmacol. 5:3 (2005).
  13. N. Chandraand J. Padiadpu. Network approaches to drug discovery. Expert Opin Drug Discov. 8:7?20 (2013).
  14. A.S. Azmi, Z. Wang, P.A. Philip, R.M. Mohammad, and F.H. Sarkar. Proof of concept: network and systems biology approaches aid in the discovery of potent anticancer drug combinations. Mol Cancer Ther. 9:3137?3144 (2010).
  15. K.I. Goh, M.E. Cusick, D. Valle, B. Childs, M. Vidal, and A.L. Barabasi. The human disease network. Proc Natl Acad Sci U S A. 104:8685?8690 (2007).
  16. M. Rask?Andersen, M.S. Almen, and H.B. Schioth. Trends in the exploitation of novel drug targets. Nat Rev Drug Discov. 10:579?590 (2011).
  17. M. Kibble, N. Saarinen, J. Tang, K. Wennerberg, S. Makela, and T. Aittokallio. Network pharmacology applications to map the unexplored target space and therapeutic potential of natural products. Natural product reports. 32:1249?1266 (2015).
  18. G.B. Zhang, Q.Y. Li, Q.L. Chen, and S.B. Su. Network pharmacology: a new approach for chinese herbal medicine research. Evid Based Complement Alternat Med. 2013:621423 (2013).
  19. W. Kooti, K. Servatyari, M. Behzadifar et al., “Efective medicinal plant in cancer treatment, part 2: Review study,” Journal of evidence-based complementary and alternative medicine, vol. 22, no. 4, pp. 982–995, 2017.
  20. A. N. Alamgir, Terapeutic Use of Medicinal Plants and their Extracts, Springer International Publishing AG, New York, NY, USA, 2017.
  21. Noor, F.; Saleem, M.H.; Aslam, M.F.; Ahmad, A.; Aslam, S. Construction of miRNA-mRNA network for the identification of key biological markers and their associated pathways in IgA nephropathy by employing the integrated bioinformatics analysis. Saudi J. Biol. Sci. 2021, 28, 4938–4945.
  22. Noor, F.; Saleem, M.H.; Aslam, M.F.; Ahmad, A.; Aslam, S. Construction of miRNA-mRNA network for the identification of key biological markers and their associated pathways in IgA nephropathy by employing the integrated bioinformatics analysis. Saudi J. Biol. Sci. 2021, 28, 4938–4945.
  23. Bergendahl, L.T.; Gerasimavicius, L.; Miles, J.; Macdonald, L.; Wells, J.N.; Welburn, J.P.; Marsh, J.A. The role of protein complexes in human genetic disease. Protein Sci. 2019, 28, 1400–1411.
  24. Wang, X.; Gulbahce, N.; Yu, H. Network-based methods for human disease gene prediction. Brief. Funct. Genom. 2011, 10, 280–293.
  25. Schrattenholz, A.; Soskic, V. What does systems biology mean for drug development? Cur. Med. Chem. 2008, 15, 1520–1528.
  26. Anighoro, A.; Bajorath, J.; Rastelli, G. Polypharmacology: Challenges and opportunities in drug discovery: Miniperspective. J. Med. Chem. 2014, 57, 7874–7887.
  27. Peters, J.-U. Polypharmacology–foe or friend? J. Med. Chem. 2013, 56, 8955–8971.
  28. S Azmi, A. Adopting network pharmacology for cancer drug discovery. Cur. Drug Discov. Technol. 2013, 10, 95–105.
  29. St ?epnicki, P.; Kondej, M.; Kosz?a, O.; Zuk, J.; Kaczor, A.A. Multi-targeted drug design strategies for the treatment of schizophrenia. ? Expert Opin. Drug Discov. 2021, 16, 101–114.
  30. Achenbach, J.; Tiikkainen, P.; Franke, L.; Proschak, E. Computational tools for polypharmacology and repurposing. Futur. Med. Chem 2011, 3, 961–968.
  31. Fitzgerald, J.B.; Schoeberl, B.; Nielsen, U.B.; Sorger, P.K. Systems biology and combination therapy in the quest for clinical efficacy. Nat. Chem. Biol. 2006, 2, 458–466.
