Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Pharmaceutics ; 15(6)2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37376121

ABSTRACT

In an era of unparalleled technical advancement, the pharmaceutical industry is struggling to transform data into increased research and development efficiency, and, as a corollary, new drugs for patients. Here, we briefly review some of the commonly discussed issues around this counterintuitive innovation crisis. Looking at both industry- and science-related factors, we posit that traditional preclinical research is front-loading the development pipeline with data and drug candidates that are unlikely to succeed in patients. Applying a first principles analysis, we highlight the critical culprits and provide suggestions as to how these issues can be rectified through the pursuit of a Human Data-driven Discovery (HD3) paradigm. Consistent with other examples of disruptive innovation, we propose that new levels of success are not dependent on new inventions, but rather on the strategic integration of existing data and technology assets. In support of these suggestions, we highlight the power of HD3, through recently published proof-of-concept applications in the areas of drug safety analysis and prediction, drug repositioning, the rational design of combination therapies and the global response to the COVID-19 pandemic. We conclude that innovators must play a key role in expediting the path to a largely human-focused, systems-based approach to drug discovery and research.

2.
CPT Pharmacometrics Syst Pharmacol ; 11(5): 540-555, 2022 05.
Article in English | MEDLINE | ID: mdl-35143713

ABSTRACT

Promising drug development efforts may frequently fail due to unintended adverse reactions. Several methods have been developed to analyze such data, aiming to improve pharmacovigilance and drug safety. In this work, we provide a brief review of key directions to quantitatively analyzing adverse events and explore the potential of augmenting these methods using additional molecular data descriptors. We argue that molecular expansion of adverse event data may provide a path to improving the insights gained through more traditional pharmacovigilance approaches. Examples include the ability to assess statistical relevance with respect to underlying biomolecular mechanisms, the ability to generate plausible causative hypotheses and/or confirmation where possible, the ability to computationally study potential clinical trial designs and/or results, as well as the further provision of advanced features incorporated in innovative methods, such as machine learning. In summary, molecular data expansion provides an elegant way to extend mechanistic modeling, systems pharmacology, and patient-centered approaches for the assessment of drug safety. We anticipate that such advances in real-world data informatics and outcome analytics will help to better inform public health, via the improved ability to prospectively understand and predict various types of drug-induced molecular perturbations and adverse events.


Subject(s)
Adverse Drug Reaction Reporting Systems , Drug-Related Side Effects and Adverse Reactions , Humans , Machine Learning , Marketing , Pharmacovigilance
3.
Clin Transl Sci ; 15(6): 1430-1438, 2022 06.
Article in English | MEDLINE | ID: mdl-35191192

ABSTRACT

Immunotherapy became a key pillar of cancer therapeutics with the approvals of ipilimumab, nivolumab, and pembrolizumab, which inhibit either cytotoxic T-lymphocyte antigen-4 (CTLA-4) or programmed death-1 (PD-1) that are negative regulators of T-cell activation. However, boosting T-cell activation is often accompanied by autoimmunity, leading to adverse drug reactions (ADRs), including high grade 3-4 colitis and its severe complications whose prevalence may reach 14% for combination checkpoint inhibitors. In this research, we investigated how mechanistic differences between anti-CTLA-4 (ipilimumab) and anti-PD-1 (nivolumab and pembrolizumab) affect colitis, a general class toxicity. The data analytical platform Molecular Health Effect was utilized to map population ADR data from the US Food and Drug Administration (FDA) Adverse Event Reporting System to chemical and biological databases for hypothesis generation regarding the underlying molecular mechanisms causing colitis. Disproportionality analysis was used to assess the statistical relevance between adverse events of interest and molecular causation. We verified that the anti-CTLA-4 drug is associated with an approximately three-fold higher proportional reporting ratio associated with colitis than those of the anti-PD-1 drugs. The signal of the molecular mechanisms, including signaling pathways of inflammatory cytokines, was statistically insignificant to test the hypothesis that the severer rate of colitis associated with ipilimumab would be due to a greater magnitude of T-cell activation as a result of earlier response of the anti-CTLA-4 drug in the immune response. This patient-centered systems-based approach provides an exploratory process to better understand drug pair adverse events at pathway and target levels through reverse translation from postmarket surveillance safety reports.


