Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2020

Open Access 01.12.2020 | Review

RNA sequencing: new technologies and applications in cancer research

verfasst von: Mingye Hong, Shuang Tao, Ling Zhang, Li-Ting Diao, Xuanmei Huang, Shaohui Huang, Shu-Juan Xie, Zhen-Dong Xiao, Hua Zhang

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2020

Abstract

Over the past few decades, RNA sequencing has significantly progressed, becoming a paramount approach for transcriptome profiling. The revolution from bulk RNA sequencing to single-molecular, single-cell and spatial transcriptome approaches has enabled increasingly accurate, individual cell resolution incorporated with spatial information. Cancer, a major malignant and heterogeneous lethal disease, remains an enormous challenge in medical research and clinical treatment. As a vital tool, RNA sequencing has been utilized in many aspects of cancer research and therapy, including biomarker discovery and characterization of cancer heterogeneity and evolution, drug resistance, cancer immune microenvironment and immunotherapy, cancer neoantigens and so on. In this review, the latest studies on RNA sequencing technology and their applications in cancer are summarized, and future challenges and opportunities for RNA sequencing technology in cancer applications are discussed.
Hinweise
Mingye Hong and Shuang Tao have contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
RNA-seq
RNA sequencing
ScRNA-seq
Single-cell RNA sequencing
SOLiD
Sequencing by Oligonucleotide Ligation and Detection
DNBS
DNA nanoball sequencing
NGS
Next-generation sequencing
PacBio SMRT
Pacific Biosciences single-molecule real-time
ONT
Oxford Nanopore Technologies
Sci-RNA-seq
Single-cell combinatorial indexing RNA sequencing
SPLiT-seq
Split-pool ligation-based transcriptome sequencing
LCM
Laser capture microdissection
FISSEQ
Fluorescent in situ RNA sequencing
RCA
Rolling-circle amplification
AML
Acute myeloid leukemia
SSRs
Simple repetitive sequences
COX2
Cyclooxygenase2
HER2
Human epidermal growth factor receptor type 2
ISG15
Interferon-stimulated gene 15
DMGDH
Dimethylglycine dehydrogenase
TMB
Tumor mutation burden
IDH
Isocitrate dehydrogenase
CNV
Copy number variations
TEPs
Tumor-educated blood platelets
SnRNA-seq
Single-nucleus RNA-sequencing
BORIS
Brother of the regulator of imprinted sites
ALL
Acute lymphocytic leukemia
LSCs
Leukemia stem cells
TILs
Tumor-infiltrating lymphocytes
TME
Tumor microenvironment
NFATC1
Nuclear factor of activated T cells 1
TCR
T cell receptor
HCC
Hepatocellular carcinoma
LAP
Microtubule-associated protein 1A/1B-light chain 3-associated phagocytosis
CSF3R
Colony stimulating factor 3 receptor
UTCαβ
Unconventional CD4CD8 αβ T cells
DCs
Dendritic cells
SOCS2
Suppressor-of-cytokine-2 protein
ICB
Immune checkpoint blockade
PD1
Targeting programmed death 1
PD-L1
Programmed death-ligand 1
CTLA4
Cytotoxic T-lymphocyte-associated protein 4
CTL
Cytotoxic T lymphocytes
Sca-1
Stem cell antigen-1
TAMs
Tumor-associated macrophages
CSF1R
Colony stimulating factor 1 receptor
INs-seq
Intracellular staining and sequencing
Mreg
Regulatory myeloid cells
HLA
Human leukocyte antigen
SGC
Salivary gland carcinoma
CCS
Circular consensus-sequencing
ATAC-seq
Assay for Transposase-Accessible Chromatin using sequencing
TF
Transcriptional factor
3C
Chromatin conformation capture analysis
4C
Circular chromosome conformation capture
5C
Carbon copy chromosome conformation capture
m6A
N6 position
m5C
5-Methylcytosine
CRISPR
Clustered regularly interspaced short palindromic repeats

Background

Cancer remains one of the major malignant diseases that endangers human life and health and comprises complex biological systems that require accurate and comprehensive analysis. Since the first appearance of high-throughput sequencing in 2005 [1], it has become possible to understand life activities at the molecular level and to conduct detailed research to elucidate the genome and transcriptome. As an essential part of high-throughput sequencing, RNA sequencing (RNA-seq), especially single-cell RNA sequencing (scRNA-seq), provides biological information on a single tumor cell, analyzes the determinants of intratumor expression heterogeneity and identifies the molecular basis of formation of many oncological diseases [2, 3]. Thus, RNA sequencing offers invaluable insights for cancer research and treatment. With the advent of the era of precision medicine, RNA sequencing will be widely used for research on many different types of cancer. This review summarizes the history of the development of RNA sequencing and focuses on the latest studies of RNA sequencing technology in cancer applications, especially single-cell RNA sequencing and spatial transcriptome sequencing. In addition, we provide a general introduction to the current bioinformatics analysis tools used for RNA sequencing and discuss future challenges and opportunities for RNA sequencing technology in cancer applications.

The development of RNA sequencing technologies

It was not until 1953 when Watson and Crick proposed the double-helix structure did people truly realize at the molecular level that the essence of life is the result of gene interactions [4]. The continuous development of RNA sequencing has ushered transcriptome analysis into a new era, with higher efficiency and lower cost. The timeline of RNA sequencing technologies is shown in Fig. 1.
The first-generation sequencing technology is also called Sanger sequencing. The chain termination method was initiated by Sanger in 1977, followed by the chemical degradation method developed by Maxam and Gilbert [5, 6]. The same year, Sanger determined the 5368 bp genome of phage φX174, which is the first DNA genome sequenced [7]. The DNA microarray has aided significant progress in many fields since it was first introduced. However, microarrays require prior knowledge of gene sequences and are unable to identify novel gene expression [8]. After the first high-throughput sequencing platform appeared in 2005 [1], multiple next-generation sequencing platforms followed (Table 1, Figs. 2, 3). The accuracy and reproducibility among different platforms depended on several factors, including the inherent features of the platform and the corresponding analysis pipelines [9, 10]. Pyrosequencing that was no longer supported after 2016, developed by 454 Life Sciences, used a “sequencing by synthesis” method [1, 1113]. The ion torrent sequencing platform is also based on the “sequencing by synthesis” method, which outperforms pyrosequencing with respect to sensitivity. SOLiD (Sequencing by Oligonucleotide Ligation and Detection) exhibits high accuracy, as each base is sequenced twice, but the read length is short [1113]. DNBS (DNA nanoball sequencing) enables large collection of DNA nanoballs for simultaneous sequencing. Illumina-based sequencing technology represents a “reversible terminator sequencing” method. High-throughput sequencing has the advantage of fast speed, low sequencing cost and high accuracy, otherwise known as next-generation sequencing (NGS). Compared to microarray, it can detect unknown gene expression sequences but is time intensive [14].
Table 1
Comparison of different RNA sequencing platforms
Platform
Company
Read length(bases)
Run time
Volume per run
Cost
Template preparation
Sequencing chemistry
The first-generation sequencing
Sanger
Life sciences
800 bp
2 h
1 read
$2400 per million bases
Bacterial cloning
Dideoxynucleosides terminator
The next-generation sequencing
Roche 454 pyrosequencing
454 Life sciences
700 bp
 < 24 h
0.7 Gb
$10 per million bases
Emulsion PCR
Sequencing by synthesis, pyrosequencing
Illumina HiSeq
Illumina
100 bp
3–10 days
120–1500 Gb
$0.02—$0.07 per million bases
Bridge PCR
Reversible terminator sequencing
Illumina MiSeq
Illumina
100 bp
1–2 days
0.3–15 Gb
$0.13 per million bases
Bridge PCR
Reversible terminator sequencing
SOLiD
Applied biosystems instruments (ABI)
50–75 bp
7–14 days
30 Gb
$0.13 per million bases
Emulsion PCR
Sequencing by ligation
DNA nanoball sequencing
Complete genomics
440–500 bp
9 days
20–60 Gb
$4400/genome
Rolling circle replication
Hybridization and ligation
Ion torrent
454 Life sciences
200–500 bp
4–5 h
660 Mb; 11 Mb
$300 to $750 per run
Emulsion PCR
Sequencing by synthesis
The third-generation sequencing
SMRT
Pacific biosciences
 > 900 bp
1-2 h
0.5–1 Gb
$2 per million bases
No need
Sequencing by synthesis
Helicos sequencing
Helicos biosciences
25–60 bp
8 days
21–35 Gb
$0.01 per million bases
No need
Hybridization and synthesis
Nanopore sequencing
Oxford nanopore technologies
Up to 98 kb
48/72 h
Up to 30 Gb
 < $1 per million bases
No need
Nanopore
In addition to NGS, there is third-generation sequencing, which allows for long-read sequencing of individual RNA molecules [15]. Single-molecule RNA sequencing enables the generation of full-length cDNA transcripts without clonal amplification or transcript assembly. Thus, third-generation sequencing is free from the shortcomings generated by PCR amplification and read mapping. It can greatly reduce the false positive rate of splice sites and capture the diversity of transcript isoforms [15]. Single-molecule sequencing platforms comprise Pacific Biosciences (PacBio) single-molecule real-time (SMRT) sequencing [16], Helicos single-molecule fluorescent sequencing [17] and Oxford Nanopore Technologies (ONT) nanopore sequencing [18]. Furthermore, RNA-seq recently evolved from bulk sequencing to single-cell sequencing. Single-cell RNA sequencing was first published in 2009 to profile the transcriptome at single-cell resolution [19]. Drop-Seq and InDrop were initially reported in 2015 by analyzing mouse retina cell and embryonic stem cell transcriptomes, identifying novel cell types. Sci-RNA-seq, single-cell combinatorial indexing RNA sequencing, was developed in 2017, and SPLiT-seq (split-pool ligation-based transcriptome sequencing) was first reported in 2018. Both approaches use a combinatorial indexing strategy in which attached RNAs are labeled with barcodes that indicate their cellular origin [20, 21].
Though single-cell data enable single-cell transcriptomics, it may lose spatial information during single-cell isolation. To solve this problem, spatial transcriptomics has emerged. Spatial transcriptomics employs unique positional barcodes to visualize RNA distributions in RNA sequencing of tissue sections and was first published in 2016 [22]. Slide-seq, reported in 2019, uses DNA barcode beads with specific positional information [23]. Geo-seq was introduced in 2017 and integrated scRNA-seq with laser capture microdissection (LCM), which can isolate individual cells [24]. In situ sequencing refers to targeted sequencing of RNA fragments in morphologically preserved tissues or cells without RNA extraction, including in situ cDNA synthesis by padlock probes or stably cross-linked cDNA amplicons in fluorescent in situ RNA sequencing (FISSEQ) and in situ amplification by rolling-circle amplification (RCA) [25, 26]. Furthermore, various new technologies based on RNA-seq have been developed for specific applications. For example, a type of targeted RNA sequencing, CaptureSeq, employs biotinylated oligonucleotide probes and results in the enrichment of certain transcripts to identify gene fusion [27, 28].