  32. Ji, H.F.; Li, X.J.; Zhang, H.Y. Natural products and drug discovery: Can thousands of years of ancient medical knowledge lead us to new and powerful drug combinations in the fight against cancer and dementia? EMBO Rep. 2009, 10, 194–200.
  33. Hopkins, A.L. Network pharmacology. Nat. Biotechnol. 2007, 25, 1110–1111.
  34. Medina-Franco, J.L.; Giulianotti, M.A.; Welmaker, G.S.; Houghten, R.A. Shifting from the single to the multitarget paradigm in drug discovery. Drug Discov. Today 2013, 18, 495–501.
  35. Chaudhari, R.; Tan, Z.; Huang, B.; Zhang, S. Computational polypharmacology: A new paradigm for drug discovery. Expert Opin. Drug Discov. 2017, 12, 279–291.
  36. Reddy, A.S.; Zhang, S. Polypharmacology: Drug discovery for the future. Expert Rev. Clin. Pharmacol. 2013, 6, 41–47.
  37. Cichonska, A.; Rousu, J.; Aittokallio, T. Identification of drug candidates and repurposing opportunities through compound–target interaction networks. Expert Opin. Drug Discov. 2015, 10, 1333–1345.
  38. Karuppasamy, R.; Veerappapillai, S.; Maiti, S.; Shin, W.-H.; Kihara, D. Current progress and future perspectives of polypharmacology: From the view of non-small cell lung cancer. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2021; pp. 84–91.
  39. Duarte, Y.; Márquez-Miranda, V.; Miossec, M.J.; González-Nilo, F. Integration of target discovery, drug discovery and drug delivery: A review on computational strategies. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2019, 11, e1554.
  40. Ekins, S.; Mestres, J.; Testa, B. In silico pharmacology for drug discovery: Methods for virtual ligand screening and profiling. Br. J. Pharmacol. 2007, 152, 9–20.
  41. Ekins, S.; Mestres, J.; Testa, B. In silico pharmacology for drug discovery: Applications to targets and beyond. Br. J. Pharmacol. 2007, 152, 21–37.
  42. R.A. Pache, A. Ceol, and P. Aloy. NetAligner??a network alignment server to compare complexes, pathways and whole interactomes. Nucleic Acids Res. 40:W157?161 (2012).
  43. A. Anighoro, J. Bajorath, and G. Rastelli. Polypharmacology: challenges and opportunities in drug discovery. J Med Chem. 57:7874?7887 (2014).
  44. M. Rask?Andersen, J. Zhang, D. Fabbro, and H.B. Schioth. Advances in kinase targeting: current clinical use and clinical trials. Trends Pharmacol Sci. 35:604?620 (2014).
  45. A.S. Azmi. Network pharmacology for cancer drug discovery: are we there yet? Future Med Chem. 4:939?941 (2012).
  46. E.E. Schadt. Molecular networks as sensors and drivers of common human diseases. Nature. 461:218?223 (2009).
  47. N. Cancer Genome Atlas Research. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 507:315?322 (2014).
  48. N. Cancer Genome Atlas Research. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202?209 (2014).
  49. C.F. Davis, C.J. Ricketts, M. Wang, L. Yang, A.D. Cherniack, H. Shen, C. Buhay, H. Kang, S.C. Kim, C.C. Fahey, K.E. Hacker, G. Bhanot, D.A. Gordenin, A. Chu, P.H. Gunaratne, M. Biehl, S. Seth, B.A. Kaipparettu, C.A. Bristow, L.A. Donehower, E.M. Wallen, A.B. Smith, S.K. Tickoo, P. Tamboli, V. Reuter, L.S. Schmidt, J.J. Hsieh, T.K. Choueiri, A.A. Hakimi, N. Cancer Genome Atlas Research, L. Chin, M. Meyerson, R. Kucherlapati, W.Y. Park, A.G. Robertson, P.W. Laird, E.P. Henske, D.J. Kwiatkowski, P.J. Park, M. Morgan, B. Shuch, D. Muzny, D.A. Wheeler, W.M. Linehan, R.A. Gibbs, W.K. Rathmell, and C.J. Creighton. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell. 26:319?330 (2014).
