Abstract
Transcriptomics, which encompasses assessments of alternative splicing and alternative polyadenylation, identification of fusion transcripts, explorations of noncoding RNAs, transcript annotation, and discovery of novel transcripts, is a valuable tool for understanding cancer mechanisms and identifying biomarkers. Recent advances in high-throughput technologies have enabled large-scale gene expression profiling. Importantly, RNA expression profiling of tumor tissue has been successfully used to determine clinically actionable molecular alterations. The WINTHER precision medicine clinical trial was the first prospective trial in diverse solid malignancies that assessed both genomics and transcriptomics to match treatments to specific molecular alterations. The use of transcriptome analysis in WINTHER and other trials increased the number of targetable -omic changes compared to genomic profiling alone. Other applications of transcriptomics involve the evaluation of tumor and circulating noncoding RNAs as predictive and prognostic biomarkers, the improvement of risk stratification by the use of prognostic and predictive multigene assays, the identification of fusion transcripts that drive tumors, and an improved understanding of the impact of DNA changes as some genomic alterations are silenced at the RNA level. Finally, RNA sequencing and gene expression analysis have been incorporated into clinical trials to identify markers predicting response to immunotherapy. Many issues regarding the complexity of the analysis, its reproducibility and variability, and the interpretation of the results still need to be addressed. The integration of transcriptomics with genomics, proteomics, epigenetics, and tumor immune profiling will improve biomarker discovery and our understanding of disease mechanisms and, thereby, accelerate the implementation of precision oncology.
Keywords: gene expression, precision oncology, noncoding RNA, RNA sequencing transcriptomics
Background
Recent advances in technology have improved our understanding of carcinogenesis and led to the discovery of novel therapeutic targets. Precision oncology combines data from tumor genomic profiling, cell-free DNA assays, proteomic and immune profile analyses, and assessments of other markers to individualize treatment according to unique patient and tumor characteristics.1,2 Artificial intelligence and innovative clinical trial designs, including adaptive and N-of-1 trials, hold promise to accelerate the collection, analysis, and application of data on predictive biomarkers and novel targeted agents.3,4 Until recently, precision oncology focused mainly on genomic profiling of tumors.5,6 Advances in next-generation sequencing (NGS) technologies have enabled the time- and cost-efficient incorporation of genomics into daily clinical practice. Several precision oncology trials have demonstrated the clinical significance of genomics in identifying molecular alterations that are successfully targeted by novel treatments.3,4,6–9 Comprehensive gene panels are currently being used to identify molecular therapeutic targets and prognostic and predictive biomarkers and prospective clinical trials are assessing the value of molecular testing in treatment selection across tumor types.10–19
Despite this significant progress in the implementation of precision oncology, several challenges need to be addressed in clinical research and practice. First, it is critical to enhance our knowledge of tumor biology, the mechanisms of carcinogenesis, and driver alterations to improve our ability to identify robust prognostic and predictive biomarkers. Additionally, to date, only a few molecular alterations have been successfully targeted by novel agents.20–23 Many of these alterations are rare, and a large number of patients need to be screened to identify a single potential therapeutic target.24 Indeed, the proportion of patients who are matched to therapy in precision oncology trials generally ranges from 5% to 50% and often depends on whether the study is conducted in a specialized clinic with access to novel agents, off-label drug use, timely molecular profiling, and the expertise of clinical trial leaders in genomics.3–7,24–31 Therefore, our understanding of cancer complexity dictates that additional precision oncology methodologies need to be incorporated to enhance patient-treatment matching and prevent the development of treatment resistance.
Transcriptomic analyses have been included in precision oncology trials only recently and infrequently (Table 1).7,32–34 Transcriptomics refers to the study of all the RNA transcripts in a cell population, typically by using high-throughput technologies, namely microarrays and RNA sequencing (RNA-seq).35 In contrast to analysis of DNA sequencing data, the assessment of RNA status and measurement of transcripts can correlate gene expression with biologic activity and cellular status (Table 2).32–34,36–64 Gene expression, in turn, is influenced by genetic and epigenetic factors, such as DNA methylation and histone modifications. Early results of clinical trials suggest that transcriptomic analysis can increase the number of patients matched to drugs.7 Therefore, transcriptomics is a potentially valuable, though underused, technique for unraveling the underlying mechanisms of cancer and moving towards the implementation of precision oncology.
Table 1:
Examples of precision medicine trials using transcriptome analysis: Design and outcomes
Year First/Last authors | Trial name | Trial type | No. of pts consented | Proportion of pts. matc hed | Biomarker(s) | Outcome | Institute (s) | Comments |
---|---|---|---|---|---|---|---|---|
| ||||||||
20197 Rodon/Kurzrock | WINTHER | Prospective, navigational | 303 | 35% | NGS, transcriptomics | Higher matching scores correlated with longer PFS (P = 0.005) and OS (P = 0.03) | 5 countries (Spain, Israel, France, Canada, USA) | First trial on solid tumors to include transcriptomics |
201832 Weidenbusch/Burdach | PROVABES | Prospective | 20 | 45% | Gene expression profiling | Matched treatment was associated with improved OS (P = 0.001) and PFS (P = 0.0011) | Germany | Refractory pediatric sarcomas |
201633 Worst/Fleischhack | INFORM | Prospective | 57 | 18% | Whole-exome, RNA sequencing | Feasibility of use of comprehensive molecular analysis to guide treatment | 20 centers, Germany | Pediatric solid and hematologic malignancies |
201634 Oberg/Kung | PIPseq | Retrospective review of prospectively recruited pts | 101 | 16% | Whole-exome, RNA sequencing | Potentially targetable genomic alterations were identified in 38/101 (38%) pts | Columbia University Medical Center, USA | Pediatric solid and hematologic malignancies |
Abbreviations: NGS = next-generation sequencing, PFS = progression-free survival, OS = overall survival, pts = patients
Table 2:
Functional implications of transcriptomics
Implication | Description | Examples/References |
---|---|---|
Identification of therapeutic targets | Identify actionable molecular alterations using RNA-seq and navigate to therapy based on the result | WINTHER trial7,32–34 |
Detection of gene fusions | Detect a hybrid gene formed from 2 different genes as a result of chromosomal rearrangements | Targeted RNA-seq for fusion gene detection37–41 |
Transcript annotation | Discover novel transcripts | Annotating genomes42,43 |
Regulation by miRNA sequencing | Explore the role of miRNA in mRNA regulation | miRNA role in mechanisms of tumorigenesis and as prognostic and predictive biomarkers44–47 |
Influence of noncoding RNA sequencing | Explore the role of noncoding RNA molecules in mRNA regulation | Noncoding RNA molecules as diagnostic, prognostic, and predictive biomarkers36,48 |
Use of prognostic gene expression signatures | Identify gene expression signatures used to assess patient prognosis | Oncotype DX,49,71 MammaPrint50,51 |
Use of predictive gene expression signatures | Identify gene expression signatures used to assess the benefit from treatments | Oncotype DX49,71 |
Identification of tissue of origin for cancer of unknown primary (CUP) | Identify the primary tumor site using gene expression profiling | Transcriptome-based prediction of primary tumor in patients with CUP52–56,120 |
Understanding of tumor heterogeneity | Use gene expression profiling to identify intrinsic cancer subtypes | Tumor classification into subtypes57–59 |
Interrogation of biomarkers for immuno-oncology | Use immune cell profiling to explore mechanisms of immune escape and identify immune cell phenotypes | Transcriptomics in cancer immunotherapy60–62,157,158 |
Silencing of the transcriptome | Understand genomic alterations that are silenced at the transcript level | Identification of discrepancies between DNA molecular alterations and RNA expression and effect on therapeutic resistance142 Silencing may also be exploitable for therapeutic purposes |
Interrogation of expression levels | Understand differences between amplified and expressed genes | Gene amplification is not always associated with increased gene expression64 |
History
Early methods to assess gene expression included Northern blotting, reverse transcriptase quantitative polymerase chain reaction (RT-qPCR), and sequencing of short nucleotide arrays (expressed sequence tags) that were generated from complementary DNAs (cDNAs). However, these methods were developed to evaluate limited numbers of transcripts and are inadequate for comprehensive RNA profiling. Subsequently, serial analysis of gene expression65 and DNA microarrays enabled the analysis of large-scale gene expression arrays.57,66–68 With microarrays, investigators can quickly assess the expression levels of thousands of genes simultaneously. Comprehensive profiling of tumor samples, normal tissues, and cancer cell lines yielded a large volume of transcriptomic data. Dedicated databases were developed to store gene expression datasets that were made publicly available to allow their use by other investigators.69,70 Examples of the implementation of gene expression analysis in clinical practice include the commercially available MammaPrint and Oncotype DX assays, which are used to assess prognosis and/or select treatment in patients with breast cancer.51,71
Transcriptome biology
Dynamic nature of the transcriptome
In multicellular organisms, the same genes, and thus the same genome, are found in almost every cell. Not every gene is transcriptionally active in every cell, however, and different patterns of gene expression appear in different types of cells. In addition, multiple RNA variants can be produced by a single gene owing to alternative splicing, RNA editing, or alternative transcription initiation and termination sites. The total transcriptional activity, that is, the full range of RNA molecules expressed, is reflected in the transcriptome of an organism. The transcriptome can be represented as the percentage of the genetic code that is transcribed into RNA molecules, which is estimated to be less than 5% of the genome in humans.72 In contrast to the genome, the transcriptome changes in response to cellular cues. Indeed, an organism’s transcriptome varies dynamically depending on many factors, including environmental conditions and developmental stage.