Computational analysis of RNA sequencing data

Computational analysis tools for RNA sequencing have dramatically increased during the past decade. The choice of a particular tool should be based on the purpose and accuracy of application [2931]. A general RNA sequencing data analysis process involves the quality control of raw data, read alignment and transcript assembly, expression quantification and differential expression analysis (Fig. 4).
The first step of data analysis is to assess and clean the raw sequencing data, which is usually provided in the form of FASTQ files [32]. Quality control visually reflects the quality of the sequencing and purposefully discards low-quality reads, eliminates poor-quality bases and trims adaptor sequences [31]. Common tools include FASTQ [33], NGSQC [34], RNA-SeQC [35], Trimmomatic [36], PRINSEQ [37] and Soapnuke [38].
The next step is to map the clean reads to either a genome or a transcriptome. There are some mapping tools available, including Tophat2 [39], HISAT2 [40], STAR [41], BWA [42] and Bowtie [43]. After alignment, another type of software, such as Cufflinks [44], StringTie [45], Trinity [46], SOAPdenovoTrans [47] and Trans-AByS [48] can be used to assemble transcripts from short-reads. When the transcript model is established, its expression can be quantified at the gene, transcript and exon levels. Commonly used software for gene-level quantification includes FeatureCount [49] and HTSeq-count [50]. Transcript level quantitative software includes Cufflinks [44], eXpress [51] and RSEM [52]. DEXSeq is a software for exon level quantification [53]. In addition, there are some alignment-free quantification tools such as Kallisto [54], Sailfish [55] and Salmon [56], which have the advantage of marked computational resource saving. After normalizing, an expression matrix is generated, and statistical methods can be used to identify differentially expressed genes. DESeq2 [57] and edgeR [58] are commonly used to perform this task.

Applications of RNA-sequencing in cancer research

Genomic data, such as RNA-seq, have become widely available due to the popularity of high-throughput sequencing technology [59]. As an important part of next-generation sequencing, RNA sequencing has made great contributions in various fields, especially cancer research, including studies on differential gene expression analysis and cancer biomarkers, cancer heterogeneity and evolution, cancer drug resistance, the cancer microenvironment and immunotherapy, neoantigens, etc. (Fig. 5).

Differential gene expression analysis and cancer biomarkers

Differential gene expression analysis is one of the most common applications of RNA sequencing [60]. Samples from different backgrounds (different species, tissues and periods) can be used for RNA sequencing to identify differentially expressed genes, revealing their function and potential molecular mechanisms [61]. More importantly, differential gene expression analysis facilitates the discovery of potential cancer biomarkers [62]. Many studies have shown that gene fusions are closely related to oncogenesis and are appreciated as both ideal cancer biomarkers and therapeutic targets [63]. Gene fusions in clinical samples are primarily detected by RNA-CaptureSeq. Compared to whole transcriptome sequencing, RNA-CaptureSeq has significantly higher sequencing depth [27, 64, 65]. It has been reported that the NUP98-PHF23 fusion gene is likely to be a novel therapeutic target in acute myeloid leukemia (AML) [66]. Recently, a variety of recurrent gene fusions, including ESR1-CCDC170, SEC16A-NOTCH1, SEC22B-NOTCH2 and ESR1-YAP1, have been identified in breast cancer, indicating that recurrent gene fusion is one of the key drivers for cancer [67]. Several novel configurations of  BRAF, NTRK3 and RET gene fusions have been identified in colorectal cancer [68]. These fusions may promote the development of malignancy and provide new targets for personalized treatment [68]. In addition, some special genomic factors have been discovered as biomarkers by RNA sequencing, including miRNA, lncRNA and circRNA, which are widely present in various types of cancer [6971]. A recent example is circRNA_0001178 and circRNA_0000826, which are biomarkers of colorectal cancer metastasis to the liver [72]. By applying both RNA sequencing and small RNA sequencing, a study on pancreatic cancer identified differential expression of simple repetitive sequences (SSRs) and demonstrated that the frequency of SSR motifs changed dramatically, which is expected to become a tumor biomarker [73]. In addition to nucleic acid biomarkers, RNA-seq combined with immunohistochemistry and western blot has also identified certain proteins as cancer biomarkers, such as nuclear COX2 (cyclooxygenase2) in combination with HER2 (human epidermal growth factor receptor type 2), which may serve as potential biomarkers for the diagnosis and prognosis of colorectal cancer [74]. Similar examples identified using RNA-seq profiling analysis include ISG15 (Interferon-stimulated gene 15) in nasopharyngeal carcinoma [75] and DMGDH (dimethylglycine dehydrogenase) in hepatocellular carcinoma [76]. Data-mining analysis of RNA sequencing data and other clinical data has identified that the isoforms of peroxiredoxins also can be expected the prognostic biomarkers for predicting overall survival and relapse-free survival in breast cancer [77]. Increasing differentially expressed genes are being identified by RNA sequencing, and new potential cancer biomarkers are being continuously discovered (Table 2). However, sufficient clinical practice is needed to confirm the diagnostic and predictive applications of these biomarkers in cancer.
Table 2
Representative potential biomarkers identified by RNA-seq in cancer
Cancer type
Biomarker name
Biomarker type
Up/Down
Value
References
Liver cancer
tRNA-ValTAC-3/tRNA-GlyTCC-5/tRNA-ValAAC-5/tRNA-GluCTC-5
tsRNA
Up
Diagnostic
[92]
 