  50. N. Cancer Genome Atlas Research, C. Kandoth, N. Schultz, A.D. Cherniack, R. Akbani, Y. Liu, H. Shen, A.G. Robertson, I. Pashtan, R. Shen, C.C. Benz, C. Yau, P.W. Laird, L. Ding, W. Zhang, G.B. Mills, R. Kucherlapati, E.R. Mardis, and D.A. Levine. Integrated genomic characterization of endometrial carcinoma. Nature. 497:67?73 (2013).
  51. N. Cancer Genome Atlas Research. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 368:2059?2074 (2013).
  52. N. Cancer Genome Atlas. Comprehensive molecular portraits of human breast tumours. Nature. 490:61?70 (2012).
  53. N. Cancer Genome Atlas Research. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 489:519?525 (2012).
  54. N. Cancer Genome Atlas. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330?337 (2012).
  55. N. Cancer Genome Atlas Research. Integrated genomic analyses of ovarian carcinoma. Nature. 474:609?615 (2011).
  56. C.W. Brennan, R.G. Verhaak, A. McKenna, B. Campos, H. Noushmehr, S.R. Salama, S. Zheng, D. Chakravarty, J.Z. Sanborn, S.H. Berman, R. Beroukhim, B. Bernard, C.J. Wu, G. Genovese, I. Shmulevich, J. Barnholtz?Sloan, L. Zou, R. Vegesna, S.A. Shukla, G. Ciriello, W.K. Yung, W. Zhang, C. Sougnez, T. Mikkelsen, K. Aldape, D.D. Bigner, E.G. Van Meir, M. Prados, A. Sloan, K.L. Black, J. Eschbacher, G. Finocchiaro, W. Friedman, D.W. Andrews, A. Guha, M. Iacocca, B.P. O'Neill, G. Foltz, J. Myers, D.J. Weisenberger, R. Penny, R. Kucherlapati, C.M. Perou, D.N. Hayes, R. Gibbs, M. Marra, G.B. Mills, E. Lander, P. Spellman, R. Wilson, C. Sander, J. Weinstein, M. Meyerson, S. Gabriel, P.W. Laird, D. Haussler, G. Getz, L. Chin, and T.R. Network. The somatic genomic landscape of glioblastoma. Cell. 155:462?477 (2013).
  57. K.A. Hoadley, C. Yau, D.M. Wolf, A.D. Cherniack, D. Tamborero, S. Ng, M.D. Leiserson, B. Niu, M.D. McLellan, V. Uzunangelov, J. Zhang, C. Kandoth, R. Akbani, H. Shen, L. Omberg, A. Chu, A.A. Margolin, L.J. Van't Veer, N. Lopez?Bigas, P.W. Laird, B.J. Raphael, L. Ding, A.G. Robertson, L.A. Byers, G.B. Mills, J.N. Weinstein, C. Van Waes, Z. Chen, E.A. Collisson, N. Cancer Genome Atlas Research, C.C. Benz, C.M. Perou, and J.M. Stuart. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell. 158:929?944 (2014).
  58. X. Tian, X. Wang, and J. Chen. Network?constrained group lasso for high?dimensional multinomial classification with application to cancer subtype prediction. Cancer Inform. 13:25? 33 (2014).
  59. W. Zhang, Y.W. Wan, G.I. Allen, K. Pang, M.L. Anderson, and Z. Liu. Molecular pathway identification using biological network?regularized logistic models. BMC Genomics. 14 Suppl 8:S7 (2013).
  60. O. Gevaert, V. Villalobos, B.I. Sikic, and S.K. Plevritis. Identification of ovarian cancer driver genes by using module network integration of multi?omics data. Interface Focus. 3:20130013 (2013).
  61. Poornima, Paramasivan, et al. "Network pharmacology of cancer: From understanding of complex interactomes to the design of multi-target specific therapeutics from nature." Pharmacological Research 111 (2016): 290-302.
  62. M. Hofree, J.P. Shen, H. Carter, A. Gross, and T. Ideker. Network?based stratification of tumor mutations. Nat Methods. 10:1108?1115 (2013).