Epitranscriptomics
Epitranscriptomics, also known as RNA epigenetics, describes the diverse posttranscriptional modifications occurring in cellular RNA. This dynamic processing occurs during RNA maturation under the regulation of RNA-binding proteins. Τo date, more than 150 types of RNA modifications have been identified, including RNA methylation and editing.73 While the exact role of these modifications is still under investigation, studies have shown that it extends from maintaining the structure of RNA to regulating critical cell systems and that disruptions in RNA processing are associated with various diseases, including cancer.74,75 Research focusing on specific modifications has revealed associations between deregulation of RNA processing and cancer progression, aggressive tumor behavior, and deregulated cellular processes.76,77 Given the oncogenic nature of these modifications, their regulators could be targeted for novel therapies.
Functional uses of transcriptomics
Identification of therapeutic targets
Transcriptomic data have been incorporated into many different tumor molecular profiles to increase the number of targetable molecular alterations and provide additional therapeutic options to patients with advanced cancer.7,32–34,78,79 In one study, gene expression profiling of longitudinally collected primary breast tumors and metastatic lesions identified several highly targetable genes.78 Administration of therapeutic agents against these alterations in patient-derived xenograft models led to a statistically significant antitumor response vs. controls. In another study of 1049 children and young adults with de novo acute megakaryocytic leukemia, RNA-seq revealed druggable targets.79 Importantly, in our WINTHER precision medicine clinical trial, which prospectively assessed both genomics and transcriptomics in diverse solid malignancies, the use of transcriptomic analysis increased the number of targetable -omic changes by a third over NGS.7
Detection of gene fusions
Conventional cytogenetic analyses, including fluorescence in situ hybridization and RT-qPCR, have been widely used for fusion gene detection. However, these methods are designed to discern the presence of specific known gene fusions, not identify novel ones. Newer techniques and algorithms have been developed for the performance of wide-scale RNA-seq to detect novel gene fusions.38–40
MicroRNA sequencing
Transcription is the process by which individual genes are copied into RNA molecules to build the transcriptome. The translation of these RNA molecules into proteins constructs the proteome. The RNA content of a cell includes coding and noncoding RNA. The coding RNA, which makes up the transcriptome, consists of messenger RNAs (mRNAs). The noncoding RNAs primarily include ribosomal RNAs (rRNAs), which are components of ribosomes (the structures on which protein synthesis takes place), and transfer RNAs (tRNAs), which are small molecules that are involved in protein synthesis by carrying amino acids to the ribosome and ensuring that they are linked together in the order specified by the nucleotide sequence of the mRNA that is being translated into a protein. In addition to rRNAs and tRNAs, short regulatory non-coding RNAs, including piwi-associated RNAs, endogenous short-interfering RNAs, and microRNAs (Figure 1), have received significant attention due to their role as regulators of gene expression.
Figure 1. Noncoding RNAs.
The role of noncoding RNAs in cancer diagnosis, prognosis, prediction of response to cancer therapy and disease monitoring is currently under investigation. MicroRNAs (miRNAs) are noncoding RNAs that play an important role as regulators of gene expression. A primary miRNA transcript is cleaved by the microprocessor complex Drosha-DGCR8 in the nucleus. The resulting pre-miRNA is exported from the nucleus. In the cytoplasm, Dicer cleaves the pre-miRNA hairpin to its mature length. The mature miRNA is loaded into the RNA-induced silencing complex (RISC), where it guides RISC to silence target mRNA through mRNA degradation or translational repression.
Regulation by microRNAs
Over the past decade, several studies have focused on revealing the microRNA (miRNA) repertoire.80–83 These small non-protein-coding RNA molecules (18–25 nucleotides) are capable of controlling gene expression by binding to mRNA targets, thus interfering in the final protein output. MiRNAs have been recognized as major regulators of biological features including proliferation,84,85 migration,86 and apoptosis.87 MiRNA profiling has been successfully used to molecularly classify tumors,88,89 to assess the prognosis of patients with different tumor types,90–92 and to predict the development of resistance to treatments.93–95 MiRNAs and miRNA mimics are currently being evaluated in clinical trials as therapeutic agents (NCT02369198, NCT01829971, NCT02580552). Intriguing data have emerged from the study of circulating miRNAs. Studies show that miRNAs, encapsulated in exosomes, are released from cells and can be detected in biological fluids.96,97 The noninvasive assessment of circulating miRNAs is an appealing approach for disease monitoring and diagnosis. Several studies are investigating the role of circulating miRNAs as cancer diagnostic and prognostic biomarkers.98
Noncoding RNA sequencing
In addition to miRNAs, gene expression profiling can reveal other molecular elements, including other noncoding RNAs. Noncoding RNAs are transcribed from non-protein-coding regions of the genome and have a wide range of regulatory functions.99,100 Depending on their transcript size, they are categorized as small (< 200 bp) or long noncoding RNAs (lncRNAs; > 200 bp, up to ~100 kb). Noncoding RNAs have been shown to play an important role in cancer, and noncoding RNA expression profiling has diagnostic, prognostic, and predictive value in patients with solid and hematologic malignancies.36,101 Recent studies show that circulating noncoding RNA levels in body fluids (eg, serum, urine) differ between individuals with and without cancer.101–104 These data suggest that noncoding RNAs could be used as diagnostic biomarkers in cancer screening. Other investigators have shown that profiling of noncoding RNAs has prognostic utility in patients with cancer.36,101 Finally, studies are currently evaluating the role of circulating noncoding RNAs in predicting response or resistance to various treatments. Preliminary data suggest that this approach might provide useful insights with which to identify patients who are likely to have a response to therapy.105–108 The clinical appeal of the use of noncoding RNAs and miRNAs as biomarkers is that they can be obtained noninvasively via liquid biopsies. However, the clinical utility of these approaches has yet to be prospectively defined and validated.
Prognostic gene expression signatures
Several studies have evaluated the presence of prognostic biomarkers through transcriptomic analysis. Gene expression signatures are alterations in the expression of single genes or sets of genes with a validated association with disease prognosis, therapeutic benefit, or cancer diagnosis. Despite recent technological advances, robust molecular prognostic biomarkers are still lacking in clinical practice. As a result, there is a great need to improve risk assessment in patients with cancer in order to identify patients at high risk of recurrence or death. When identified, these patients might be treated more aggressively or with different therapeutic strategies.
To improve risk stratification, prognostic multigene assays have been developed and validated in lung,109 breast,51,57,110 colon,111,112 and other tumor types.113–117 These gene expression signatures provide prognostic information independently of clinicopathologic features and have been shown to improve the stratification of patients based on risk of recurrence. However, only a few, such as Oncotype DX in breast cancer, are recommended in international patient management guidelines for use in clinical practice to predict whether a patient is likely to have a recurrence of the disease.71 Future studies might reveal the prognostic value of additional gene expression signatures in different tumor types.
Predictive gene expression signatures
Other studies have employed transcriptome analysis to identify gene expression signatures that can predict response to specific cancer therapies.118,119 However, only a small number of predictive signatures have been validated in prospective randomized clinical trials. Therefore, only a few gene expression signatures with verified clinical validity are used in daily practice. For instance, the validated predictive model Oncotype DX is often used in patients with hormone-receptor–positive, human epidermal growth factor receptor type 2–negative, early-stage breast cancer to predict benefit from adjuvant chemotherapy.71 It is critical that these signatures are robust, highly reproducible, and validated in diverse populations beyond the tightly controlled environment of clinical trials.