ACVR2B-AS1
LncRNA
Up
Prognostic/therapeutic target
[93]
Lung cancer
LINC01537
LncRNA
Down
Prognostic/therapeutic target
[94]
circFARSA
CircRNA
Up
Noninvasive biomarker
[95]
LINC01123
LncRNA
Up
Prognostic/therapeutic target
[96]
Gastric cancer
CTD2510F5.4
LncRNA
Up
Diagnostic/prognostic
[97]
MEF2C-AS1/FENDRR
LncRNA
Down
Diagnostic/prognostic
[98]
Prostate cancer
PSLNR
LncRNA
Down
Diagnostic/therapeutic target
[99]
Colorectal cancer
RAMS11
LncRNA
Up
Therapeutic target
[100]
CRCAL-1/CRCAL-2 /CRCAL-3/ CRCAL-4
LncRNA
Up
Therapeutic target
[101]
Colon cancer
AFAP1-AS1
LncRNA
Up
Prognostic/ therapeutic target
[102]
Head and neck squamous cell carcinoma
LINC00460
LncRNA
Up
Prognostic
[103]
HCG22
LncRNA
Down
Prognostic
[104]
HOXA11-AS/LINC00964/MALAT1
LncRNA
Up
Diagnostic
[105]
Clear cell renal cell carcinomas
SLINKY
LncRNA
Up
Prognostic
[106]
Leukemia
LUCAT1
LncRNA
Up
Therapeutic target
[107]
circ-HIPK2
CircRNA
Down
Diagnostic/prognostic
[108]
RNA-seq could detect early mutations as well as high molecular risk mutations, thus can discover novel cancer biomarkers and potential therapeutic targets, monitoring of diseases and guiding targeted therapy during early treatment decisions. Tumor mutation burden (TMB) is considered as a potential biomarker for immune checkpoint therapy and prognosis [78, 79]. RNA-seq can be used to explore the application value of TMB in diffuse glioma [78]. Through the RNA-seq, MET exon 14 mutation and isocitrate dehydrogenase 1 (IDH1) mutation were identified as new potential therapeutic targets in lung adenocarcinoma and chondrosarcoma patients, respectively [80, 81]. Several studies have shown that RNA sequencing can effectively improve the detection rate on the basis of DNA sequencing, provide more comprehensive detection results and achieve a better curative effect for targeted therapy [82]. In addition, it has been proved that IDH mutation is a good prognostic marker for glioma by RNA-seq [83]. Targeted therapy is also considered to enhance or replace cytotoxic chemotherapy regimen in cancer including AML [8486].
ScRNA-seq also has some new discoveries in diagnosis. For example, scRNA-seq data can be used to infer copy number variations (CNV) and to distinguish malignant from non-malignant cells. The infer CNV algorithm, which was used in the study of glioblastoma, uses averaging relative expression levels over large genomic regions to infer chromosome copy number variation [87]. Similar examples include head and neck cancer [88] and human oligodendroglioma [89]. It is reported that RNA sequence of tumor-educated blood platelets (TEPs) can also become a blood-based cancer diagnosis method [90]. It should be noted that the lack of detailed functional implications of the identified RNAs in platelets in the field of platelet RNA research is also an urgent problem to be solved [91].

Cancer heterogeneity and evolution

Heterogeneity has always existed during the transformation of normal cells to cancer cells. The continuous accumulation of heterogeneity may reflect the evolution of cancer [109]. Early RNA sequencing detected all RNA transcripts in a given tissue or cell group, ignoring differences in individual cells. Transcriptome profiling of single-cell RNA sequencing solves this problem by providing single-cell resolution of the transcriptome [3]. In melanoma, single-cell RNA-seq was used to analyze 4645 tumor cells from 19 patients, including cancer cells, immune cells, mesenchymal cells and endothelial cells. Transcriptomic data from different single cells revealed that heterogeneity of cells within the same cancer is associated with cell cycle, spatial background and drug resistance [110]. A recent single-cell RNA-seq study of 49 samples of metastatic lung cancer revealed changes in plasticity induced by non-small cell lung cancer treatment, providing new directions for clinical treatment [111]. Single-cell RNA sequencing also integrates a variety of information in a single cancer cell, deciphering the secrets of cancer heterogeneity and evolution [112]. Compared with scRNA-seq, another emerging technology spatial transcriptome sequencing incorporates information on the spatial location of cells. In prostate cancer, using spatial transcriptomics technology, the transcriptome of nearly 6750 tissue regions was analyzed, revealing the whole-tissue gene expression heterogeneity of the entire multifocal prostate cancer and accurately describing the range of cancer foci [113]. In a study of breast cancer tissues, the results of spatial transcriptome sequencing revealed that gene expression among different regions was surprisingly highly heterogeneous [22]. In recent years, single-nucleus RNA sequencing (snRNA-seq) has also received extensive attention due to its solving the problem that single-cell RNA sequencing cannot be applied to frozen specimens and cannot obtain all cell types in a given tissue [114, 115]. The emerging technology of RNA sequencing will contribute to research on cancer heterogeneity and evolution.

Cancer drug resistance

Drug resistance is a main reason leading to cancer treatment failure. However, the molecular mechanisms underlying drug resistance are still poorly understood [116]. RNA sequencing became a vital tool for revealing the mechanisms of cancer drug resistance. In breast cancer, single-cell RNA sequencing identified a tumor-infiltrating immunosuppressive immature myeloid cell that leads to drug resistance [117]. Another study identified a new COX7B gene related to platinum resistance and a surrogate marker CD63 in cancer cells by single-cell RNA-seq [118]. RNA sequencing has also demonstrated that cancer cells that wake up from a dormant state produce large amounts of BORIS (brother of the regulator of imprinted sites), which can regulate the expression of survival genes in drug-resistant neuroblastoma cells [119]. Identifying special molecules that mediate these processes could help us understand the occurrence of drug resistance. Single-cell transcriptomics can be used to study different modes of chemoresistance in tumor cells and has shown that pre-existing drug-resistant cells can be selected through higher phenotypic intratumoral heterogeneity, while phenotypic homogeneous cells use other mechanisms to trans-differentiate under drug-selection [120]. In one study of pancreatic ductal adenocarcinoma, human pancreatic cancer (PANC-1) cells and gemcitabine-resistant PANC-1 cell lines were compared by RNA sequencing, and two circRNAs were identified as both novel biomarkers and potential therapeutic targets for gemcitabine resistant patients [121]. RNA-seq has also conducted in-depth research on the drug resistance of hematological malignancies. Through RNA-seq, it has been found that non-coding RNAs and fusion genes play an important role in mediating the drug resistance of hematological malignancies [122]. A good example is to compare the circRNA expression profile of the drug-resistant acute myeloid leukemia cell with its parent cell, and determine the circRNAs involved in drug resistance [123]. Similarly, the novel MEF2D-BCL9 fusion transcript identified by RNA-seq was found to increase HDAC9 (histone deacetylase 9) expression and to enhance the resistance to dexamethasone in acute lymphocytic leukemia (ALL) [124]. Leukemia stem cells (LSCs), a rare cell population assumed to be responsible for relapse, is crucial to improve the prognosis of patients [125, 126]. RNA-seq analysis showed that LSCs have a unique lncRNA signature with functional relevance and therapeutic potential, providing an explanation for chemotherapy resistance and disease recurrence [127].