  63. S. Marsh. Cancer pharmacogenetics. Methods Mol Biol. 448:437?446 (2008).
  64. T. Efferth and M. Volm. Pharmacogenetics for individualized cancer chemotherapy. Pharmacol Ther. 107:155?176 (2005).
  65. K.M. Giacomini, C.M. Brett, R.B. Altman, N.L. Benowitz, M.E. Dolan, D.A. Flockhart, J.A. Johnson, D.F. Hayes, T. Klein, R.M. Krauss, D.L. Kroetz, H.L. McLeod, A.T. Nguyen, M.J. Ratain, M.V. Relling, V. Reus, D.M. Roden, C.A. Schaefer, A.R. Shuldiner, T. Skaar, K. Tantisira, R.F. Tyndale, L. Wang, R.M. Weinshilboum, S.T. Weiss, I. Zineh, and N. Pharmacogenetics Research. The pharmacogenetics research network: from SNP discovery to clinical drug response. Clin Pharmacol Ther. 81:328?345 (2007).
  66.  J.T. Changand J.R. Nevins. GATHER: a systems approach to interpreting genomic signatures. Bioinformatics. 22:2926?2933 (2006).
  67. D.J. Weisenberger. Characterizing DNA methylation alterations from The Cancer Genome Atlas. J Clin Invest. 124:17?23 (2014).
  68. M.P. Pradhan, A. Desai, and M.J. Palakal. Systems biology approach to stage?wise characterization of epigenetic genes in lung adenocarcinoma. BMC Syst Biol. 7:141 (2013).
  69. T. Huang, J. Yang, and Y.D. Cai. Novel candidate key drivers in the integrative network of genes, microRNAs, methylations, and copy number variations in squamous cell lung carcinoma. Biomed Res Int. 2015:358125 (2015).
  70. F. Gnad, S. Doll, G. Manning, D. Arnott, and Z. Zhang. Bioinformatics analysis of thousands of TCGA tumors to determine the involvement of epigenetic regulators in human cancer. BMC Genomics. 16 Suppl 8:S5 (2015).
  71. M. Marin?Kuan, C. Cavin, T. Delatour, and B. Schilter. Ochratoxin A carcinogenicity involves a complex network of epigenetic mechanisms. Toxicon. 52:195?202 (2008).
  72. A. Jones, A.E. Teschendorff, Q. Li, J.D. Hayward, A. Kannan, T. Mould, J. West, M. Zikan, D. Cibula, H. Fiegl, S.H. Lee, E. Wik, R. Hadwin, R. Arora, C. Lemech, H. Turunen, P. Pakarinen, I.J. Jacobs, H.B. Salvesen, M.K. Bagchi, I.C. Bagchi, and M. Widschwendter. Role of DNA methylation and epigenetic silencing of HAND2 in endometrial cancer development. PLoS medicine. 10:e1001551 (2013).
  73. J.N. Fisher, M. Terao, M. Fratelli, M. Kurosaki, G. Paroni, A. Zanetti, M. Gianni, M. Bolis, M. Lupi, A. Tsykin, G.J. Goodall, and E. Garattini. MicroRNA networks regulated by all?trans retinoic acid and Lapatinib control the growth, survival and motility of breast cancer cells. Oncotarget. 6:13176?13200 (2015).
  74. S.A. Knaack, A.F. Siahpirani, and S. Roy. A pan?cancer modular regulatory network analysis to identify common and cancer?specific network components. Cancer Inform. 13:69?84 (2014).
  75. T. Mashima, M. Ushijima, M. Matsuura, S. Tsukahara, K. Kunimasa, A. Furuno, S. Saito, M. Kitamura, T. Soma?Nagae, H. Seimiya, S. Dan, T. Yamori, and A. Tomida. Comprehensive transcriptomic analysis of molecularly targeted drugs in cancer for target pathway evaluation. Cancer Sci. 106:909?920 (2015).
  76. N. Matsumura, Z. Huang, S. Mori, T. Baba, S. Fujii, I. Konishi, E.S. Iversen, A. Berchuck, and S.K. Murphy. Epigenetic suppression of the TGF?beta pathway revealed by transcriptome profiling in ovarian cancer. Genome research. 21:74?82 (2011).
  77. J. Wang, G. Wu, L. Chen, and W. Zhang. Integrated Analysis of Transcriptomic and Proteomic Datasets Reveals Information on Protein Expressivity and Factors Affecting Translational Efficiency. Methods Mol Biol (2015).