Classification of cancer of unknown primary
Transcriptomics holds promise as an additional tool for the accurate classification of cancer of unknown primary (CUP). Several investigators have explored gene expression profiles and revealed biomarkers indicative of the origin of the tumor in patients with CUP.52–56,120 In one study, transcriptome analysis of 16 674 tumors corresponding to 22 tumor types revealed a 154-gene expression signature that aided the identification of tumor origin.120 Independent validation of the signature was successfully performed using 9626 primary tumors. In another study, a cancer type classifier was developed using gene expression data from more than 10 000 tissue samples from 30 tumor types.56 The accuracy of the classifier was high (77%–88%) and varied according to the primary tumor type, the purity of the tumor sample, and the site of tumor tissue (primary or metastatic). Finally, computational algorithms have been employed to mine RNA expression datasets and identify diagnostic classifiers.55 Gene expression profiling can be incorporated into diagnostic algorithms for patients with CUP to increase rates of accurate classification and improve understanding of patients’ prognoses. However, in randomized trials, treating CUP according to tissue-of-origin signatures did not effectively improve outcomes.121,122
Assessment of tumor heterogeneity
Investigators exploring the intratumor heterogeneity of renal tumors (primary and corresponding metastatic sites) demonstrated that tumors are not only genomically but also transcriptomically heterogeneous.123 Specifically, they showed that gene expression signatures suggestive of good and poor prognoses can be identified within the same tumor. Others explored tumor heterogeneity between foci of multifocal or multicentric invasive lobular breast carcinoma and observed heterogenous transcriptional profiles in expression analysis of 730 genes.124 More recently, studies have incorporated single-cell RNA-seq methods to explore tumor heterogeneity in detail.59,125–127
Prediction of response to immuno-oncology
Despite unprecedented improvement in patient outcomes by the use of immune checkpoint inhibitors, mechanisms of resistance significantly limit the benefit from these treatments. Several genomic alterations are being evaluated as predictive biomarkers for immunotherapy.128–132
In recent studies, RNA-seq and gene expression analysis have been incorporated to predict responsiveness to immunotherapy. In 1 study, analysis of the genome and transcriptome of melanoma tissue samples identified biomarkers that predicted response to anti-PD-1 therapy.133 In addition, transcriptomic profiles suggested that innate tumor resistance to anti-PD-1 immunotherapy was associated with mesenchymal and inflammatory tumor phenotypes.133 In a study of metastatic melanoma tumors, gene expression profiling showed that tumors with PTEN loss had lower expression of inflammation-related genes, suggesting that PTEN loss could be associated with resistance to immunotherapy.134 Other studies identified gene expression profiles associated with response or resistance to immunotherapeutic agents.135,136 Transcriptome analysis has also been used to study how the tumor microenvironment evolves after treatment with immunotherapy,137 as well as tumor immune heterogeneity136 and tumor immune classification.138
Transcriptomic silencing
Gene expression silencing is a mechanism of transcriptional regulation in mammalian cells. It is mediated by RNA interference (RNAi), in which a small noncoding RNA associates with RNA-induced silencing complex and degrades target mRNAs.139 Gene silencing is seen with exogenous (e.g., viral, bacterial) and endogenous (e.g., transgene, transposon) sequences. In addition, RNAi is involved in the regulation of gene expression and other biologic processes. Transcriptomic silencing is used in combination with gene expression profiling to identify novel biomarkers and therapeutic targets.140 For instance, in 1 study of 9873 prostate tissue samples, gene expression profiling revealed 295 genes that had high mRNA expression in prostate cancer samples. An RNAi-based cell viability assay was incorporated to demonstrate the role of gene silencing in prostate cancer cell lines and silencing of a candidate gene (ERGIC1) led to inhibition of ERG mRNA expression and decreased proliferation of ERG-positive prostate cancer cells.140 These data suggested that ERG could be tested as a candidate drug target.
RNAi-mediated gene silencing is an appealing approach for use in cancer treatment because it can silence oncogenes and other driver genes involved in cancer cell proliferation, the cell cycle, and tumor progression. In this vein, clinical trials are currently evaluating the clinical benefit from the use of short interfering RNA (siRNA)-based treatments in patients with cancer. PROTACT is a phase II trial assessing the clinical utility of siG12D LODER in combination with chemotherapy in patients with locally advanced pancreatic cancer (NCT01676259). In this study, SiG12D LODER, a miniature biodegradable polymeric matrix that encloses siRNAs targeting G12D-mutated KRAS, is implanted in the tumor via endoscopic ultrasound. Published data from another phase I/IIa trial evaluating the administration of SiG12D LODER as first-line treatment (in combination with chemotherapy) in 15 patients with locally advanced pancreatic cancer showed that this therapeutic approach was safe and well tolerated.141 Another phase I trial is evaluating the administration of a liposomal agent consisting of siRNAs against EphA2 that are encapsulated into 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine liposomes in patients with advanced cancer (NCT01591356). Both trials are currently recruiting patients.
In addition, RNA-seq, along with NGS, has revealed transcriptomic silencing of DNA mutations in advanced cancers, which has important implications for resistance to targeted therapeutics. In a study of 45 patients with cancer, 86 pathogenic DNA alterations were identified, including 17 (19.8%) alterations that were not observed at the RNA level.142 Among these patients, 31% (14/45) had 1 or more DNA alterations that were not expressed at the RNA level. Examples of genes that had pathogenic DNA alterations not seen at the RNA level included ALK, KDR, and GNAS. On the other hand, alterations involving other genes, including TP53, PIK3CA, and FGFR3, showed 100% concordance between DNA and RNA. This study concluded that a significant number of patients had DNA alterations that are silenced at the RNA level and that transcriptomic silencing merits additional investigation as a mechanism of therapeutic resistance.
Interrogation of gene expression levels
Gene amplification has frequently been described as a mechanism leading to carcinogenesis. Even though gene amplification would be expected to correlate with overexpression, there are cases where these phenomena are not associated.64 In order to characterize a gene amplification as a “driver” alteration, overexpression is required. Gene expression profiling alone or in combination with copy-number variation (CNV) analysis is used to identify candidate driver genes for molecular therapeutic targeting.34,143,144 In one study, investigators analyzed gene CNV and mRNA expression data from The Cancer Genome Atlas project. They identified 42 candidate cancer driver genes and validated their oncogenic activity via siRNA knockdown.143 In another study, transcriptome and CNV analysis enabled the identification of targetable molecular alterations as well as prognostic and diagnostic biomarkers.34 Importantly, some studies suggest that the variability in normal transcript levels between tissues and between individuals means that cancer transcript expression levels must be compared to their normal-tissue counterparts for accuracy.7
Novel bioinformatic approaches in large datasets are essential for exploring and describing the association of gene amplification and expression with mechanisms of carcinogenesis. Launched by the National Institutes of Health in 2010, the Genotype-Tissue Expression (GTEx) Program aims to explore the association between genetic variants and gene expression145 and consists of a tissue bank with multiple tissue samples for each donor along with whole-genome and RNA-seq data from approximately 960 deceased adult donors. This Program provides publicly available data to the research community, which investigators can use in studies of mechanisms of gene regulation, genetic variation and its association with gene expression and disease risk, and novel methods of gene expression analysis.145,146
Machine learning
Machine learning, a subfield of artificial intelligence, has been playing an increasingly central role in biomedical and pharmaceutical sciences,147 primarily owing to the need to develop new tools to analyze the influx of complex, heterogeneous, and multidimensional biological datasets. Machine learning approaches can be used to detect key genomic and epigenetic features that can help classify patients who may have different responses to a drug. For this purpose, multiple feature-selection algorithms have been proposed, including filters (e.g., Markov blanket filtering), wrappers (e.g., gradient-based-leave-one-out gene selection), and embedded techniques (e.g., block diagonal linear discriminant analysis). Next, a variety of established machine learning algorithms, including artificial neural networks, support vector machines, and decision trees, can build predictive drug response models based on the features. As an example, a multilayer perceptron neural network can be trained to generate an output (e.g., whether the patient responds to targeted treatments) in response to a set of input variables (e.g., genetic variations). Most recent developments focus on deep learning methods, a subset of machine learning based on artificial neural networks. Currently, deep learning approaches hold significant promise as they are capable of unsupervised learning using data that are unstructured or unlabeled,148 potentially addressing some of the inherent limitations of machine learning.149
Transcriptomics in clinical trials
The WINTHER trial was one of the first studies to incorporate transcriptional analysis, in addition to genomics, in order to match patients with solid tumors to therapeutic agents (Table 1).7 WINTHER was an international clinical trial directed by the Worldwide Innovative Network for Personalized Cancer Medicine (WIN Consortium) and involved centers in 5 countries. The rationale for the trial was the need to expand the identification of predictive biomarkers beyond genomic aberrations. Therefore, a double biopsy of tumor (primary or metastatic) and normal tissue from each patient was performed after study enrollment. Tissue was used for DNA analysis by NGS of 236 cancer-related genes. In addition, the study included gene expression analysis of matched tumor and normal tissue pairs. Transcriptional data were evaluated for treatment selection if genomic data generated no recommended treatment option. The selection of targeted therapy using transcriptomic analysis occurred in a stepwise process as follows: gene expression analysis was performed on tumor tissue and paired normal samples from individual patients. Bioinformatic analysis was performed, and the differential expression of these genes in tumor versus analogous normal tissue was used to select treatment. Details for each overexpressed or under-expressed mRNA were provided, including gene fold changes, expression intensity, and type of molecular abnormality. Each alteration was evaluated as a potential match with a targeted agent on the basis of a knowledge database. The WINTHER database included information on the gene expression “targeting” and efficacy of both registered drugs and drugs evaluated in clinical trials. Treatment selection was based on the recommendations of a clinical management committee. Finally, the trial evaluated the use of an exploratory matching score. The score, which was calculated in a post hoc, blinded fashion, was derived by dividing the number of molecular alterations that could be associated with a therapeutic agent by the total number of alterations (for DNA analysis) or by adding the reciprocal of the ranks of each matched drug received by the patient according to the WINTHER algorithm (for RNA analysis).