The cancer microenvironment and immunotherapy

The immune system plays a critical role in the cancer microenvironment, affecting several stages of cancer development, including tumorigenesis, progression and metastasis, through tumor-infiltrating lymphocytes (TILs) [128]. TILs and their interactions with malignant cells and stromal cells make up the cancer immune microenvironment. Due to the heterogeneity of cancer, it is difficult to define the exact pro- or anti-cancer function of certain immune cells. Cancer heterogeneity also causes the varied clinical efficacy observed in patients treated with immunotherapies due to different responses of different subclones [129]. Transcriptomic profiling by RNA-seq, in particular scRNA-seq, provides comprehensive information on cellular activity and interactions among cells in the tumor microenvironment (TME). ScRNA-seq enables genomic and molecular profiling of high quantity and quality individual immune cells and assessment of cellular heterogeneity to depict the immune system spectrum in the cancer microenvironment [130132]. ScRNA-seq data demonstrated that compared to normal tissues, cancer tissues exhibited significantly higher heterogeneity in the immune microenvironment, and a continuity in T cell activation resulting from polyclonal T cells and heterogeneous antigen-presenting cells has been identified [133].
ScRNA-seq of tumor-infiltrating T cells in metastatic melanoma identified transcription factor NFATC1 (nuclear factor of activated T cells 1) as a potential molecular signature of T cell exhaustion programs and revealed the depletion of low-exhaustion T cells in expanded clones of T cells [110]. Combining scRNA-seq with assembled T cell receptor (TCR) sequences, 11 T cell subsets, such as CD8 + T cells and CD8 + FOXP3 + regulatory-like cells, and their genomic signatures, were identified in hepatocellular carcinoma (HCC), providing valuable insights for understanding the immune landscape of infiltrating T cells in HCC [134]. Cancer infiltrating T cells also play an anti-tumor role through impairment of an autophagy protein, LC3 (microtubule-associated protein 1A/1B-light chain 3, often short for LC3)-associated phagocytosis (LAP), demonstrating the role of autophagy in oncogenesis and suppression revealed by scRNA-seq [135]. In addition to solid tumors, scRNA-seq of acute myeloid leukemia patients detected diverse immunomodulatory genes that suppress T cell function [136]. By CSOmap, a computational tool for scRNA-seq, the CCL4-CCR8 directed interaction between Tregs and Texs, as well as reduced proliferation of Texs, was characterized [137]. Notably, findings also revealed that tumor-infiltrating T cells exhibited more interactions among themselves than with T cells from peripheral blood and different interactions between tumors and T cells, indicating a varied response to immunotherapy and a potential trend for immune escape [137].
In the cancer immune microenvironment, neutrophils, in addition to T cells, are also key components of cancer progression and cancer drug resistance [138141]. Through scRNA-seq of murine sarcomas and certain human cancers, neutrophils with CSF3R (colony stimulating factor 3 receptor) expression were found to be a part of type 1 antitumor immunity associated with unconventional CD4CD8αβ T cells (UTCαβ) in anti-cancer immunity, indicating better prognosis [142]. ScRNA-seq of metastatic breast cancer and CD45 cells from primary cancer identified neutrophils as pro- and anti-tumorigenic or metastatic, in which pro-tumorigenic and metastatic neutrophils are induced by IL11 expressing cancer subclones, resulting in polyclonal metastasis [143]. This observation also provides new insight into anti-cancer immunotherapy by targeting neutrophils [143]. With scRNA-seq of CD4 and CD8 T cells, several crucial pathways with anti-cancer function were revealed [144].
The balance between immune reaction and immune tolerance is the basis of immune homeostasis, which is also involved in anti-cancer immunity and oncogenesis. scRNA-seq of monocytes and dendritic cells (DCs) separated from a single lymph node melanoma metastasis revealed a conserved homeostatic module regulated by suppressor-of-cytokine-2 (SOCS2) protein and IFNγ [145]. SOCS2 serves an essential regulatory role in anti-tumor immunity and T cell priming through DCs. This highly conserved homeostatic program establishes a connection between autoimmune prevention and immune surveillance in cancer [145].
Immunotherapies, especially immune checkpoint blockade (ICB), has opened a new chapter for anti-cancer therapy with remarkable responses from targeting programmed death 1 (PD1), programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA4) [146148]. However, only a few patients benefit from ICB, and severe side effects were observed [149, 150]. Obviously, various unknown determinants are correlated with the outcome of immunotherapies in addition to well-known factors such as PD1/PD-L1/CTLA-4 expression and mismatch repair deficiency [151155]. Therefore, it is paramount to identify potential effectors for ICB efficacy. By analyzing RNA-seq data from melanoma patients who underwent anti-PD1 and anti-CTLA4 treatment, a potential ICB resistance effector SERPINB9 (a member of the serine protease inhibitor (serpin) family) and the connection between cytotoxic T lymphocytes (CTL) infiltration level and ICB response were characterized [156]. A sub-population cells with immunotherapy persistence have been identified by scRNA-seq and were found to have stem cell-like states with the expression of stem cell antigen-1 (Sca-1) and Snai1 [157].
Another immunotherapy, myeloid-targeted immunotherapy, is based on the complexity of tumor-infiltrating myeloid cells, including DCs and tumor-associated macrophages (TAMs) revealed by scRNA-seq [158]. Through scRNA-seq of immune cells from colorectal cancer patients, C1QC + and SPP1 + TAMs, two subsets of TAMs, were identified, and the mechanism of myeloid-targeted immunotherapy, such as anti-CSF1R (colony stimulating factor 1 receptor) and CD40 agonist, was revealed [159]. Intracellular staining and sequencing (INs-seq), a novel technology integrating scRNA-seq and intracellular protein activity measurements, revealed novel Arg1 + Trem2 + regulatory myeloid (Mreg) cells and demonstrated that depletion of Trem2 led to deduction of exhausted CD8 T cells with increased NK and cytotoxic T cells and cancer suppression by reducing accumulation of intratumoral Mreg cells [160].

Cancer neoantigens

Neoantigens, human leukocyte antigen (HLA)-bound peptides derived from cancer-specific somatic mutations or gene fusions during tumor growth, are another crucial regulator of the clinical response to immunotherapy [161]. Higher intratumor neoantigen heterogeneity and clonal neoantigen burden increases sensitivity to ICB and contributes to better clinical outcome in patients with melanoma and advanced non-small cell lung cancer [162]. This kind of antigen is an optimal target for anti-cancer immunotherapy, enhancing neoantigen-specific T cell activity, and a vaccine targeting personal neoantigens for melanoma patients has been developed [163, 164]. Given all these promising features of personalized medicine targeting neoantigens in tumors, massive parallel profiling of tumor neoantigen burden is necessary for improving clinical efficacy and a deeper understanding of the neoantigen landscape. An RNA-seq-based transcriptomic approach is an efficient tool for neoantigen profiling in many studies. It was revealed that homology of neoantigen and somatic-mutation induced pathogens are important in response prediction in anti-CTLA4 treated melanoma [165]. In addition to melanoma, other studies found reduced neoantigen load in triple-negative and HER2 breast cancers [166], diverse neoantigen abundance in non-small-cell lung cancer patients with different treatment strategies [167], a decreased ratio of neoantigen expression to predicted neoantigens in recurrent glioma due to immune selection pressure [168], a negative correlation between neoantigen abundance and clinical outcome in selected solid tumors [169] and different neoantigen landscapes in immune filtration and T cell dysfunction based on histology in salivary gland carcinoma (SGC) patients [170].
A neoantigen prediction program, Neopepsee, based on RNA-seq data and somatic mutation, can be utilized to detect potential neoantigens for personal vaccine development with reduced false-positive rate compared to binding affinity prediction [171]. ScanNeo is another prediction computational pipeline based on RNA-seq that aims to identify insertion and deletion derived neoantigens, which was validated in prostate cancer [172], and ASNEO, which identifies personal-specific alternative splicing derived neoantigens [173]. Several neoantigens have been identified to be related to cancer prognosis and might be potential targets of immunotherapies, such as the TP53 neoantigen for HCC patients [174]. For the anti-neoantigen immunotherapy in cancer, a new strategy involving neoantigen-specific TCRs modification has been proposed, and scRNA-seq has been applied to isolate neoantigen-specific TCRs for further clinical application [175].