  78. S. Haiderand R. Pal. Integrated analysis of transcriptomic and proteomic data. Curr Genomics. 14:91?110 (2013).
  79. H. Naya, J.I. Urioste, Y.M. Chang, M. Rodrigues?Motta, R. Kremer, and D. Gianola. A comparison between Poisson and zero?inflated Poisson regression models with an application to number of black spots in Corriedale sheep. Genetics, selection, evolution : GSE. 40:379?394 (2008).
  80. P.D. Thomas, A. Kejariwal, M.J. Campbell, H. Mi, K. Diemer, N. Guo, I. Ladunga, B. Ulitsky? Lazareva, A. Muruganujan, S. Rabkin, J.A. Vandergriff, and O. Doremieux. PANTHER: a browsable database of gene products organized by biological function, using curated protein family and subfamily classification. Nucleic acids research. 31:334?341 (2003).
  81. W. Huang da, B.T. Sherman, and R.A. Lempicki. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols. 4:44?57 (2009).
  82. R. Akbani, P.K. Ng, H.M. Werner, M. Shahmoradgoli, F. Zhang, Z. Ju, W. Liu, J.Y. Yang, K. Yoshihara, J. Li, S. Ling, E.G. Seviour, P.T. Ram, J.D. Minna, L. Diao, P. Tong, J.V. Heymach, S.M. Hill, F. Dondelinger, N. Stadler, L.A. Byers, F. Meric?Bernstam, J.N. Weinstein, B.M. Broom, R.G. Verhaak, H. Liang, S. Mukherjee, Y. Lu, and G.B. Mills. A pan?cancer proteomic perspective on The Cancer Genome Atlas. Nat Commun. 5:3887 (2014).
  83. R. Akbani, P.K. Ng, H.M. Werner, M. Shahmoradgoli, F. Zhang, Z. Ju, W. Liu, J.Y. Yang, K. Yoshihara, J. Li, S. Ling, E.G. Seviour, P.T. Ram, J.D. Minna, L. Diao, P. Tong, J.V. Heymach, S.M. Hill, F. Dondelinger, N. Stadler, L.A. Byers, F. Meric?Bernstam, J.N. Weinstein, B.M. Broom, R.G. Verhaak, H. Liang, S. Mukherjee, Y. Lu, and G.B. Mills. Corrigendum: A pan?cancer proteomic perspective on The Cancer Genome Atlas. Nat Commun. 6:4852 (2015).
  84. J. Reimand, O. Wagih, and G.D. Bader. The mutational landscape of phosphorylation signaling in cancer. Sci Rep. 3:2651 (2013).
  85. N. Barkaiand S. Leibler. Robustness in simple biochemical networks. Nature. 387:913?917 (1997).
  86. S. Iadevaia, Y. Lu, F.C. Morales, G.B. Mills, and P.T. Ram. Identification of optimal drug combinations targeting cellular networks: integrating phospho?proteomics and computational network analysis. Cancer Res. 70:6704?6714 (2010).
  87. J.D. Wulfkuhle, K.H. Edmiston, L.A. Liotta, and E.F. Petricoin, 3rd. Technology insight: pharmacoproteomics for cancer??promises of patient?tailored medicine using protein microarrays. Nature clinical practice Oncology. 3:256?268 (2006).
  88. C.V. Dang, K.A. O'Donnell, K.I. Zeller, T. Nguyen, R.C. Osthus, and F. Li. The c?Myc target gene network. Semin Cancer Biol. 16:253?264 (2006).
  89. K. Yizhak, B. Chaneton, E. Gottlieb, and E. Ruppin. Modeling cancer metabolism on a genome scale. Mol Syst Biol. 11:817 (2015).
  90. N. Pornputtapong, I. Nookaew, and J. Nielsen. Human metabolic atlas: an online resource for human metabolism. Database (Oxford). 2015:bav068 (2015).
  91. L. Li, X. Zhou, W.K. Ching, and P. Wang. Predicting enzyme targets for cancer drugs by profiling human metabolic reactions in NCI?60 cell lines. BMC bioinformatics. 11:501 (2010).
  92. M. Hur, A.A. Campbell, M. Almeida?de?Macedo, L. Li, N. Ransom, A. Jose, M. Crispin, B.J. Nikolau, and E.S. Wurtele. A global approach to analysis and interpretation of metabolic data for plant natural product discovery. Natural product reports. 30:565?583 (2013).