Of 303 patients who consented, 107 (35%) patients received 1 or more agents and were evaluable for analysis. Patients were heavily pretreated with a median of 3 prior therapies. Among the 107 evaluable patients, 15% had stable disease for 6 months or longer, and 11% had partial or complete responses. Having 2 or fewer previous therapies, an Eastern Cooperative Oncology Group (ECOG) performance status of 0, and a higher matching score were independently associated with longer progression-free survival. ECOG performance status of 0 and a higher matching score were also associated with longer overall survival on multivariate analysis. The WINTHER trial demonstrated that transcriptomic analysis can be an indispensable tool for the navigation of treatment in selected patients. However, the analysis was complex and required bioinformatic expertise.
Other clinical trials focusing on pediatric patients have matched therapies to molecular alterations identified by transcriptomic analysis.32–34 In 1 study, gene expression profiling was performed in 20 patients with refractory pediatric sarcoma to identify overexpressed genes and deregulated pathways that could be therapeutically targeted.32 The actionable targets most commonly identified were TOP2A and FGFR1 upregulation. Nine of the 20 patients received a targeted therapy. Patients who received targeted treatments had higher overall (P = 0.0014) and progression-free (P = 0.0011) survival rates compared to patients who did not.
The Individualized Therapy for Relapsed Malignancies in Childhood (INFORM) study used RNA sequencing in addition to whole-exome and low-coverage whole-genome sequencing in prospectively recruited children with high-risk relapsed or refractory malignancies.33 An expert multidisciplinary panel prioritized the identified molecular alterations using a customized prioritization algorithm. The algorithm used a 7-step priority scale, ranging from “very low” to “very high,” based on the biological relevance, type of alteration, and potential druggability. Investigators created an internal database comprising genes considered to be potential therapeutic targets. Treatment was selected on the basis of physician choice. In the initial report of 52 patients, candidate targetable molecular alterations were identified in 26 (50%) of the patients, 10 of whom received the respective treatment. Even though patients were heavily pretreated, clinical responses were noted.
The investigators recently reported updated results of the INFORM study. Of 1,300 patients who were enrolled at 72 centers, 525 were included in the analysis.150 A “very high-” or “high-” priority actionable target was identified in 8% and 14.8% of patients, respectively. Patients who received targeted treatment had longer progression-free survival compared to patients who received non-targeted therapy (204.5 vs. 114 days, P = 0.0095), although no difference was noted in overall survival between the 2 groups. Patients who received treatment matched to a “very high-” priority target had higher time-to-progression ratios (time to progression before/time to progression after enrollment in INFORM) compared to the remaining patients.
The clinical utility of molecular analysis in pediatric tumors was also demonstrated in the Precision in Pediatric Sequencing (PIPseq) Program study.34 In this study, whole-exome and RNA sequencing were performed in 101 patients with solid and hematologic malignancies. Molecular alterations (variant calls, copy number variations [CNV], fusions, and overexpressed genes) were initially reviewed by a multidisciplinary molecular tumor board comprising molecular pathologists, pediatric oncologists, medical geneticists, bioinformaticians, and cancer biologists. The final report included the following clinically relevant alterations: driver mutations, gain- or loss-of-function molecular alterations in oncogenes and tumor suppressor genes, respectively, and fusions known or expected to be oncogenic drivers. Investigators included “clinically meaningful” molecular alterations, used for diagnosis, prognosis, treatment (as therapeutic targets), refinement of a therapeutic plan, and/or health maintenance interventions. On the basis of the level of evidence for clinical actionability,151 a tiered report of all clinically relevant alterations was provided to the referring physician for treatment selection. Transcriptomic analysis identified diagnostic, prognostic, and predictive molecular alterations in 37 (57%) of 65 patients. Overall, 15 (23%) patients received targetable therapeutic agents on the basis of sequencing findings. These studies serve as proof of concept showing that the use of comprehensive gene expression profiling in daily practice is feasible and provides useful diagnostic, prognostic, and predictive data. Targeted therapeutic agents can be matched to molecular alterations, providing clinical responses even in heavily pretreated patients. Finally, future prospective trials validating the clinical utility of this approach are warranted and should also address the cost, time burden, and inconsistent analysis of transcriptomic data. Ongoing clinical trials are exploring the importance of transcriptomics in cancer therapy.
Challenges and limitations
The main challenges associated with transcriptomic analyses are related to the handling of tissue samples and the application of advanced computational methodologies. For example, RNA-seq protocols include sample preparation, RNA isolation and selection, and cDNA synthesis and sequencing followed by bioinformatic analysis. Formalin-fixed, paraffin-embedded tissue samples can include RNA that is degraded, fragmented,152 or contaminated.153 Contamination caused by errors during sample preparation or by inadequate equipment sterilization can lead to the presence of sequence data from a different sample. Additionally, tumor samples can be contaminated by normal cells that infiltrate or surround the tumor. Experimental methods (cell sorting or laser capture micro-dissection)154 and bioinformatics algorithms aim to eliminate contamination effects.155 Novel methods have been developed to address the issue of low levels of RNA in archival tissue. Additionally, methodologic artifacts are often encountered in transcriptome analysis and require careful assessment.
Another challenge involves the application of advanced computational methodologies. High-level bioinformatic infrastructure is required to conduct complex analyses of profiling data. For instance, in the WINTHER trial, RNA analysis required the systematic development of an algorithm by bioinformaticians.7 Therefore, the implementation of transcriptomic analysis in clinical workflows may be more complicated than that of genomic analysis. In addition, reproducibility issues need to be addressed. RNA profiling can be used to compare tumor tissue with normal tissue from the same organ, such as in the WINTHER trial; however, some investigators believe that peripheral blood or buccal swab samples could also be used for comparison. This difference may introduce variability in the interpretation of the results.
Overall, investigators who used transcriptomics in clinical trials developed diverse and complex algorithms for characterizing the actionability of molecular alterations.7,33,34 Consequently, the use of transcriptomics in clinical practice is arduous and expensive. For instance, in the INFORM study, the average cost per patient for the molecular analysis, including tissue sample shipment, data processing and storage, labor, and general costs, was approximately €7,000.33 The time from tissue processing to start of analysis ranged from 0 to 112 days.33 Therefore, transcriptomic analysis requires significant optimization, validation and cost decrease in order to be optimally implemented in clinical practice. Standardization of bioinformatic analysis through expert consensus would make the use of transcriptomic analysis in routine clinical practice more consistent.
Conclusions and future perspectives
Tumor genomic profiling approaches provide average signatures and a snapshot of the tumor state at the time of biopsy but often do not reflect the complete tumor biology, all tumor components, or the intrinsic heterogeneity of individual cell populations. Innovative emerging techniques, including single-cell transcriptome profiling technologies, will improve our understanding of tumor biology in individual patients and will provide a plethora of translational discovery opportunities.59,125,156 However, understanding tumor complexity and heterogeneity, as well as the dynamic expression of the genome, requires the incorporation of several newer methodologies. The future of precision medicine lies in the integration of genomics, transcriptomics, proteomics, and epigenetics in order to fully elucidate tumor immune and -omic profiles, optimize comprehensive tumor molecular profiles, and inform treatment decisions. By focusing on the role of transcriptomics in identifying appropriate targeted therapies, clinical trials can validate and enrich the available data demonstrating that the use of transcriptomics can increase the number of patients treated with matched targeted therapy and lead to favorable outcomes, hence providing the next frontier for precision medicine.