Subject(s)
Colitis , Drug-Related Side Effects and Adverse Reactions , Colitis/chemically induced , Drug-Related Side Effects and Adverse Reactions/diagnosis , Drug-Related Side Effects and Adverse Reactions/etiology , Humans , Immune Checkpoint Inhibitors , Ipilimumab/adverse effects , Nivolumab/adverse effects , Patient-Centered Care
4.
Clin Transl Sci ; 15(4): 1003-1013, 2022 04.
Article in English | MEDLINE | ID: mdl-35014203

ABSTRACT

Adverse drug reactions (ADRs) of targeted therapy drugs (TTDs) are frequently unexpected and long-term toxicities detract from exceptional efficacy of new TTDs. In this proof-of-concept study, we explored how molecular causation involved in trastuzumab-induced cardiotoxicity changes when trastuzumab was given in combination with doxorubicin, tamoxifen, paroxetine, or lapatinib. The data analytical platform Molecular Health Effect was utilized to map population ADR data from the US Food and Drug Administration (FDA) Adverse Event Reporting System to chemical and biological databases (such as UniProt and Reactome), for hypothesis generation regarding the underlying molecular mechanisms causing cardiotoxicity. Disproportionality analysis was used to assess the statistical relevance between adverse events of interest and molecular causation. Literature search was performed to compare the established hypotheses to published experimental findings. We found that the combination therapy of trastuzumab and doxorubicin may affect mitochondrial dysfunction in cardiomyocytes through different molecular pathways such as BCL-X and PGC-1α proteins, leading to a synergistic effect of cardiotoxicity. We found, on the other hand, that trastuzumab-induced cardiotoxicity would be diminished by concomitant use of tamoxifen, paroxetine, and/or lapatinib. Tamoxifen and paroxetine may cause less cardiotoxicity through an increase in antioxidant activities, such as glutathione conjugation. Lapatinib may decrease the apoptotic effects in cardiomyocytes by altering the effects of trastuzumab on BCL-X proteins. This patient-centered systems-based approach provides, based on the trastuzumab-induced ADR cardiotoxicity, an example of how to apply reverse translation to investigate ADRs at the molecular pathway and target level to understand the causality and prevalence during drug development of novel therapeutics.


Subject(s)
Cardiotoxicity , Drug-Related Side Effects and Adverse Reactions , Cardiotoxicity/etiology , Doxorubicin/adverse effects , Drug Development , Drug-Related Side Effects and Adverse Reactions/diagnosis , Humans , Lapatinib/adverse effects , Paroxetine/adverse effects , Patient-Centered Care , Tamoxifen , Trastuzumab/adverse effects
5.
Clin Pharmacol Ther ; 109(5): 1232-1243, 2021 05.
Article in English | MEDLINE | ID: mdl-33090463

ABSTRACT

We improved a previous pharmacological target adverse-event (TAE) profile model to predict adverse events (AEs) on US Food and Drug Administration (FDA) drug labels at the time of approval. The new model uses more drugs and features for learning as well as a new algorithm. Comparator drugs sharing similar target activities to a drug of interest were evaluated by aggregating AEs from the FDA Adverse Event Reporting System (FAERS), FDA drug labels, and medical literature. An ensemble machine learning model was used to evaluate FAERS case count, disproportionality scores, percent of comparator drug labels with a specific AE, and percent of comparator drugs with the reports of the event in the literature. Overall classifier performance was F1 of 0.71, area under the precision-recall curve of 0.78, and area under the receiver operating characteristic curve of 0.87. TAE analysis continues to show promise as a method to predict adverse events at the time of approval.


Subject(s)
Adverse Drug Reaction Reporting Systems , Algorithms , Pharmacovigilance , Data Mining , Drug Labeling , Drug-Related Side Effects and Adverse Reactions , Humans , Machine Learning , United States , United States Food and Drug Administration
6.
Int J Mol Sci ; 21(11)2020 May 29.
Article in English | MEDLINE | ID: mdl-32486089