Conclusions and perspectives

High-throughput RNA-seq technology has been a major tool to explore the transcriptome. The rapid development of RNA-seq technology not only saves time and cost but also sheds light on many new research fields. However, there are still limitations of RNA-seq technology that need to be improved.
For short-read length RNA-seq technologies, bias and imperfections are primarily generated in sequencing library preparation and short read assembly. It is difficult for these methods to correctly identify multiple isoforms from a certain gene. To overcome the disadvantage of short read length, improved read coverage and sequencing depth is required. Long-read length RNA-seq technologies avoid shortcomings in template amplification, reduce the false positive rate in splice junction detection and enable the identification of unannotated longer transcripts, overcoming the common limitations of short-read sequencing [176, 177]. However, this method suffers from the drawback of reduced throughput, higher cost and higher sequencing error rate, especially insertion-deletion errors. To reduce random errors, PacBio circular consensus-sequencing (CCS) was developed to increase sequencing depth by rereading molecules several times. However, it also reduces the identification rate of unique isoforms. In addition, the sensitivity of long-read sequencing for identification of differentially expressed genes is lower compared to short-read sequencing [178180]. Thus, hybridization of long-read and short-read sequencing has been reported to yield a more comprehensive and accurate analysis [181].
Improvements in the throughput of RNA sequencing technology have resulted in billions of sequencing reads, bringing great challenges to the computational process, such as data storage, transmission, quality control and data analysis, including read mapping, transcript assembly and read normalization. Therefore, it is important for bioinformatics to keep pace with the continuous developments of RNA-seq technologies. Notably, bias could be produced due to differences in read data handling, necessitating the improvement of current bioinformatics pipelines.
RNA-seq measures gene expression by the read counts, which always containing missing values, thus results in information loss of specific gene and negative impact on downstream analysis. To overcome this problem, missing data need to be imputed and analyzed by several methods, such as optimal clustering with missing values [182]. For scRNA-seq, the proportion of genes with zero or low expression varies across cells due to biological or technical bias. For example, batch effects can come from cells captured and sequenced in different conditions [183]. Imputation methods, such as SAVER, MAGIC and kNN-smoothing, are recommended for scRNA-seq [184]. Another method named batch effects correction with unknown subtypes for scRNA-seq data (BUSseq) utilizes Bayesian hierarchical model and can also be used to correct batch effects and missing data [185].
Combination of data from multi-omics sequencing can undoubtedly expand the application of RNA-seq. For example, Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) was developed by utilizing hyperactive Tn5 transposase to identify open chromatin region and transcriptional factor (TF) binding sites [186]. The integration of ATAC-seq and RNA-seq enables the reveal of TF-targeted genes and their transcripts [187, 188]. Chromatin conformation capture analysis (3C) technology and its several derivatives including circular chromosome conformation capture (4C), carbon copy chromosome conformation capture (5C), ChIP-Loop, Hi-C and capture Hi-C were developed and improved to detect chromatin structure as well as unknown interacting regions [189191]. It has been reported that combined analysis of RNA-seq and chromatin structure can detect structure variation-related differentially expressed genes [192194].
Epitranscriptomics is a crucial part of gene expression, and methylation of adenosine at the N6 position (m6A) is the most abundant [195]. Traditional RNA-seq needs reverse transcription before sequencing and thus easily loses the information of transcriptome complexity. This shortcoming can be overcome by directly sequencing native RNA molecules using methods such as nanopore sequencing. Transcript modifications could be inferred from the current signal as the modified RNA molecules passing nanopore cause a characteristic temporary current blockade, which enables the detection of diverse modifications such as m6A or 5-methylcytosine (m5C) [196198].
ScRNA-seq is a powerful technology to facilitate further exploration in cancer research and also has been employed in the detection of cancer stem cell subpopulation, metabolic switch in cancer-draining lymph nodes and therapy-induced adaption of cancer cells [111, 199, 200]. Combined with cell sorting or ligand-receptor interaction, scRNA-seq was utilized in cellular interaction, cell spatial organization as well as molecular crosstalk characterization [137, 201, 202]. Coupling of parallel CRISPR (clustered regularly interspaced short palindromic repeats)-pooled screen, scRNA-seq enables the simultaneous analysis of genomic perturbation and transcriptional activity to detect heterogeneous cell type as well as crucial factors of complexity regulatory mechanism [203205]. ScNT-seq, single-cell metabolically labeled new RNA tagging sequencing, brings RNA-seq into time resolution by identifying RNAs transcribed at different stage [206]. Utilizing SNP-based demultiplexing of scRNA-seq data, MIX-Seq was developed to study cancer cell reaction to pharmacologic treatment [207]. Another technology, snRNA-seq, is invaluable for detecting cellular heterogeneity of cancer and has been employed to identification of a sub-population of adipocytes regulating cancer genesis [208].
Taken together, RNA-seq has been applied in an impressively wide range of cancer research. All applications in cancer research rely on the boost of advanced RNA-seq technologies, especially the combination of scRNA-seq and spatial transcriptomics as well as data from multi-omics, which will bring RNA-seq technologies into single-cell resolution and tissue-level transcriptomics, providing new insight into cancer diagnosis, treatment and prevention.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Margulies M, Egholm M, Altman WE, Attiya S, Bader JS, Bemben LA, et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature. 2005;437:376–80.PubMedPubMedCentralCrossRef Margulies M, Egholm M, Altman WE, Attiya S, Bader JS, Bemben LA, et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature. 2005;437:376–80.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Cieślik M, Chinnaiyan AM. Cancer transcriptome profiling at the juncture of clinical translation. Nat Rev Genet. 2018;19:93–109.PubMedCrossRef Cieślik M, Chinnaiyan AM. Cancer transcriptome profiling at the juncture of clinical translation. Nat Rev Genet. 2018;19:93–109.PubMedCrossRef
3.
Zurück zum Zitat Suvà ML, Tirosh I. Single-cell RNA sequencing in cancer: lessons learned and emerging challenges. Mol Cell. 2019;75:7–12.PubMedCrossRef Suvà ML, Tirosh I. Single-cell RNA sequencing in cancer: lessons learned and emerging challenges. Mol Cell. 2019;75:7–12.PubMedCrossRef
4.
Zurück zum Zitat Watson JD, Crick FH. Molecular structure of nucleic acids; a structure for deoxyribose nucleic acid. Nature. 1953;171:737–8.PubMedCrossRef Watson JD, Crick FH. Molecular structure of nucleic acids; a structure for deoxyribose nucleic acid. Nature. 1953;171:737–8.PubMedCrossRef
7.
Zurück zum Zitat Sanger F, Air GM, Barrell BG, Brown NL, Coulson AR, Fiddes CA, et al. Nucleotide sequence of bacteriophage phi X174 DNA. Nature. 1977;265:687–95.PubMedCrossRef Sanger F, Air GM, Barrell BG, Brown NL, Coulson AR, Fiddes CA, et al. Nucleotide sequence of bacteriophage phi X174 DNA. Nature. 1977;265:687–95.PubMedCrossRef
8.
Zurück zum Zitat Russo G, Zegar C, Giordano A. Advantages and limitations of microarray technology in human cancer. Oncogene. 2003;22:6497–507.PubMedCrossRef Russo G, Zegar C, Giordano A. Advantages and limitations of microarray technology in human cancer. Oncogene. 2003;22:6497–507.PubMedCrossRef
9.
Zurück zum Zitat Li S, Tighe S, Nicolet C, Grove D, Levy S, Farmerie W, et al. Multi-platform assessment of transcriptome profiling using RNA-seq in the ABRF next-generation sequencing study. Nat Biotechnol. 2014;32:915–25.PubMedPubMedCentralCrossRef Li S, Tighe S, Nicolet C, Grove D, Levy S, Farmerie W, et al. Multi-platform assessment of transcriptome profiling using RNA-seq in the ABRF next-generation sequencing study. Nat Biotechnol. 2014;32:915–25.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat A comprehensive assessment of RNA-seq accuracy, reproducibility and information content by the Sequencing Quality Control Consortium. Nat Biotechnol. 2014;32:903–14. A comprehensive assessment of RNA-seq accuracy, reproducibility and information content by the Sequencing Quality Control Consortium. Nat Biotechnol. 2014;32:903–14.