Acknowledgments
Editorial support was provided by Amy Ninetto in Scientific Publications, Research Medical Library, The University of Texas MD Anderson Cancer Center.
Funding
NIH/NCI, award number P30 CA016672
Abbreviations
- cDNA
complementary DNA
- CNV
copy-number variation
- CUP
cancer of unknown primary
- EGOG
Eastern Cooperative Oncology Group
- GTEx
Genotype-Tissue Expression
- INFORM
Individualized Therapy for Relapsed Malignancies in Childhood
- mRNA
messenger RNA
- NGS
next-generation sequencing
- PIPseq
Precision in Pediatric Sequencing
- RNAi
RNA interference
- RNA-seq
RNA sequencing
- rRNA
ribosomal RNA
- RT-qPCR
reverse transcriptase quantitative polymerase chain reaction
- siRNA
short interfering RNA
- tRNA
transfer RNA
- WIN
Worldwide Innovative Network
Footnotes
Competing interests
Dr. Apostolia-Maria Tsimberidou has the following financial relationships to disclose: Research Funding (Institution): Immatics, Parker Institute for Cancer Immunotherapy, Tempus, OBI Pharma, EMD Serono, Baxalta, ONYX, Bayer, Boston Biomedical, Placon Therapeutics, Karus Therapeutics, and Tvardi Therapeutics. Consulting or Advisory Role: Covance, Genentech and Tempus.
Dr. Elena Fountzilas has the following financial relationships to disclose: Travel grants: Merck, Pfizer, and K.A.M Oncology/Hematology; Speaker fees: from Roche, Leo; stock ownership Deciphera Pharmaceuticals, Inc.
Leonidas Bleris has the following financial relationships to disclose: Research funding from US National Science Foundation (NSF) CAREER grant 1351354, NSF 1361355, and a Cecil H. and Ida Green Endowment.
Dr. Razelle Kurzrock has the following financial relationships to disclose: Research Funding (Institution): Incyte, Genentech, Merck Serono, Pfizer, Sequenom, Foundation Medicine, Konica Minolta, Grifols, Biologic Dynamics, and Guardant. Consulting role: X-Biotech, Loxo, and Actuate Therapeutics. Speaker fees: Roche. Ownership interest: IDbyDNA and CureMatch, Inc. Board member: CureMatch and CureMetrix.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Kato S, Subbiah V, Kurzrock R: Counterpoint: Successes in the Pursuit of Precision Medicine: Biomarkers Take Credit. J Natl Compr Canc Netw 15:863–866, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Subbiah V, Kurzrock R: Debunking the Delusion That Precision Oncology Is an Illusion. Oncologist 22:881–882, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sicklick JK, Kato S, Okamura R, et al. : Molecular profiling of cancer patients enables personalized combination therapy: the I-PREDICT study. Nat Med 25:744–750, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Tsimberidou AM, Iskander NG, Hong DS, et al. : Personalized medicine in a phase I clinical trials program: the MD Anderson Cancer Center initiative. Clin Cancer Res 18:6373–83, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wheler JJ, Janku F, Naing A, et al. : Cancer Therapy Directed by Comprehensive Genomic Profiling: A Single Center Study. Cancer Res 76:3690–701, 2016 [DOI] [PubMed] [Google Scholar]
- 6.Massard C, Michiels S, Ferte C, et al. : High-Throughput Genomics and Clinical Outcome in Hard-to-Treat Advanced Cancers: Results of the MOSCATO 01 Trial. Cancer Discov 7:586–595, 2017 [DOI] [PubMed] [Google Scholar]
- 7.Rodon J, Soria JC, Berger R, et al. : Genomic and transcriptomic profiling expands precision cancer medicine: the WINTHER trial. Nat Med 25:751–758, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tsimberidou AM, Wen S, Hong DS, et al. : Personalized medicine for patients with advanced cancer in the phase I program at MD Anderson: validation and landmark analyses. Clin Cancer Res 20:4827–36, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Tsimberidou AM, Hong DS, Ye Y, et al. : Initiative for Molecular Profiling and Advanced Cancer Therapy (IMPACT): An MD Anderson Precision Medicine Study. JCO Precis Oncol 2017, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Aftimos PG, Antunes De Melo e Oliveira AM, Hilbers F, et al. : 152OFirst report of AURORA, the breast international group (BIG) molecular screening initiative for metastatic breast cancer (MBC) patients (pts). Annals of Oncology 30, 2019 [Google Scholar]
- 11.Folprecht G, Aust DE, Roth A, et al. : Improving access to molecularly defined clinical trials for patients with colorectal cancer: The EORTC SPECTAcolor platform. Journal of Clinical Oncology 33:575–575, 2015. 25584004 [Google Scholar]
- 12.Gerber DE, Oxnard GR, Mandrekar SJ, et al. : ALCHEMIST: a clinical trial platform to bring genomic discovery and molecularly targeted therapies to early-stage lung cancer. Journal of Clinical Oncology 33:TPS7583–TPS7583, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Herbst RS, Gandara DR, Hirsch FR, et al. : Lung Master Protocol (Lung-MAP)-A Biomarker-Driven Protocol for Accelerating Development of Therapies for Squamous Cell Lung Cancer: SWOG S1400. Clin Cancer Res 21:1514–24, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Jhaveri KL, Makker V, Wang XV, et al. : Ado-trastuzumab emtansine (T-DM1) in patients (pts) with HER2 amplified (amp) tumors excluding breast and gastric/gastro-esophageal junction (GEJ) adenocarcinomas: Results from the National Cancer Institute (NCI) Molecular Analysis for Therapy Choice (MATCH) trial. Journal of Clinical Oncology 36:100–100, 2018 [Google Scholar]
- 15.Krop IE, Jegede O, Grilley-Olson JE, et al. : Results from molecular analysis for therapy choice (MATCH) arm I: Taselisib for PIK3CA-mutated tumors. Journal of Clinical Oncology 36:101–101, 2018. 29220288 [Google Scholar]
- 16.Azad N, Overman M, Gray R, et al. : Nivolumab in mismatch-repair deficient (MMR-d) cancers: NCI-MATCH Trial (Molecular Analysis for Therapy Choice. Journal for ImmunoTherapy of Cancer 89, 2017 [Google Scholar]
- 17.Chae YK, Vaklavas C, Cheng HH, et al. : Molecular analysis for therapy choice (MATCH) arm W: Phase II study of AZD4547 in patients with tumors with aberrations in the FGFR pathway. Journal of Clinical Oncology 36:2503–2503, 2018 [Google Scholar]
- 18.Joshi SS, Maron SB, Lomnicki S, et al. : Personalized antibodies for gastroesophageal adenocarcinoma (PANGEA): A phase II precision medicine trial (NCT02213289). Journal of Clinical Oncology 36:TPS198–TPS198, 2018 [Google Scholar]
- 19.Slosberg ED, Kang BP, Peguero J, et al. : Signature program: a platform of basket trials. Oncotarget 9:21383–21395, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Demetri GD, von Mehren M, Blanke CD, et al. : Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 347:472–80, 2002 [DOI] [PubMed] [Google Scholar]
- 21.Chapman PB, Hauschild A, Robert C, et al. : Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 364:2507–16, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Swain SM, Baselga J, Kim SB, et al. : Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med 372:724–34, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ramalingam SS, Vansteenkiste J, Planchard D, et al. : Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. The New England journal of medicine 382:41–50, 2020 [DOI] [PubMed] [Google Scholar]
- 24.Zehir A, Benayed R, Shah RH, et al. : Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 23:703–713, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Aisner DL, Sholl LM, Berry LD, et al. : The Impact of Smoking and TP53 Mutations in Lung Adenocarcinoma Patients with Targetable Mutations-The Lung Cancer Mutation Consortium (LCMC2). Clin Cancer Res 24:1038–1047, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Andre F, Bachelot T, Commo F, et al. : Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER). Lancet Oncol 15:267–74, 2014 [DOI] [PubMed] [Google Scholar]
- 27.Kris MG, Johnson BE, Berry LD, et al. : Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 311:1998–2006, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Schwaederle M, Parker BA, Schwab RB, et al. : Precision Oncology: The UC San Diego Moores Cancer Center PREDICT Experience. Mol Cancer Ther 15:743–52, 2016 [DOI] [PubMed] [Google Scholar]
- 29.Von Hoff DD, Stephenson JJ Jr., Rosen P, et al. : Pilot study using molecular profiling of patients’ tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol 28:4877–83, 2010 [DOI] [PubMed] [Google Scholar]
- 30.Stockley TL, Oza AM, Berman HK, et al. : Molecular profiling of advanced solid tumors and patient outcomes with genotype-matched clinical trials: the Princess Margaret IMPACT/COMPACT trial. Genome Med 8:109, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Tredan O, Wang Q, Pissaloux D, et al. : Molecular screening program to select molecular-based recommended therapies for metastatic cancer patients: analysis from the ProfiLER trial. Ann Oncol 30:757–765, 2019 [DOI] [PubMed] [Google Scholar]
- 32.Weidenbusch B, Richter GHS, Kesper MS, et al. : Transcriptome based individualized therapy of refractory pediatric sarcomas: feasibility, tolerability and efficacy. Oncotarget 9:20747–20760, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Worst BC, van Tilburg CM, Balasubramanian GP, et al. : Next-generation personalised medicine for high-risk paediatric cancer patients - The INFORM pilot study. Eur J Cancer 65:91–101, 2016 [DOI] [PubMed] [Google Scholar]