  93. Y. Asgari, Z. Zabihinpour, A. Salehzadeh?Yazdi, F. Schreiber, and A. Masoudi?Nejad. Alterations in cancer cell metabolism: the Warburg effect and metabolic adaptation. Genomics. 105:275?281 (2015).
  94. J. Gertsch. Botanical drugs, synergy, and network pharmacology: forth and back to intelligent mixtures. Planta Med. 77:1086?1098 (2011).
  95. X. Xu. New concepts and approaches for drug discovery based on traditional Chinese medicine. Drug Discov Today Technol. 3:247?253 (2006).
  96. F. Cheung. TCM: Made in China. Nature. 480:S82?83 (2011).
  97. J. Tangand T. Aittokallio. Network pharmacology strategies toward multi?target anticancer therapies: from computational models to experimental design principles. Curr Pharm Des. 20:23?36 (2014).
  98. A.D. Boranand R. Iyengar. Systems approaches to polypharmacology and drug discovery. Curr Opin Drug Discov Devel. 13:297?309 (2010).
  99. C. Chakraborty, C.G. Doss, L. Chen, and H. Zhu. Evaluating protein?protein interaction (PPI) networks for diseases pathway, target discovery, and drug?design using 'in silico pharmacology'. Curr Protein Pept Sci. 15:561?571 (2014).
  100. S. Ekins, J. Mestres, and B. Testa. In silico pharmacology for drug discovery: methods for virtual ligand screening and profiling. Br J Pharmacol. 152:9?20 (2007).
  101. J.K. Morrow, L. Tian, and S. Zhang. Molecular networks in drug discovery. Crit Rev Biomed Eng. 38:143?156 (2010).
  102. H.B. Engin, A. Gursoy, R. Nussinov, and O. Keskin. Network?based strategies can help mono? and poly?pharmacology drug discovery: a systems biology view. Curr Pharm Des. 20:1201?1207 (2014).
  103. A.S. Azmi AS. Adopting network pharmacology for cancer drug discovery. Curr Drug Discov Technol. 10:95?105 (2013).
  104. T. Korcsmáros, M.S. Szalay, C. Böde, I.A. Kovács, and P. Csermely. How to design multi?target drugs. Expert Opin Drug Discov. 2:799?808 (2007).
  105. H. Jeong, S.P. Mason, A.L. Barabási, and Z.N. Oltvai. Lethality and centrality in protein networks. Nature. 411:41?2 (2001).
  106. C. Hao da and P.G. Xiao. Network pharmacology: a Rosetta Stone for traditional Chinese medicine. Drug Dev Res. 75:299?312 (2014).
  107. R. Morphy, C. Kay and Z. Rankovic. From magic bullets to designed multiple ligands. Drug Discov Today. 9:641?51 (2004).
  108. A.L. Hopkins, J.S. Mason, J.P. Overington. Can we rationally design promiscuous drugs? Curr Opin Struct Biol. 16:127?36 (2006).
  109. R.A. Jackson and E.S. Chen. Synthetic lethal approaches for assessing combinatorial efficacy of chemotherapeutic drugs. Pharmacol Ther. pii: S0163?7258(16)00015?2 (2016).
  110. Z. Wang, J. Li, R. Dang, L. Liang, and J. Lin. PhIN: A Protein Pharmacology Interaction Network Database. CPT Pharmacometrics Syst Pharmacol. 4:e00025 (2015).
  111. H. Kitano Towards a theory of biological robustness. Mol Syst Biol. 3:137 (2007).
  112. S. Zhang, L. Shan, Q. Li, X. Wang, S. Li, Y. Zhang, J. Fu, X. Liu, H. Li, and W. Zhang. Systematic Analysis of the Multiple Bioactivities of Green Tea through a Network Pharmacology Approach. Evid Based Complement Alternat Med. 2014:512081 (2014).
  113. F. Luo, J. Gu, L. Chen, and X. Xu. Systems pharmacology strategies for anticancer drug discovery based on natural products. Mol Biosyst. 10:1912?1917 (2014).