11.
Zurück zum Zitat Mardis E. Next-generation DNA sequencing methods. Annu Rev Genomics Hum Genet. 2008;9:387–402.PubMedCrossRef Mardis E. Next-generation DNA sequencing methods. Annu Rev Genomics Hum Genet. 2008;9:387–402.PubMedCrossRef
12.
13.
Zurück zum Zitat Mardis E. Next-generation sequencing platforms. Annu Rev Anal Chem (Palo Alto Calif). 2013;6:287–303.CrossRef Mardis E. Next-generation sequencing platforms. Annu Rev Anal Chem (Palo Alto Calif). 2013;6:287–303.CrossRef
14.
Zurück zum Zitat Marioni JC, Mason CE, Mane SM, Stephens M, Gilad Y. RNA-seq: an assessment of technical reproducibility and comparison with gene expression arrays. Genome Res. 2008;18:1509–17.PubMedPubMedCentralCrossRef Marioni JC, Mason CE, Mane SM, Stephens M, Gilad Y. RNA-seq: an assessment of technical reproducibility and comparison with gene expression arrays. Genome Res. 2008;18:1509–17.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Schadt EE, Turner S, Kasarskis A. A window into third-generation sequencing. Hum Mol Genet. 2010;19:R227–40.PubMedCrossRef Schadt EE, Turner S, Kasarskis A. A window into third-generation sequencing. Hum Mol Genet. 2010;19:R227–40.PubMedCrossRef
16.
Zurück zum Zitat Eid J, Fehr A, Gray J, Luong K, Lyle J, Otto G, et al. Real-time DNA sequencing from single polymerase molecules. Science. 2009;323:133–8.PubMedCrossRef Eid J, Fehr A, Gray J, Luong K, Lyle J, Otto G, et al. Real-time DNA sequencing from single polymerase molecules. Science. 2009;323:133–8.PubMedCrossRef
17.
Zurück zum Zitat Hart C, Lipson D, Ozsolak F, Raz T, Steinmann K, Thompson J, et al. Single-molecule sequencing: sequence methods to enable accurate quantitation. Methods Enzymol. 2010;472:407–30.PubMedCrossRef Hart C, Lipson D, Ozsolak F, Raz T, Steinmann K, Thompson J, et al. Single-molecule sequencing: sequence methods to enable accurate quantitation. Methods Enzymol. 2010;472:407–30.PubMedCrossRef
18.
19.
Zurück zum Zitat Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. 2009;6:377–82.PubMedCrossRef Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. 2009;6:377–82.PubMedCrossRef
20.
Zurück zum Zitat Cao J, Packer J, Ramani V, Cusanovich D, Huynh C, Daza R, et al. Comprehensive single-cell transcriptional profiling of a multicellular organism. Science. 2017;357:661–7.PubMedPubMedCentralCrossRef Cao J, Packer J, Ramani V, Cusanovich D, Huynh C, Daza R, et al. Comprehensive single-cell transcriptional profiling of a multicellular organism. Science. 2017;357:661–7.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Rosenberg A, Roco C, Muscat R, Kuchina A, Sample P, Yao Z, et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science. 2018;360:176–82.PubMedPubMedCentralCrossRef Rosenberg A, Roco C, Muscat R, Kuchina A, Sample P, Yao Z, et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science. 2018;360:176–82.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Ståhl P, Salmén F, Vickovic S, Lundmark A, Navarro J, Magnusson J, et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 2016;353:78–82.PubMedCrossRef Ståhl P, Salmén F, Vickovic S, Lundmark A, Navarro J, Magnusson J, et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 2016;353:78–82.PubMedCrossRef
23.
Zurück zum Zitat Rodriques SG, Stickels RR, Goeva A, Martin CA, Murray E, Vanderburg CR, et al. Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science. 2019;363:1463–7.PubMedPubMedCentralCrossRef Rodriques SG, Stickels RR, Goeva A, Martin CA, Murray E, Vanderburg CR, et al. Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science. 2019;363:1463–7.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Chen J, Suo S, Tam PP, Han JJ, Peng G, Jing N. Spatial transcriptomic analysis of cryosectioned tissue samples with geo-seq. Nat Protoc. 2017;12:566–80.PubMedCrossRef Chen J, Suo S, Tam PP, Han JJ, Peng G, Jing N. Spatial transcriptomic analysis of cryosectioned tissue samples with geo-seq. Nat Protoc. 2017;12:566–80.PubMedCrossRef
25.
Zurück zum Zitat Ke R, Mignardi M, Pacureanu A, Svedlund J, Botling J, Wählby C, et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat Methods. 2013;10:857–60.PubMedCrossRef Ke R, Mignardi M, Pacureanu A, Svedlund J, Botling J, Wählby C, et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat Methods. 2013;10:857–60.PubMedCrossRef
26.
Zurück zum Zitat Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Yang JL, Ferrante TC, et al. Highly multiplexed subcellular RNA sequencing in situ. Science. 2014;343:1360–3.PubMedPubMedCentralCrossRef Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Yang JL, Ferrante TC, et al. Highly multiplexed subcellular RNA sequencing in situ. Science. 2014;343:1360–3.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Mercer TR, Gerhardt DJ, Dinger ME, Crawford J, Trapnell C, Jeddeloh JA, et al. Targeted RNA sequencing reveals the deep complexity of the human transcriptome. Nat Biotechnol. 2011;30:99–104.PubMedPubMedCentralCrossRef Mercer TR, Gerhardt DJ, Dinger ME, Crawford J, Trapnell C, Jeddeloh JA, et al. Targeted RNA sequencing reveals the deep complexity of the human transcriptome. Nat Biotechnol. 2011;30:99–104.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Heyer EE, Deveson IW, Wooi D, Selinger CI, Lyons RJ, Hayes VM, et al. Diagnosis of fusion genes using targeted RNA sequencing. Nat Commun. 2019;10:1388.PubMedPubMedCentralCrossRef Heyer EE, Deveson IW, Wooi D, Selinger CI, Lyons RJ, Hayes VM, et al. Diagnosis of fusion genes using targeted RNA sequencing. Nat Commun. 2019;10:1388.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Soverini S, Abruzzese E, Bocchia M, Bonifacio M, Galimberti S, Gozzini A, et al. Next-generation sequencing for BCR-ABL1 kinase domain mutation testing in patients with chronic myeloid leukemia: a position paper. J Hematol Oncol. 2019;12:131.PubMedPubMedCentralCrossRef Soverini S, Abruzzese E, Bocchia M, Bonifacio M, Galimberti S, Gozzini A, et al. Next-generation sequencing for BCR-ABL1 kinase domain mutation testing in patients with chronic myeloid leukemia: a position paper. J Hematol Oncol. 2019;12:131.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Chatterjee A, Ahn A, Rodger EJ, Stockwell PA, Eccles MR. A guide for designing and analyzing RNA-seq data. Methods Mol Biol. 2018;1783:35–80.PubMedCrossRef Chatterjee A, Ahn A, Rodger EJ, Stockwell PA, Eccles MR. A guide for designing and analyzing RNA-seq data. Methods Mol Biol. 2018;1783:35–80.PubMedCrossRef
31.
Zurück zum Zitat Conesa A, Madrigal P, Tarazona S, Gomez-Cabrero D, Cervera A, McPherson A, et al. A survey of best practices for RNA-seq data analysis. Genome Biol. 2016;17:13.PubMedPubMedCentralCrossRef Conesa A, Madrigal P, Tarazona S, Gomez-Cabrero D, Cervera A, McPherson A, et al. A survey of best practices for RNA-seq data analysis. Genome Biol. 2016;17:13.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Cock PJ, Fields CJ, Goto N, Heuer ML, Rice PM. The Sanger FASTQ file format for sequences with quality scores, and the Solexa/Illumina FASTQ variants. Nucleic Acids Res. 2010;38:1767–71.PubMedCrossRef Cock PJ, Fields CJ, Goto N, Heuer ML, Rice PM. The Sanger FASTQ file format for sequences with quality scores, and the Solexa/Illumina FASTQ variants. Nucleic Acids Res. 2010;38:1767–71.PubMedCrossRef
35.
Zurück zum Zitat DeLuca DS, Levin JZ, Sivachenko A, Fennell T, Nazaire MD, Williams C, et al. RNA-SeQC: RNA-seq metrics for quality control and process optimization. Bioinformatics. 2012;28:1530–2.PubMedPubMedCentralCrossRef DeLuca DS, Levin JZ, Sivachenko A, Fennell T, Nazaire MD, Williams C, et al. RNA-SeQC: RNA-seq metrics for quality control and process optimization. Bioinformatics. 2012;28:1530–2.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Chen Y, Chen Y, Shi C, Huang Z, Zhang Y, Li S, et al. SOAPnuke: a MapReduce acceleration-supported software for integrated quality control and preprocessing of high-throughput sequencing data. Gigascience. 2018;7:1–6.PubMedPubMedCentral Chen Y, Chen Y, Shi C, Huang Z, Zhang Y, Li S, et al. SOAPnuke: a MapReduce acceleration-supported software for integrated quality control and preprocessing of high-throughput sequencing data. Gigascience. 2018;7:1–6.PubMedPubMedCentral
39.
Zurück zum Zitat Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013;14:R36.PubMedPubMedCentralCrossRef Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013;14:R36.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.PubMedCrossRef Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.PubMedCrossRef
43.
Zurück zum Zitat Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10:R25.PubMedPubMedCentralCrossRef Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10:R25.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28:511–5.PubMedPubMedCentralCrossRef Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28:511–5.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33:290–5.PubMedPubMedCentralCrossRef Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33:290–5.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Grabherr MG, Haas BJ, Yassour M, Levin JZ, Thompson DA, Amit I, et al. Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat Biotechnol. 2011;29:644–52.PubMedPubMedCentralCrossRef Grabherr MG, Haas BJ, Yassour M, Levin JZ, Thompson DA, Amit I, et al. Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat Biotechnol. 2011;29:644–52.PubMedPubMedCentralCrossRef