- 34.Oberg JA, Glade Bender JL, Sulis ML, et al. : Implementation of next generation sequencing into pediatric hematology-oncology practice: moving beyond actionable alterations. Genome Med 8:133, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Cieslik M, Chinnaiyan AM: Cancer transcriptome profiling at the juncture of clinical translation. Nat Rev Genet 19:93–109, 2018 [DOI] [PubMed] [Google Scholar]
- 36.Tsai C-H, Yao C-Y, Tien F-M, et al. : Incorporation of long non-coding RNA expression profile in the 2017 ELN risk classification can improve prognostic prediction of acute myeloid leukemia patients. EBioMedicine 40:240–250, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Heyer EE, Deveson IW, Wooi D, et al. : Diagnosis of fusion genes using targeted RNA sequencing. Nat Commun 10:1388, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kim P, Jang YE, Lee S: FusionScan: accurate prediction of fusion genes from RNA-Seq data. Genomics Inform 17:e26, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Vu TN, Deng W, Trac QT, et al. : A fast detection of fusion genes from paired-end RNA-seq data. BMC Genomics 19:786, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Winters JL, Davila JI, McDonald AM, et al. : Development and Verification of an RNA Sequencing (RNA-Seq) Assay for the Detection of Gene Fusions in Tumors. J Mol Diagn 20:495–511, 2018 [DOI] [PubMed] [Google Scholar]
- 41.Maher CA, Kumar-Sinha C, Cao X, et al. : Transcriptome sequencing to detect gene fusions in cancer. Nature 458:97–101, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Denoeud F, Aury J-M, Da Silva C, et al. : Annotating genomes with massive-scale RNA sequencing. Genome Biology 9:R175, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Garber M, Grabherr MG, Guttman M, et al. : Computational methods for transcriptome annotation and quantification using RNA-seq. Nature methods 8:469–477, 2011 [DOI] [PubMed] [Google Scholar]
- 44.Wang N, Zheng J, Chen Z, et al. : Single-cell microRNA-mRNA co-sequencing reveals non-genetic heterogeneity and mechanisms of microRNA regulation. Nat Commun 10:95, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Martinez-Gutierrez AD, Catalan OM, Vazquez-Romo R, et al. : miRNA profile obtained by nextgeneration sequencing in metastatic breast cancer patients is able to predict the response to systemic treatments. Int J Mol Med 44:1267–1280, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yu N, Yong S, Kim HK, et al. : Identification of tumor suppressor miRNAs by integrative miRNA and mRNA sequencing of matched tumor-normal samples in lung adenocarcinoma. Mol Oncol 13:1356–1368, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Szeto CY, Lin CH, Choi SC, et al. : Integrated mRNA and microRNA transcriptome sequencing characterizes sequence variants and mRNA-microRNA regulatory network in nasopharyngeal carcinoma model systems. FEBS Open Bio 4:128–40, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xie L, Yao Z, Zhang Y, et al. : Deep RNA sequencing reveals the dynamic regulation of miRNA, lncRNAs, and mRNAs in osteosarcoma tumorigenesis and pulmonary metastasis. Cell Death Dis 9:772, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Paik S, Shak S, Tang G, et al. : A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 351:2817–26, 2004 [DOI] [PubMed] [Google Scholar]
- 50.Krop I, Ismaila N, Andre F, et al. : Use of Biomarkers to Guide Decisions on Adjuvant Systemic Therapy for Women With Early-Stage Invasive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline Focused Update. J Clin Oncol 35:2838–2847, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cardoso F, van’t Veer LJ, Bogaerts J, et al. : 70-Gene Signature as an Aid to Treatment Decisions in Early-Stage Breast Cancer. N Engl J Med 375:717–29, 2016 [DOI] [PubMed] [Google Scholar]
- 52.Bridgewater J, van Laar R, Floore A, et al. : Gene expression profiling may improve diagnosis in patients with carcinoma of unknown primary. British Journal of Cancer 98:1425–1430, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Loriot Y, Gross-Goupil M, Lesimple T, et al. : Identifying the primary site using gene expression profiling in patients with carcinoma of an unknown primary (CUP): a feasibility study from the GEFCAPI. Journal of Clinical Oncology 27:e22168–e22168, 2009 [DOI] [PubMed] [Google Scholar]
- 54.Tothill RW, Shi F, Paiman L, et al. : Development and validation of a gene expression tumour classifier for cancer of unknown primary. Pathology 47:7–12, 2015 [DOI] [PubMed] [Google Scholar]
- 55.Wei IH, Shi Y, Jiang H, et al. : RNA-Seq accurately identifies cancer biomarker signatures to distinguish tissue of origin. Neoplasia (New York, N.Y.) 16:918–927, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Michuda J, Igartua C, Taxter T, et al. : Transcriptome-based cancer type prediction for tumors of unknown origin. Journal of Clinical Oncology 37:3081–3081, 2019. 31398081 [Google Scholar]
- 57.Perou CM, Sørlie T, Eisen MB, et al. : Molecular portraits of human breast tumours. Nature 406:747–752, 2000 [DOI] [PubMed] [Google Scholar]
- 58.Patel AP, Tirosh I, Trombetta JJ, et al. : Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science (New York, N.Y.) 344:1396–1401, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Li H, Courtois ET, Sengupta D, et al. : Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nature genetics 49:708–718, 2017 [DOI] [PubMed] [Google Scholar]
- 60.Parulkar R, Nguyen A, Sanborn JZ, et al. : Evidence for selective silencing of MHC-binding neoepitopes to avoid immune surveillance. Journal of Clinical Oncology 37:2591–2591, 2019 [Google Scholar]
- 61.Azizi E, Carr AJ, Plitas G, et al. : Single-Cell Map of Diverse Immune Phenotypes in the Breast Tumor Microenvironment. Cell 174:1293–1308.e36, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Savas P, Virassamy B, Ye C, et al. : Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis. Nature medicine 24:986–993, 2018 [DOI] [PubMed] [Google Scholar]
- 63.Adashek JJ, Kato S, Parulkar R, et al. : Transcriptomic silencing as a potential mechanism of treatment resistance. JCI Insight 5, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Santarius T, Shipley J, Brewer D, et al. : A census of amplified and overexpressed human cancer genes. Nature reviews. Cancer 10:59–64, 2010 [DOI] [PubMed] [Google Scholar]
- 65.Stein WD, Litman T, Fojo T, et al. : A Serial Analysis of Gene Expression (SAGE) database analysis of chemosensitivity: comparing solid tumors with cell lines and comparing solid tumors from different tissue origins. Cancer Res 64:2805–16, 2004 [DOI] [PubMed] [Google Scholar]
- 66.Bittner M, Meltzer P, Chen Y, et al. : Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature 406:536–40, 2000 [DOI] [PubMed] [Google Scholar]
- 67.Ramaswamy S, Ross KN, Lander ES, et al. : A molecular signature of metastasis in primary solid tumors. Nat Genet 33:49–54, 2003 [DOI] [PubMed] [Google Scholar]
- 68.Ntzani EE, Ioannidis JP: Predictive ability of DNA microarrays for cancer outcomes and correlates: an empirical assessment. Lancet 362:1439–44, 2003 [DOI] [PubMed] [Google Scholar]
- 69.Clough E, Barrett T: The Gene Expression Omnibus Database. Methods in molecular biology (Clifton, N.J.) 1418:93–110, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Adetunji MO, Lamont SJ, Schmidt CJ: TransAtlasDB: an integrated database connecting expression data, metadata and variants. Database (Oxford) 2018, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sparano JA, Gray RJ, Makower DF, et al. : Adjuvant Chemotherapy Guided by a 21-Gene Expression Assay in Breast Cancer. N Engl J Med 379:111–121, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Frith MC, Pheasant M, Mattick JS: The amazing complexity of the human transcriptome. European journal of human genetics : EJHG 13:894–897, 2005 [DOI] [PubMed] [Google Scholar]
- 73.Boccaletto P, Machnicka MA, Purta E, et al. : MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Research 46:D303–D307, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Alexandrov LB, Nik-Zainal S, Wedge DC, et al. : Signatures of mutational processes in human cancer. Nature 500:415–421, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Avesson L, Barry G: The emerging role of RNA and DNA editing in cancer. Biochimica et biophysica acta 1845:308–316, 2014 [DOI] [PubMed] [Google Scholar]