ABSTRACT

BRCA1/2 variants are prognostic biomarkers for hereditary breast and/or ovarian cancer (HBOC) syndrome and predictive biomarkers for PARP inhibition. In this study, we benchmarked the classification of BRCA1/2 variants from patients with HBOC-related cancer using MH BRCA, a novel computational technology that combines the ACMG guidelines with expert-curated variant annotations. Evaluation of BRCA1/2 variants (n = 1040) taken from four HBOC studies showed strong concordance within the pathogenic (98.1%) subset. Comparison of MH BRCA's ACMG classification to ClinVar submitter content from ENIGMA, the international consortium of investigators on the clinical significance of BRCA1/2 variants, the ARUP laboratories, a clinical testing lab of the University of UTAH, and the German Cancer Consortium showed 99.98% concordance (4975 out of 4976 variants) in the pathogenic subset. In our patient cohort, refinement of patients with variants of unknown significance reduced the uncertainty of cancer-predisposing syndromes by 64.7% and identified three cases with potential family risk to HBOC due to a likely pathogenic variant BRCA1 p.V1653L (NM_007294.3:c.4957G > T; rs80357261). To assess whether classification results predict PARP inhibitor efficacy, contextualization with functional impact information on DNA repair activity were performed, using MH Guide. We found a strong correlation between treatment efficacy association and MH BRCA classifications. Importantly, low efficacy to PARP inhibition was predicted in 3.95% of pathogenic variants from four examined HBOC studies and our patient cohort, indicating the clinical relevance of the consolidated variant interpretation.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1 , Genes, BRCA2 , Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Biomarkers, Tumor/genetics , Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Computational Biology , DNA Repair , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genetic Testing , Genetic Variation , Germ-Line Mutation , Germany , Humans , Japan , Male , Ovarian Neoplasms/blood , Ovarian Neoplasms/diagnosis , Prostatic Neoplasms/blood , Prostatic Neoplasms/genetics , Reproducibility of Results , Retrospective Studies
7.
Cancers (Basel) ; 12(4)2020 Apr 19.
Article in English | MEDLINE | ID: mdl-32325840

ABSTRACT

Immune checkpoint inhibition represents an important therapeutic option for advanced melanoma patients. Results from clinical studies have shown that treatment with the PD-1 inhibitors Pembrolizumab and Nivolumab provides improved response and survival rates. Moreover, combining Nivolumab with the CTLA-4 inhibitor Ipilimumab is superior to the respective monotherapies. However, use of these immunotherapies frequently associated with, sometimes life-threatening, immune-related adverse events. Thus, more evidence-based studies are required to characterize the underlying mechanisms, towards more effective clinical management and treatment monitoring. Our study examines two sets of public adverse event data coming from FAERS and VigiBase, each with more than two thousand melanoma patients treated with Pembrolizumab. Standard disproportionality metrics are utilized to characterize the safety of Pembrolizumab and its reaction profile is compared to those of the widely used Ipilimumab and Nivolumab based on melanoma cases that report only one of them. Our results confirm known toxicological considerations for their related and distinct side-effect profiles and highlight specific immune-related adverse reactions. Our retrospective computational analysis includes more patients than examined in other studies and relies on evidence coming from public pharmacovigilance data that contain safety reports from clinical and controlled studies as well as reports of suspected adverse events coming from real-world post-marketing setting. Despite these informative insights, more prospective studies are necessary to fully characterize the efficacy of these agents.

8.
Pharmaceuticals (Basel) ; 12(4)2019 Sep 20.
Article in English | MEDLINE | ID: mdl-31546999

ABSTRACT

The development of monoclonal antibodies has dramatically changed the outcome of patients with non-Hodgkin's lymphoma (NHL), the most common hematological malignancy. However, despite the satisfying results of monoclonal antibody treatment, only few NHL patients are permanently cured with single-agent therapies. In this context, radioimmunotherapy, the administration of radionuclides conjugated to monoclonal antibodies, is aimed to augment the single-agent efficacy of immunotherapy in order to deliver targeted radiation to tumors, particularly CD20+ B-cell lymphomas. Based on evidence from several trials in NHL, the radiolabeled antibodies 90Y-ibritumomab tiuxetan (Zevalin, Spectrum Pharmaceuticals) and 131I-tositumomab (Bexxar, GlaxoSmithKline) received FDA approval in 2002 and 2003, respectively. However, none of the two radioimmunotherapeutic agents has been broadly applied in clinical practice. The main reason for the under-utilization of radioimmunotherapy includes economic and logistic considerations. However, concerns about potential side effects have also been raised. Driven by these developments, we performed retrospective analysis of adverse events reporting Zevalin or Bexxar, extracted from the FDA's Adverse Event Reporting System (FAERS) and the World Health Organization's VigiBase repository. Our results indicate that the two radioimmunotherapeutic agents have both related and distinct side effect profiles and confirm their known toxicological considerations. Our work also suggests that computational analysis of real-world post-marketing data can provide informative clinical insights. While more prospective studies are necessary to fully characterize the efficacy and safety of radioimmunotherapy, we expect that it has not yet reached its full therapeutic potential in modern hematological oncology.