47.
48.
Zurück zum Zitat Robertson G, Schein J, Chiu R, Corbett R, Field M, Jackman SD, et al. De novo assembly and analysis of RNA-seq data. Nat Methods. 2010;7:909–12.PubMedCrossRef Robertson G, Schein J, Chiu R, Corbett R, Field M, Jackman SD, et al. De novo assembly and analysis of RNA-seq data. Nat Methods. 2010;7:909–12.PubMedCrossRef
49.
Zurück zum Zitat Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–30.PubMedCrossRef Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–30.PubMedCrossRef
50.
Zurück zum Zitat Anders S, Pyl PT, Huber W. HTSeq: a python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–9.PubMedCrossRef Anders S, Pyl PT, Huber W. HTSeq: a python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–9.PubMedCrossRef
51.
Zurück zum Zitat Roberts A, Pachter L. Streaming fragment assignment for real-time analysis of sequencing experiments. Nat Methods. 2013;10:71–3.PubMedCrossRef Roberts A, Pachter L. Streaming fragment assignment for real-time analysis of sequencing experiments. Nat Methods. 2013;10:71–3.PubMedCrossRef
52.
Zurück zum Zitat Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011;12:323.CrossRef Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011;12:323.CrossRef
54.
Zurück zum Zitat Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016;34:525–7.PubMedCrossRef Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016;34:525–7.PubMedCrossRef
55.
Zurück zum Zitat Patro R, Mount SM, Kingsford C. Sailfish enables alignment-free isoform quantification from RNA-seq reads using lightweight algorithms. Nat Biotechnol. 2014;32:462–4.PubMedPubMedCentralCrossRef Patro R, Mount SM, Kingsford C. Sailfish enables alignment-free isoform quantification from RNA-seq reads using lightweight algorithms. Nat Biotechnol. 2014;32:462–4.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Patro R, Duggal G, Love MI, Irizarry RA, Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods. 2017;14:417–9.PubMedPubMedCentralCrossRef Patro R, Duggal G, Love MI, Irizarry RA, Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods. 2017;14:417–9.PubMedPubMedCentralCrossRef
57.
58.
Zurück zum Zitat Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.PubMedCrossRef Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.PubMedCrossRef
59.
Zurück zum Zitat Moon M, Nakai K. Stable feature selection based on the ensemble L (1)-norm support vector machine for biomarker discovery. BMC Genomics. 2016;17:1026.PubMedPubMedCentralCrossRef Moon M, Nakai K. Stable feature selection based on the ensemble L (1)-norm support vector machine for biomarker discovery. BMC Genomics. 2016;17:1026.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Zubovic L, Piazza S, Tebaldi T, Cozzuto L, Palazzo G, Sidarovich V, et al. The altered transcriptome of pediatric myelodysplastic syndrome revealed by RNA sequencing. J Hematol Oncol. 2020;13:135.PubMedPubMedCentralCrossRef Zubovic L, Piazza S, Tebaldi T, Cozzuto L, Palazzo G, Sidarovich V, et al. The altered transcriptome of pediatric myelodysplastic syndrome revealed by RNA sequencing. J Hematol Oncol. 2020;13:135.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Govindarajan M, Wohlmuth C, Waas M, Bernardini MQ, Kislinger T. High-throughput approaches for precision medicine in high-grade serous ovarian cancer. J Hematol Oncol. 2020;13:134.PubMedPubMedCentralCrossRef Govindarajan M, Wohlmuth C, Waas M, Bernardini MQ, Kislinger T. High-throughput approaches for precision medicine in high-grade serous ovarian cancer. J Hematol Oncol. 2020;13:134.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Reeser JW, Martin D, Miya J, Kautto EA, Lyon E, Zhu E, et al. Validation of a targeted RNA sequencing assay for kinase fusion detection in solid tumors. J Mol Diagn. 2017;19:682–96.PubMedPubMedCentralCrossRef Reeser JW, Martin D, Miya J, Kautto EA, Lyon E, Zhu E, et al. Validation of a targeted RNA sequencing assay for kinase fusion detection in solid tumors. J Mol Diagn. 2017;19:682–96.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Mercer TR, Clark MB, Crawford J, Brunck ME, Gerhardt DJ, Taft RJ, et al. Targeted sequencing for gene discovery and quantification using RNA CaptureSeq. Nat Protoc. 2014;9:989–1009.PubMedCrossRef Mercer TR, Clark MB, Crawford J, Brunck ME, Gerhardt DJ, Taft RJ, et al. Targeted sequencing for gene discovery and quantification using RNA CaptureSeq. Nat Protoc. 2014;9:989–1009.PubMedCrossRef
66.
Zurück zum Zitat Togni M, Masetti R, Pigazzi M, Astolfi A, Zama D, Indio V, et al. Identification of the NUP98-PHF23 fusion gene in pediatric cytogenetically normal acute myeloid leukemia by whole-transcriptome sequencing. J Hematol Oncol. 2015;8:69.PubMedPubMedCentralCrossRef Togni M, Masetti R, Pigazzi M, Astolfi A, Zama D, Indio V, et al. Identification of the NUP98-PHF23 fusion gene in pediatric cytogenetically normal acute myeloid leukemia by whole-transcriptome sequencing. J Hematol Oncol. 2015;8:69.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Veeraraghavan J, Ma J, Hu Y, Wang XS. Recurrent and pathological gene fusions in breast cancer: current advances in genomic discovery and clinical implications. Breast Cancer Res Treat. 2016;158:219–32.PubMedPubMedCentralCrossRef Veeraraghavan J, Ma J, Hu Y, Wang XS. Recurrent and pathological gene fusions in breast cancer: current advances in genomic discovery and clinical implications. Breast Cancer Res Treat. 2016;158:219–32.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Kloosterman WP, Coebergh van den Braak RRJ, Pieterse M, van Roosmalen MJ, Sieuwerts AM, Stangl C, et al. A systematic analysis of oncogenic gene fusions in primary colon cancer. Cancer Res. 2017;77:3814–22. Kloosterman WP, Coebergh van den Braak RRJ, Pieterse M, van Roosmalen MJ, Sieuwerts AM, Stangl C, et al. A systematic analysis of oncogenic gene fusions in primary colon cancer. Cancer Res. 2017;77:3814–22.
69.
Zurück zum Zitat Sun YM, Chen YQ. Principles and innovative technologies for decrypting noncoding RNAs: from discovery and functional prediction to clinical application. J Hematol Oncol. 2020;13:109.PubMedPubMedCentralCrossRef Sun YM, Chen YQ. Principles and innovative technologies for decrypting noncoding RNAs: from discovery and functional prediction to clinical application. J Hematol Oncol. 2020;13:109.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Zhou X, Zhan L, Huang K, Wang X. The functions and clinical significance of circRNAs in hematological malignancies. J Hematol Oncol. 2020;13:138.PubMedPubMedCentralCrossRef Zhou X, Zhan L, Huang K, Wang X. The functions and clinical significance of circRNAs in hematological malignancies. J Hematol Oncol. 2020;13:138.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Liu Y, Cheng Z, Pang Y, Cui L, Qian T, Quan L, et al. Role of microRNAs, circRNAs and long noncoding RNAs in acute myeloid leukemia. J Hematol Oncol. 2019;12:51.PubMedPubMedCentralCrossRef Liu Y, Cheng Z, Pang Y, Cui L, Qian T, Quan L, et al. Role of microRNAs, circRNAs and long noncoding RNAs in acute myeloid leukemia. J Hematol Oncol. 2019;12:51.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Xu H, Wang C, Song H, Xu Y, Ji G. RNA-Seq profiling of circular RNAs in human colorectal Cancer liver metastasis and the potential biomarkers. Mol Cancer. 2019;18:8.PubMedPubMedCentralCrossRef Xu H, Wang C, Song H, Xu Y, Ji G. RNA-Seq profiling of circular RNAs in human colorectal Cancer liver metastasis and the potential biomarkers. Mol Cancer. 2019;18:8.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Alisoltani A, Fallahi H, Shiran B, Alisoltani A, Ebrahimie E. RNA-seq SSRs and small RNA-seq SSRs: new approaches in cancer biomarker discovery. Gene. 2015;560:34–43.PubMedCrossRef Alisoltani A, Fallahi H, Shiran B, Alisoltani A, Ebrahimie E. RNA-seq SSRs and small RNA-seq SSRs: new approaches in cancer biomarker discovery. Gene. 2015;560:34–43.PubMedCrossRef
74.