- 76.Cui Q, Shi H, Ye P, et al. : m(6)A RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells. Cell reports 18:2622–2634, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Li Z, Weng H, Su R, et al. : FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N(6)-Methyladenosine RNA Demethylase. Cancer cell 31:127–141, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Vareslija D, Priedigkeit N, Fagan A, et al. : Transcriptome Characterization of Matched Primary Breast and Brain Metastatic Tumors to Detect Novel Actionable Targets. J Natl Cancer Inst 111:388–398, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Smith JL, Ries RE, Hylkema T, et al. : Comprehensive Transcriptome Profiling of Cryptic CBFA2T3-GLIS2 Fusion-positive AML Defines Novel Therapeutic Options - A COG and TARGET Pediatric AML Study. Clin Cancer Res, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Buffa FM, Camps C, Winchester L, et al. : microRNA-associated progression pathways and potential therapeutic targets identified by integrated mRNA and microRNA expression profiling in breast cancer. Cancer research 71:5635–5645, 2011 [DOI] [PubMed] [Google Scholar]
- 81.Jacobsen A, Silber J, Harinath G, et al. : Analysis of microRNA-target interactions across diverse cancer types. Nature structural & molecular biology 20:1325–1332, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Dhawan A, Scott JG, Harris AL, et al. : Pan-cancer characterisation of microRNA across cancer hallmarks reveals microRNA-mediated downregulation of tumour suppressors. Nature Communications 9:5228, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Czech MP: MicroRNAs as therapeutic targets. The New England journal of medicine 354:1194–1195, 2006 [DOI] [PubMed] [Google Scholar]
- 84.Roccaro AM, Sacco A, Thompson B, et al. : MicroRNAs 15a and 16 regulate tumor proliferation in multiple myeloma. Blood 113:6669–6680, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Hayashita Y, Osada H, Tatematsu Y, et al. : A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer research 65:9628–9632, 2005 [DOI] [PubMed] [Google Scholar]
- 86.Korpal M, Lee ES, Hu G, et al. : The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. The Journal of biological chemistry 283:14910–14914, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.He L, He X, Lim LP, et al. : A microRNA component of the p53 tumour suppressor network. Nature 447:1130–1134, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lu J, Getz G, Miska EA, et al. : MicroRNA expression profiles classify human cancers. Nature 435:834–8, 2005 [DOI] [PubMed] [Google Scholar]
- 89.Fountzilas E, Kelly AD, Perez-Atayde AR, et al. : A microRNA activity map of human mesenchymal tumors: connections to oncogenic pathways; an integrative transcriptomic study. BMC genomics 13:332–332, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yanaihara N, Caplen N, Bowman E, et al. : Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 9:189–98, 2006 [DOI] [PubMed] [Google Scholar]
- 91.Foekens JA, Sieuwerts AM, Smid M, et al. : Four miRNAs associated with aggressiveness of lymph node-negative, estrogen receptor-positive human breast cancer. Proc Natl Acad Sci U S A 105:13021–6, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lamichhane SR, Thachil T, De Ieso P, et al. : Prognostic Role of MicroRNAs in Human Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. Disease markers 2018:8309015–8309015, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hu S, Yuan Y, Song Z, et al. : Expression Profiles of microRNAs in Drug-Resistant Non-Small Cell Lung Cancer Cell Lines Using microRNA Sequencing. Cellular Physiology and Biochemistry 51:2509–2522, 2018 [DOI] [PubMed] [Google Scholar]
- 94.Ma T, Yang L, Zhang J: MiRNA-542-3p downregulation promotes trastuzumab resistance in breast cancer cells via AKT activation. Oncology reports 33:1215–1220, 2015 [DOI] [PubMed] [Google Scholar]
- 95.Wagenseller AG, Shada A, D’Auria KM, et al. : MicroRNAs induced in melanoma treated with combination targeted therapy of Temsirolimus and Bevacizumab. Journal of translational medicine 11:218–218, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hannafon BN, Ding W-Q: Intercellular communication by exosome-derived microRNAs in cancer. International journal of molecular sciences 14:14240–14269, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Tran N: Cancer Exosomes as miRNA Factories. Trends in cancer 2:329–331, 2016 [DOI] [PubMed] [Google Scholar]
- 98.Anfossi S, Babayan A, Pantel K, et al. : Clinical utility of circulating non-coding RNAs - an update. Nature reviews. Clinical oncology 15:541–563, 2018 [DOI] [PubMed] [Google Scholar]
- 99.Mercer TR, Dinger ME, Mattick JS: Long non-coding RNAs: insights into functions. Nature reviews. Genetics 10:155–159, 2009 [DOI] [PubMed] [Google Scholar]
- 100.Cabili MN, Trapnell C, Goff L, et al. : Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes & development 25:1915–1927, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Liu L, Meng T, Yang X-H, et al. : Prognostic and predictive value of long non-coding RNA GAS5 and mircoRNA-221 in colorectal cancer and their effects on colorectal cancer cell proliferation, migration and invasion. Cancer biomarkers : section A of Disease markers 22:283–299, 2018 [DOI] [PubMed] [Google Scholar]
- 102.Yuan T, Huang X, Woodcock M, et al. : Plasma extracellular RNA profiles in healthy and cancer patients. Scientific reports 6:19413–19413, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Zhang R, Xia Y, Wang Z, et al. : Serum long non coding RNA MALAT-1 protected by exosomes is up-regulated and promotes cell proliferation and migration in non-small cell lung cancer. Biochemical and biophysical research communications 490:406–414, 2017 [DOI] [PubMed] [Google Scholar]
- 104.Nilsson J, Skog J, Nordstrand A, et al. : Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer. British journal of cancer 100:1603–1607, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Xiao Y, Yurievich UA, Yosypovych SV: Long noncoding RNA XIST is a prognostic factor in colorectal cancer and inhibits 5-fluorouracil-induced cell cytotoxicity through promoting thymidylate synthase expression. Oncotarget 8:83171–83182, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Li L, Shang J, Zhang Y, et al. : MEG3 is a prognostic factor for CRC and promotes chemosensitivity by enhancing oxaliplatin-induced cell apoptosis. Oncology reports 38:1383–1392, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Godinho MFE, Sieuwerts AM, Look MP, et al. : Relevance of BCAR4 in tamoxifen resistance and tumour aggressiveness of human breast cancer. British Journal of Cancer 103:1284–1291, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Shi S-J, Wang L-J, Yu B, et al. : LncRNA-ATB promotes trastuzumab resistance and invasion-metastasis cascade in breast cancer. Oncotarget 6:11652–11663, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Tang H, Wang S, Xiao G, et al. : Comprehensive evaluation of published gene expression prognostic signatures for biomarker-based lung cancer clinical studies. Annals of oncology : official journal of the European Society for Medical Oncology 28:733–740, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Liu MC, Pitcher BN, Mardis ER, et al. : PAM50 gene signatures and breast cancer prognosis with adjuvant anthracycline- and taxane-based chemotherapy: correlative analysis of C9741 (Alliance). NPJ breast cancer 2:15023, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Maak M, Simon I, Nitsche U, et al. : Independent validation of a prognostic genomic signature (ColoPrint) for patients with stage II colon cancer. Annals of surgery 257:1053–1058, 2013 [DOI] [PubMed] [Google Scholar]
- 112.Srivastava G, Renfro LA, Behrens RJ, et al. : Prospective multicenter study of the impact of oncotype DX colon cancer assay results on treatment recommendations in stage II colon cancer patients. The oncologist 19:492–497, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Schwartz GW, Petrovic J, Zhou Y, et al. : Differential Integration of Transcriptome and Proteome Identifies Pan-Cancer Prognostic Biomarkers. Front Genet 9:205, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Smyth EC, Nyamundanda G, Cunningham D, et al. : A seven-Gene Signature assay improves prognostic risk stratification of perioperative chemotherapy treated gastroesophageal cancer patients from the MAGIC trial. Annals of oncology : official journal of the European Society for Medical Oncology 29:2356–2362, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Wang B, Wan F, Sheng H, et al. : Identification and validation of an 18-gene signature highly-predictive of bladder cancer metastasis. Scientific Reports 8:374, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Fountzilas E, Markou K, Vlachtsis K, et al. : Identification and validation of gene expression models that predict clinical outcome in patients with early-stage laryngeal cancer. Annals of oncology : official journal of the European Society for Medical Oncology 23:2146–2153, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Konstantinopoulos PA, Cannistra SA, Fountzilas H, et al. : Integrated analysis of multiple microarray datasets identifies a reproducible survival predictor in ovarian cancer. PloS one 6:e18202–e18202, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Bertucci F, Ueno NT, Finetti P, et al. : Gene expression profiles of inflammatory breast cancer: correlation with response to neoadjuvant chemotherapy and metastasis-free survival. Annals of oncology : official journal of the European Society for Medical Oncology 25:358–365, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Cao B, Luo L, Feng L, et al. : A network-based predictive gene-expression signature for adjuvant chemotherapy benefit in stage II colorectal cancer. BMC cancer 17:844–844, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Xu Q, Chen J, Ni S, et al. : Pan-cancer transcriptome analysis reveals a gene expression signature for the identification of tumor tissue origin. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 29:546–556, 2016 [DOI] [PubMed] [Google Scholar]