9.
J Pers Med ; 9(3)2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31492009

ABSTRACT

The molecular characterization of patient tumors provides a rational and highly promising approach for guiding oncologists in treatment decision-making. Notwithstanding, genomic medicine still remains in its infancy, with innovators and early adopters continuing to carry a significant portion of the clinical and financial risk. Numerous innovative precision oncology trials have emerged globally to address the associated need for evidence of clinical utility. These studies seek to capitalize on the power of predictive biomarkers and/or treatment decision support analytics, to expeditiously and cost-effectively demonstrate the positive impact of these technologies on drug resistance/response, patient survival, and/or quality of life. Here, we discuss the molecular foundations of these approaches and highlight the diversity of innovative trial strategies that are capitalizing on this emergent knowledge. We conclude that, as increasing volumes of clinico-molecular outcomes data become available, in future, we will begin to transition away from expert systems for treatment decision support (TDS), towards the power of AI-assisted TDS-an evolution that may truly revolutionize the nature and success of cancer patient care.

10.
Healthcare (Basel) ; 7(1)2019 Mar 19.
Article in English | MEDLINE | ID: mdl-30893930

ABSTRACT

Adverse events are a common and for the most part unavoidable consequence of therapeutic intervention. Nevertheless, available tomes of such data now provide us with an invaluable opportunity to study the relationship between human phenotype and drug-induced protein perturbations within a patient system. Deciphering the molecular basis of such adverse responses is not only paramount to the development of safer drugs but also presents a unique opportunity to dissect disease systems in search of novel response biomarkers, drug targets, and efficacious combination therapies. Inspired by the potential applications of this approach, we first examined adverse event circumstances reported in FAERS and then performed a molecular level interrogation of cancer patient adverse events to investigate the prevalence of drug-drug interactions in the context of patient responses. We discuss avoidable and/or preventable cases and how molecular analytics can help optimize therapeutic use of co-medications. While up to one out of three adverse events in this dataset might be explicable by iatrogenic, patient, and product/device related factors, almost half of the patients in FAERS received multiple drugs and one in four may have experienced effects attributable to drug interactions.

11.
High Throughput ; 7(4)2018 Nov 23.
Article in English | MEDLINE | ID: mdl-30477159

ABSTRACT

We present a novel approach for the molecular transformation and analysis of patient clinical phenotypes. Building on the fact that drugs perturb the function of targets/genes, we integrated data from 8.2 million clinical reports detailing drug-induced side effects with the molecular world of drug-target information. Using this dataset, we extracted 1.8 million associations of clinical phenotypes to 770 human drug-targets. This collection is perhaps the largest phenotypic profiling reference of human targets to-date, and unique in that it enables rapid development of testable molecular hypotheses directly from human-specific information. We also present validation results demonstrating analytical utilities of the approach, including drug safety prediction, and the design of novel combination therapies. Challenging the long-standing notion that molecular perturbation studies cannot be performed in humans, our data allows researchers to capitalize on the vast tomes of clinical information available throughout the healthcare system.

12.
Mol Oncol ; 11(10): 1413-1429, 2017 10.
Article in English | MEDLINE | ID: mdl-28675654

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a tumor with an extremely poor prognosis, predominantly as a result of chemotherapy resistance and numerous somatic mutations. Consequently, PDAC is a prime candidate for the use of sequencing to identify causative mutations, facilitating subsequent administration of targeted therapy. In a feasibility study, we retrospectively assessed the therapeutic recommendations of a novel, evidence-based software that analyzes next-generation sequencing (NGS) data using a large panel of pharmacogenomic biomarkers for efficacy and toxicity. Tissue from 14 patients with PDAC was sequenced using NGS with a 620 gene panel. FASTQ files were fed into treatmentmap. The results were compared with chemotherapy in the patients, including all side effects. No changes in therapy were made. Known driver mutations for PDAC were confirmed (e.g. KRAS, TP53). Software analysis revealed positive biomarkers for predicted effective and ineffective treatments in all patients. At least one biomarker associated with increased toxicity could be detected in all patients. Patients had been receiving one of the currently approved chemotherapy agents. In two patients, toxicity could have been correctly predicted by the software analysis. The results suggest that NGS, in combination with an evidence-based software, could be conducted within a 2-week period, thus being feasible for clinical routine. Therapy recommendations were principally off-label use. Based on the predominant KRAS mutations, other drugs were predicted to be ineffective. The pharmacogenomic biomarkers indicative of increased toxicity could be retrospectively linked to reported negative side effects in the respective patients. Finally, the occurrence of somatic and germline mutations in cancer syndrome-associated genes is noteworthy, despite a high frequency of these particular variants in the background population. These results suggest software-analysis of NGS data provides evidence-based information on effective, ineffective and toxic drugs, potentially forming the basis for precision cancer medicine in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genomics , High-Throughput Nucleotide Sequencing , Pancreatic Neoplasms/genetics , Precision Medicine , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Feasibility Studies , Genomics/methods , Germ-Line Mutation , High-Throughput Nucleotide Sequencing/methods , Humans , Middle Aged , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Precision Medicine/methods , Prospective Studies , Proto-Oncogene Proteins p21(ras)/genetics , Software , Tumor Suppressor Protein p53/genetics
13.
Gynecol Oncol ; 141(1): 17-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27016224