  114. A. Wongand B.B. Ma. Personalizing therapy for colorectal cancer. Clin Gastroenterol Hepatol. 12:139?144 (2014).
  115. L.K. Teh, S. Hamzah, H. Hashim, Z. Bannur, Z.A. Zakaria, Z. Hasbullani, J.K. Shia, H. Fijeraid, A. Md Nor, M. Zailani, P. Ramasamy, H. Ngow, S. Sood, and M.Z. Salleh. Potential of dihydropyrimidine dehydrogenase genotypes in personalizing 5?fluorouracil therapy among colorectal cancer patients. Ther Drug Monit. 35:624?630 (2013).
  116. B.A. Aksoy, E. Demir, O. Babur, W. Wang, X. Jing, N. Schultz, and C. Sander. Prediction of individualized therapeutic vulnerabilities in cancer from genomic profiles. Bioinformatics. 30:2051?2059 (2014).
  117. D. Hanahan. Rethinking the war on cancer. Lancet. 383:558?563 (2014).
  118. R. Roskoski, Jr. The ErbB/HER family of protein?tyrosine kinases and cancer. Pharmacological research. 79:34?74 (2014).
  119. N. Serkovaand L.G. Boros. Detection of resistance to imatinib by metabolic profiling: clinical and drug development implications. Am J Pharmacogenomics. 5:293?302 (2005).
  120. D. Kim, R. Li, S.M. Dudek, and M.D. Ritchie. Predicting censored survival data based on the interactions between meta?dimensional omics data in breast cancer. J Biomed Inform. 56:220? 228 (2015).
  121. H.H. Jeong, S. Leem, K. Wee, and K.A. Sohn. Integrative network analysis for survival?associated gene?gene interactions across multiple genomic profiles in ovarian cancer. J Ovarian Res. 8:42 (2015).
  122. B. Wiench, T. Eichhorn, M. Paulsen, and T. Efferth. Shikonin directly targets mitochondria and causes mitochondrial dysfunction in cancer cells. Evid Based Complement Alternat Med. 2012:726025 (2012).
  123. A. Panossian, R. Hamm, O. Kadioglu, G. Wikman, and T. Efferth. Synergy and antagonism of active constituents of ADAPT?232 on transcriptional level of metabolic regulation of isolated neuroglial cells. Front Neurosci. 7:16 (2013).
  124. A. Panossian, R. Hamm, G. Wikman, and T. Efferth. Mechanism of action of Rhodiola, salidroside, tyrosol and triandrin in isolated neuroglial cells: an interactive pathway analysis of the downstream effects using RNA microarray data. Phytomedicine 21:1325?48 (2014).  
  125. A. Panossian, E.J. Seo, G. Wikman, and T. Efferth. Synergy assessment of fixed combinations of Herba Andrographidis and Radix Eleutherococci extracts by transcriptome?wide microarray profiling. Phytomedicine 22:981?92 (2015).  
  126. E. Martinez?Ledesma, R.G. Verhaak, and V. Trevino. Identification of a multi?cancer gene expression biomarker for cancer clinical outcomes using a network?based algorithm. Sci Rep. 5:11966 (2015).
  127. G. Suarez?Kurtz, D.D. Vargens, A.B. Santoro, M.H. Hutz, M.E. de Moraes, S.D. Pena, A. Ribeiro? dos?Santos, M.A. Romano?Silva, and C.J. Struchiner. Global pharmacogenomics: distribution of CYP3A5 polymorphisms and phenotypes in the Brazilian population. PLoS One. 9:e83472 (2014).