- 121.Hayashi H, Kurata T, Takiguchi Y, et al. : Randomized Phase II Trial Comparing Site-Specific Treatment Based on Gene Expression Profiling With Carboplatin and Paclitaxel for Patients With Cancer of Unknown Primary Site. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 37:570–579, 2019 [DOI] [PubMed] [Google Scholar]
- 122.Fizazi K, Maillard A, Penel N, et al. : A phase 3 trial of empiric chemotherapy with cisplatin and gemcitabine or systemic treatment tailored by molecular gene expression analysis in patients with carcinomas of an unknown primary (CUP) site (GEFCAPI 04), ESMO 2019 Congress, Annals of Oncology, 2019, pp v851–v934 [Google Scholar]
- 123.Gerlinger M, Rowan AJ, Horswell S, et al. : Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. The New England journal of medicine 366:883–892, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Norton N, Advani PP, Serie DJ, et al. : Assessment of Tumor Heterogeneity, as Evidenced by Gene Expression Profiles, Pathway Activation, and Gene Copy Number, in Patients with Multifocal Invasive Lobular Breast Tumors. PloS one 11:e0153411–e0153411, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Fan J, Lee H-O, Lee S, et al. : Linking transcriptional and genetic tumor heterogeneity through allele analysis of single-cell RNA-seq data. Genome research 28:1217–1227, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hong SP, Chan TE, Lombardo Y, et al. : Single-cell transcriptomics reveals multi-step adaptations to endocrine therapy. Nature Communications 10:3840, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Levitin HM, Yuan J, Sims PA: Single-Cell Transcriptomic Analysis of Tumor Heterogeneity. Trends in cancer 4:264–268, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Goodman AM, Piccioni D, Kato S, et al. : Prevalence of PDL1 Amplification and Preliminary Response to Immune Checkpoint Blockade in Solid Tumors. JAMA oncology 4:1237–1244, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Le DT, Uram JN, Wang H, et al. : PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. The New England journal of medicine 372:2509–2520, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Goodman AM, Kato S, Bazhenova L, et al. : Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Molecular cancer therapeutics 16:2598–2608, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Goodman AM, Sokol ES, Frampton GM, et al. : Microsatellite-Stable Tumors with High Mutational Burden Benefit from Immunotherapy. Cancer immunology research 7:1570–1573, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zaretsky JM, Garcia-Diaz A, Shin DS, et al. : Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. The New England journal of medicine 375:819–829, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hugo W, Zaretsky JM, Sun L, et al. : Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 165:35–44, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Peng W, Chen JQ, Liu C, et al. : Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer discovery 6:202–216, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Danaher P, Warren S, Dennis L, et al. : Gene expression markers of Tumor Infiltrating Leukocytes. Journal for immunotherapy of cancer 5:18–18, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Lee W-C, Diao L, Wang J, et al. : Multiregion gene expression profiling reveals heterogeneity in molecular subtypes and immunotherapy response signatures in lung cancer. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 31:947–955, 2018 [DOI] [PubMed] [Google Scholar]
- 137.Riaz N, Havel JJ, Makarov V, et al. : Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell 171:934–949.e16, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Mandal R, Şenbabaoğlu Y, Desrichard A, et al. : The head and neck cancer immune landscape and its immunotherapeutic implications. JCI insight 1:e89829–e89829, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Groenenboom MAC, Marée AFM, Hogeweg P: The RNA silencing pathway: the bits and pieces that matter. PLoS computational biology 1:155–165, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Vainio P, Mpindi J-P, Kohonen P, et al. : High-throughput transcriptomic and RNAi analysis identifies AIM1, ERGIC1, TMED3 and TPX2 as potential drug targets in prostate cancer. PloS one 7:e39801–e39801, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Golan T, Khvalevsky EZ, Hubert A, et al. : RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget 6:24560–24570, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Adashek JJ, Kato S, Parulkar R, et al. : RNAseq in addition to next generation sequencing in advanced genitourinary cancers reveals transcriptomic silencing of DNA mutations: Implications for resistance to targeted therapeutics. Journal of Clinical Oncology 37:583–583, 2019 [Google Scholar]
- 143.Chen Y, McGee J, Chen X, et al. : Identification of druggable cancer driver genes amplified across TCGA datasets. PloS one 9:e98293–e98293, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Caburet S, Anttonen M, Todeschini A-L, et al. : Combined comparative genomic hybridization and transcriptomic analyses of ovarian granulosa cell tumors point to novel candidate driver genes. BMC cancer 15:251–251, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Consortium GT, Laboratory DA, Coordinating Center —Analysis Working G, et al. : Genetic effects on gene expression across human tissues. Nature 550:204–213, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Li X, Kim Y, Tsang EK, et al. : The impact of rare variation on gene expression across tissues. Nature 550:239–243, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Kourou K, Exarchos TP, Exarchos KP, et al. : Machine learning applications in cancer prognosis and prediction. Computational and structural biotechnology journal 13:8–17, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Levine AB, Schlosser C, Grewal J, et al. : Rise of the Machines: Advances in Deep Learning for Cancer Diagnosis. Trends in cancer 5:157–169, 2019 [DOI] [PubMed] [Google Scholar]
- 149.Adamson AS, Welch HG: Machine Learning and the Cancer-Diagnosis Problem - No Gold Standard. The New England journal of medicine 381:2285–2287, 2019 [DOI] [PubMed] [Google Scholar]
- 150.van Tilburg CM, Pfaff E, Pajtler KW, et al. : The pediatric precision oncology study INFORM: Clinical outcome and benefit for molecular subgroups. Journal of Clinical Oncology 38:LBA10503–LBA10503, 2020 [Google Scholar]
- 151.Wagle N, Berger MF, Davis MJ, et al. : High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. Cancer discovery 2:82–93, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Sinicropi D, Qu K, Collin F, et al. : Whole transcriptome RNA-Seq analysis of breast cancer recurrence risk using formalin-fixed paraffin-embedded tumor tissue. PLoS One 7:e40092, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Nieuwenhuis TO, Yang SY, Verma RX, et al. : Consistent RNA sequencing contamination in GTEx and other data sets. Nature Communications 11:1933, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Ong C-AJ, Tan QX, Lim HJ, et al. : An Optimised Protocol Harnessing Laser Capture Microdissection for Transcriptomic Analysis on Matched Primary and Metastatic Colorectal Tumours. Scientific Reports 10:682, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Shen Q, Hu J, Jiang N, et al. : contamDE: differential expression analysis of RNA-seq data for contaminated tumor samples. Bioinformatics 32:705–712, 2015 [DOI] [PubMed] [Google Scholar]
- 156.Ramalingam N, Lee Y, Szpankowski L, et al. : Abstract LB-326: Full-Length mRNA transcriptome analysis of matched circulating tumor and immune cells from breast cancer subjects. Cancer Research 79:LB-326–LB-326, 2019 [Google Scholar]
- 157.Chung W, Eum HH, Lee H-O, et al. : Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer. Nature communications 8:15081–15081, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Li B, Cui Y, Diehn M, et al. : Development and Validation of an Individualized Immune Prognostic Signature in Early-Stage Nonsquamous Non-Small Cell Lung Cancer. JAMA oncology 3:1529–1537, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]