ABSTRACT

Oncology is undergoing a data-driven metamorphosis. Armed with new and ever more efficient molecular and information technologies, we have entered an era where data is helping us spearhead the fight against cancer. This technology driven data explosion, often referred to as "big data", is not only expediting biomedical discovery, but it is also rapidly transforming the practice of oncology into an information science. This evolution is critical, as results to-date have revealed the immense complexity and genetic heterogeneity of patients and their tumors, a sobering reminder of the challenge facing every patient and their oncologist. This can only be addressed through development of clinico-molecular data analytics that provide a deeper understanding of the mechanisms controlling the biological and clinical response to available therapeutic options. Beyond the exciting implications for improved patient care, such advancements in predictive and evidence-based analytics stand to profoundly affect the processes of cancer drug discovery and associated clinical trials.


Subject(s)
Clinical Trials as Topic , Datasets as Topic , Drug Discovery , Humans , Research Design
14.
Cancer Cell ; 28(5): 610-622, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26481148

ABSTRACT

While recombinant human erythropoietin (rhEpo) has been widely used to treat anemia in cancer patients, concerns about its adverse effects on patient survival have emerged. A lack of correlation between expression of the canonical EpoR and rhEpo's effects on cancer cells prompted us to consider the existence of an alternative Epo receptor. Here, we identified EphB4 as an Epo receptor that triggers downstream signaling via STAT3 and promotes rhEpo-induced tumor growth and progression. In human ovarian and breast cancer samples, expression of EphB4 rather than the canonical EpoR correlated with decreased disease-specific survival in rhEpo-treated patients. These results identify EphB4 as a critical mediator of erythropoietin-induced tumor progression and further provide clinically significant dimension to the biology of erythropoietin.


Subject(s)
Breast Neoplasms/genetics , Erythropoietin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Ovarian Neoplasms/genetics , Receptor, EphB4/genetics , Adult , Aged , Aged, 80 and over , Animals , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Erythropoietin/genetics , Female , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Binding/drug effects , Receptor, EphB4/metabolism , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Young Adult
15.
Nat Commun ; 4: 1403, 2013.
Article in English | MEDLINE | ID: mdl-23360994

ABSTRACT

Noradrenaline can modulate multiple cellular functions important for cancer progression; however, how this single extracellular signal regulates such a broad array of cellular processes is unknown. Here we identify Src as a key regulator of phosphoproteomic signalling networks activated in response to beta-adrenergic signalling in cancer cells. These results also identify a new mechanism of Src phosphorylation that mediates beta-adrenergic/PKA regulation of downstream networks, thereby enhancing tumour cell migration, invasion and growth. In human ovarian cancer samples, high tumoural noradrenaline levels were correlated with high pSrc(Y419) levels. Moreover, among cancer patients, the use of beta blockers was significantly associated with reduced cancer-related mortality. Collectively, these data provide a pivotal molecular target for disrupting neural signalling in the tumour microenvironment.


Subject(s)
Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Receptors, Adrenergic, beta/metabolism , src-Family Kinases/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Female , Humans , Mice , Models, Molecular , Neoplasm Invasiveness , Neoplasm Metastasis , Norepinephrine/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Phosphorylation/drug effects , Phosphoserine/metabolism , Signal Transduction/drug effects , Stress, Physiological/drug effects , Survival Analysis , Tyrosine/metabolism , src-Family Kinases/chemistry
16.
Nat Rev Clin Oncol ; 8(12): 735-41, 2011 10 11.
Article in English | MEDLINE | ID: mdl-21989071

ABSTRACT

It was Hippocrates, the father of Western medicine, who first emphasized the patient as the most important determinant of therapeutic efficacy. Although the principle of adjusting treatment to specific patient characteristics has since been the strategy of physicians, this is undermined by a population-biased approach to drug development. Therefore, it is generally true to say that our current evidential approach to cancer treatment is driven more by drug-regulation requirements and market considerations than the specific needs of an individual patient. But, with cancer drug costs now spiraling out of control and the modest efficacy typically seen in patients, the community is again turning to Hippocrates' ancient paradigm--this time with emphasis on molecular considerations. Rapidly evolving technologies are empowering us to describe the molecular 'nature' of a patient and/or tumor and with this has come the beginning of truly personalized medicine, with maximized efficacy, cost effectiveness and hopefully improved survival for the patient.