  128. M. Radovich, S.E. Clare, R. Atale, I. Pardo, B.A. Hancock, J.P. Solzak, N. Kassem, T. Mathieson, A.M. Storniolo, C. Rufenbarger, H.A. Lillemoe, R.J. Blosser, M.R. Choi, C.A. Sauder, D. Doxey, J.E. Henry, E.E. Hilligoss, O. Sakarya, F.C. Hyland, M. Hickenbotham, J. Zhu, J. Glasscock, S. Badve, M. Ivan, Y. Liu, G.W. Sledge, and B.P. Schneider. Characterizing the heterogeneity of triple?negative breast cancers using microdissected normal ductal epithelium and RNA?sequencing. Breast cancer research and treatment. 143:57?68 (2014)

Photo
Akanksha Chavan
Corresponding author

Ashokrao Mane College of Pharmacy, Peth Vadgaon, Kolhapur, Maharashtra, 416112, India

Photo
Prashant Kumbhar
Co-author

Ashokrao Mane College of Pharmacy, Peth Vadgaon, Kolhapur, Maharashtra, 416112, India

Photo
Dr. Sanganna Burli
Co-author

Ashokrao Mane College of Pharmacy, Peth Vadgaon, Kolhapur, Maharashtra, 416112, India

Photo
Vikas Dhole
Co-author

Ashokrao Mane College of Pharmacy, Peth Vadgaon, Kolhapur, Maharashtra, 416112, India

Akanksha Chavan*, Prashant Kumbhar, Dr. Sanganna Burli, Vikas Dhole, Unraveling Complex Interactomes: The Role of Network Pharmacology in Cancer Therapeutics, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 3, 1204-1220. https://doi.org/10.5281/zenodo.15019471

More related articles
An Overview of Pharmacological Models and Phytocon...
Anjali Patil, Vikas Dhole, Dr. Sanganna Burli, Prashant Kumbhar, ...
A Review On Role Of Artificial Intelligence In Dru...
Rama Brahma Reddy D, Malleswari K, Chetan M, Adarsh Babu B, Bhuva...
Revealing the Molecular Interactions: Investigatin...
Mayur Bagane, Rutuparna Karkare, Rajesh Jorgewad, Saee Thakur, M...
Management of Psoriasis: A Review of Current Treatments and Ethnobotanical Surve...
Samruddhi Raje , Prashant Kumbhar, Dr. Sanganna Burli, Vikas Dhole, ...
Pharmacological Screening Technique: Elisa Techhnique ...
Harshwardhan Yesugade , Omkar Shelake, Shubham Salunkhe, Manoj Bachche, J. R. Kambale, Dr. Nilesh Ch...
Aspirin Redesigned: A CADD-Guided Exploration of Optimized Aspirin Analogs for T...
Ranjeet V Pingale , Raj R Jagtap , Rohan B chavan , Pratik B lakade, ...
Related Articles
Artificial Intelligence in Pharmacy ...
Prabhakar kolhe, Radika Kotame, Omkar Phopse, Abhishek Dhanwate, Pravin Binnar, ...
Gastric Ulcers: Understanding Pathophysiology and Advances in Treatment...
Gaurav Sawant, Prashant Kumbhar, Dr. Sanganna Burli, Vikas Dhole, ...
Machine Learning Meets Medicine: AI’s Role in Drug Discovery and Development...
Aman Bhardwaj, Shalu Bharti, Ujwal, Vikas Kumar, Sidharath Kumar Gaud, ...
An Overview of Pharmacological Models and Phytoconstituents for The Anti-Inflamm...
Anjali Patil, Vikas Dhole, Dr. Sanganna Burli, Prashant Kumbhar, ...
More related articles
An Overview of Pharmacological Models and Phytoconstituents for The Anti-Inflamm...
Anjali Patil, Vikas Dhole, Dr. Sanganna Burli, Prashant Kumbhar, ...
A Review On Role Of Artificial Intelligence In Drug Discovery...
Rama Brahma Reddy D, Malleswari K, Chetan M, Adarsh Babu B, Bhuvan Chandra Durga Eswar J., ...
Revealing the Molecular Interactions: Investigating the Docking Studies of (N-(4...
Mayur Bagane, Rutuparna Karkare, Rajesh Jorgewad, Saee Thakur, Medha Petkar, Amruta Patil, Sneha K...
An Overview of Pharmacological Models and Phytoconstituents for The Anti-Inflamm...
Anjali Patil, Vikas Dhole, Dr. Sanganna Burli, Prashant Kumbhar, ...
A Review On Role Of Artificial Intelligence In Drug Discovery...
Rama Brahma Reddy D, Malleswari K, Chetan M, Adarsh Babu B, Bhuvan Chandra Durga Eswar J., ...
Revealing the Molecular Interactions: Investigating the Docking Studies of (N-(4...
Mayur Bagane, Rutuparna Karkare, Rajesh Jorgewad, Saee Thakur, Medha Petkar, Amruta Patil, Sneha K...