Subject(s)
Neoplasms/economics , Neoplasms/therapy , Precision Medicine/economics , Humans
17.
Expert Rev Mol Diagn ; 11(6): 567-77, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21745011

ABSTRACT

While current trials of anticancer agents serve to provide a population-based validation of therapeutic activity, clinical success is typically restricted to tumors of select molecular subtype. Recent insights have yielded a growing catalogue of germline and tumor-based aberrations that can predetermine whether a patient will achieve clinical benefit from a drug or not. Thus, in order to realize the true potential of anticancer agents, we need to define the molecular contexts under which they will prove both efficacious and safe. In this article, we provide an overview of such molecular determinants and introduce the concept of 'cancer patient profiling' - the process and science of defining the optimal therapy for a given patient through the generation and analysis of system-wide molecular information.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Precision Medicine/trends , Animals , Antineoplastic Agents/pharmacokinetics , Humans , Membrane Transport Proteins/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Patient Compliance , Polymorphism, Genetic
18.
BMC Med Genomics ; 4: 9, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21251323

ABSTRACT

BACKGROUND: Colon cancer has been classically described by clinicopathologic features that permit the prediction of outcome only after surgical resection and staging. METHODS: We performed an unsupervised analysis of microarray data from 326 colon cancers to identify the first principal component (PC1) of the most variable set of genes. PC1 deciphered two primary, intrinsic molecular subtypes of colon cancer that predicted disease progression and recurrence. RESULTS: Here we report that the most dominant pattern of intrinsic gene expression in colon cancer (PC1) was tightly correlated (Pearson R = 0.92, P < 10(-135)) with the EMT signature-- both in gene identity and directionality. In a global micro-RNA screen, we further identified the most anti-correlated microRNA with PC1 as MiR200, known to regulate EMT. CONCLUSIONS: These data demonstrate that the biology underpinning the native, molecular classification of human colon cancer--previously thought to be highly heterogeneous-- was clarified through the lens of comprehensive transcriptome analysis.


Subject(s)
Colonic Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Principal Component Analysis , Cell Line, Tumor , Colonic Neoplasms/pathology , Disease Progression , Gene Expression Profiling/methods , Humans , Recurrence , Vimentin/metabolism
19.
Drug Discov Today ; 15(17-18): 749-56, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20601095

ABSTRACT

Off-target hits of drugs can lead to serious adverse effects or, conversely, to unforeseen alternative medical utility. Selectivity profiling against large panels of potential targets is essential for the drug discovery process to minimize attrition and maximize therapeutic utility. Lately, it has become apparent that drug promiscuity (polypharmacology) goes well beyond target families; therefore, lowering the profiling costs and expanding the coverage of targets is an industry-wide challenge to improve predictions. Here, we review current and promising drug profiling alternatives and commercial solutions in these exciting emerging fields.


Subject(s)
Database Management Systems , Drug Discovery/methods , Information Storage and Retrieval/methods , Systems Biology/methods , Animals , Humans
20.
Drug Discov Today ; 14(7-8): 373-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19200455

ABSTRACT

With the advent of targeted therapies promising to revolutionise the nature and success of patient care, the field of clinical oncology is facing a highly exciting future. While much of this enthusiasm comes from the hope for improved patient outcomes, a review of clinical response/relapse rates for current therapies provides a more sobering perspective. Given that the majority of patients are intrinsically resistant to the therapeutic potential of these molecules, efforts are now directed at characterising such non-responsive system behaviour and causative molecular insults. Testament to this is an expanding catalogue of target and system-based aberrations, often defined through retrospective analyses of clinical tissue and associated outcome data. What has emerged is a complex picture, where numerous potential sources of cancer-specific aberration can contribute to refractory tumour behaviour. Clinicians, regulators and sponsors must now collaborate to determine how such knowledge should be used to enhance the clinical decision process and associated regulatory guidance.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Biomedical Research , Drug Design , Humans , Research Design
SELECTION OF CITATIONS
SEARCH DETAIL
...