Zurück zum Zitat Zhou FF, Huang R, Jiang J, Zeng XH, Zou SQ. Correlated non-nuclear COX2 and low HER2 expression confers a good prognosis in colorectal cancer. Saudi J Gastroenterol. 2018;24:301–6.PubMedPubMedCentralCrossRef Zhou FF, Huang R, Jiang J, Zeng XH, Zou SQ. Correlated non-nuclear COX2 and low HER2 expression confers a good prognosis in colorectal cancer. Saudi J Gastroenterol. 2018;24:301–6.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Chen RH, Du Y, Han P, Wang HB, Liang FY, Feng GK, et al. ISG15 predicts poor prognosis and promotes cancer stem cell phenotype in nasopharyngeal carcinoma. Oncotarget. 2016;7:16910–22.PubMedPubMedCentralCrossRef Chen RH, Du Y, Han P, Wang HB, Liang FY, Feng GK, et al. ISG15 predicts poor prognosis and promotes cancer stem cell phenotype in nasopharyngeal carcinoma. Oncotarget. 2016;7:16910–22.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Liu G, Hou G, Li L, Li Y, Zhou W, Liu L. Potential diagnostic and prognostic marker dimethylglycine dehydrogenase (DMGDH) suppresses hepatocellular carcinoma metastasis in vitro and in vivo. Oncotarget. 2016;7:32607–16.PubMedPubMedCentralCrossRef Liu G, Hou G, Li L, Li Y, Zhou W, Liu L. Potential diagnostic and prognostic marker dimethylglycine dehydrogenase (DMGDH) suppresses hepatocellular carcinoma metastasis in vitro and in vivo. Oncotarget. 2016;7:32607–16.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Mei J, Hao L, Liu X, Sun G, Xu R, Wang H, et al. Comprehensive analysis of peroxiredoxins expression profiles and prognostic values in breast cancer. Biomark Res. 2019;7:16.PubMedPubMedCentralCrossRef Mei J, Hao L, Liu X, Sun G, Xu R, Wang H, et al. Comprehensive analysis of peroxiredoxins expression profiles and prognostic values in breast cancer. Biomark Res. 2019;7:16.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Wang L, Ge J, Lan Y, Shi Y, Luo Y, Tan Y, et al. Tumor mutational burden is associated with poor outcomes in diffuse glioma. BMC Cancer. 2020;20:213.PubMedPubMedCentralCrossRef Wang L, Ge J, Lan Y, Shi Y, Luo Y, Tan Y, et al. Tumor mutational burden is associated with poor outcomes in diffuse glioma. BMC Cancer. 2020;20:213.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Jiang T, Shi J, Dong Z, Hou L, Zhao C, Li X, et al. Genomic landscape and its correlations with tumor mutational burden, PD-L1 expression, and immune cells infiltration in Chinese lung squamous cell carcinoma. J Hematol Oncol. 2019;12:75.PubMedPubMedCentralCrossRef Jiang T, Shi J, Dong Z, Hou L, Zhao C, Li X, et al. Genomic landscape and its correlations with tumor mutational burden, PD-L1 expression, and immune cells infiltration in Chinese lung squamous cell carcinoma. J Hematol Oncol. 2019;12:75.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Seo JS, Ju YS, Lee WC, Shin JY, Lee JK, Bleazard T, et al. The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res. 2012;22:2109–19.PubMedPubMedCentralCrossRef Seo JS, Ju YS, Lee WC, Shin JY, Lee JK, Bleazard T, et al. The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res. 2012;22:2109–19.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Nakagawa M, Nakatani F, Matsunaga H, Seki T, Endo M, Ogawara Y, et al. Selective inhibition of mutant IDH1 by DS-1001b ameliorates aberrant histone modifications and impairs tumor activity in chondrosarcoma. Oncogene. 2019;38:6835–49.PubMedCrossRef Nakagawa M, Nakatani F, Matsunaga H, Seki T, Endo M, Ogawara Y, et al. Selective inhibition of mutant IDH1 by DS-1001b ameliorates aberrant histone modifications and impairs tumor activity in chondrosarcoma. Oncogene. 2019;38:6835–49.PubMedCrossRef
82.
Zurück zum Zitat Davies KD, Lomboy A, Lawrence CA, Yourshaw M, Bocsi GT, Camidge DR, et al. DNA-based versus RNA-based detection of MET Exon 14 skipping events in lung cancer. J Thorac Oncol. 2019;14:737–41.PubMedCrossRef Davies KD, Lomboy A, Lawrence CA, Yourshaw M, Bocsi GT, Camidge DR, et al. DNA-based versus RNA-based detection of MET Exon 14 skipping events in lung cancer. J Thorac Oncol. 2019;14:737–41.PubMedCrossRef
83.
Zurück zum Zitat Unruh D, Zewde M, Buss A, Drumm MR, Tran AN, Scholtens DM, et al. Methylation and transcription patterns are distinct in IDH mutant gliomas compared to other IDH mutant cancers. Sci Rep. 2019;9:8946.PubMedPubMedCentralCrossRef Unruh D, Zewde M, Buss A, Drumm MR, Tran AN, Scholtens DM, et al. Methylation and transcription patterns are distinct in IDH mutant gliomas compared to other IDH mutant cancers. Sci Rep. 2019;9:8946.PubMedPubMedCentralCrossRef
84.
87.
Zurück zum Zitat Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344:1396–401.PubMedPubMedCentralCrossRef Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344:1396–401.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell. 2017;171(1611–24):e24. Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell. 2017;171(1611–24):e24.
89.
Zurück zum Zitat Tirosh I, Venteicher AS, Hebert C, Escalante LE, Patel AP, Yizhak K, et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature. 2016;539:309–13.PubMedPubMedCentralCrossRef Tirosh I, Venteicher AS, Hebert C, Escalante LE, Patel AP, Yizhak K, et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature. 2016;539:309–13.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Best MG, Sol N, Kooi I, Tannous J, Westerman BA, Rustenburg F, et al. RNA-seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and molecular pathway cancer diagnostics. Cancer Cell. 2015;28:666–76.PubMedPubMedCentralCrossRef Best MG, Sol N, Kooi I, Tannous J, Westerman BA, Rustenburg F, et al. RNA-seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and molecular pathway cancer diagnostics. Cancer Cell. 2015;28:666–76.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Best MG, Vancura A, Wurdinger T. Platelet RNA as a circulating biomarker trove for cancer diagnostics. J Thromb Haemost. 2017;15:1295–306.PubMedCrossRef Best MG, Vancura A, Wurdinger T. Platelet RNA as a circulating biomarker trove for cancer diagnostics. J Thromb Haemost. 2017;15:1295–306.PubMedCrossRef
92.
Zurück zum Zitat Zhu L, Li J, Gong Y, Wu Q, Tan S, Sun D, et al. Exosomal tRNA-derived small RNA as a promising biomarker for cancer diagnosis. Mol Cancer. 2019;18:74.PubMedPubMedCentralCrossRef Zhu L, Li J, Gong Y, Wu Q, Tan S, Sun D, et al. Exosomal tRNA-derived small RNA as a promising biomarker for cancer diagnosis. Mol Cancer. 2019;18:74.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Nie Y, Jiao Y, Li Y, Li W. Investigation of the clinical significance and prognostic value of the lncRNA ACVR2B-As1 in liver cancer. Biomed Res Int. 2019;2019:4602371.PubMedPubMedCentral Nie Y, Jiao Y, Li Y, Li W. Investigation of the clinical significance and prognostic value of the lncRNA ACVR2B-As1 in liver cancer. Biomed Res Int. 2019;2019:4602371.PubMedPubMedCentral
94.
Zurück zum Zitat Gong W, Yang L, Wang Y, Xian J, Qiu F, Liu L, et al. Analysis of survival-related lncRNA landscape identifies a role for LINC01537 in energy metabolism and lung cancer progression. Int J Mol Sci. 2019;20. Gong W, Yang L, Wang Y, Xian J, Qiu F, Liu L, et al. Analysis of survival-related lncRNA landscape identifies a role for LINC01537 in energy metabolism and lung cancer progression. Int J Mol Sci. 2019;20.
95.
Zurück zum Zitat Hang D, Zhou J, Qin N, Zhou W, Ma H, Jin G, et al. A novel plasma circular RNA circFARSA is a potential biomarker for non-small cell lung cancer. Cancer Med. 2018;7:2783–91.PubMedPubMedCentralCrossRef Hang D, Zhou J, Qin N, Zhou W, Ma H, Jin G, et al. A novel plasma circular RNA circFARSA is a potential biomarker for non-small cell lung cancer. Cancer Med. 2018;7:2783–91.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Hua Q, Jin M, Mi B, Xu F, Li T, Zhao L, et al. LINC01123, a c-Myc-activated long non-coding RNA, promotes proliferation and aerobic glycolysis of non-small cell lung cancer through miR-199a-5p/c-Myc axis. J Hematol Oncol. 2019;12:91.PubMedPubMedCentralCrossRef Hua Q, Jin M, Mi B, Xu F, Li T, Zhao L, et al. LINC01123, a c-Myc-activated long non-coding RNA, promotes proliferation and aerobic glycolysis of non-small cell lung cancer through miR-199a-5p/c-Myc axis. J Hematol Oncol. 2019;12:91.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Wang Z, Qin B. Prognostic and clinicopathological significance of long noncoding RNA CTD-2510F5.4 in gastric cancer. Gastric Cancer. 2019;22:692–704. Wang Z, Qin B. Prognostic and clinicopathological significance of long noncoding RNA CTD-2510F5.4 in gastric cancer. Gastric Cancer. 2019;22:692–704.
98.
Zurück zum Zitat Luo T, Zhao J, Lu Z, Bi J, Pang T, Cui H, et al. Characterization of long non-coding RNAs and MEF2C-AS1 identified as a novel biomarker in diffuse gastric cancer. Transl Oncol. 2018;11:1080–9.PubMedPubMedCentralCrossRef Luo T, Zhao J, Lu Z, Bi J, Pang T, Cui H, et al. Characterization of long non-coding RNAs and MEF2C-AS1 identified as a novel biomarker in diffuse gastric cancer. Transl Oncol. 2018;11:1080–9.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Wang D, Wan X, Zhang Y, Kong Z, Lu Y, Sun X, et al. A novel androgen-reduced prostate-specific lncRNA, PSLNR, inhibits prostate-cancer progression in part by regulating the p53-dependent pathway. Prostate. 2019;79:1362–77.PubMed Wang D, Wan X, Zhang Y, Kong Z, Lu Y, Sun X, et al. A novel androgen-reduced prostate-specific lncRNA, PSLNR, inhibits prostate-cancer progression in part by regulating the p53-dependent pathway. Prostate. 2019;79:1362–77.PubMed
100.
Zurück zum Zitat Silva-Fisher JM, Dang HX, White NM, Strand MS, Krasnick BA, Rozycki EB, et al. Long non-coding RNA RAMS11 promotes metastatic colorectal cancer progression. Nat Commun. 2020;11:2156.PubMedPubMedCentralCrossRef Silva-Fisher JM, Dang HX, White NM, Strand MS, Krasnick BA, Rozycki EB, et al. Long non-coding RNA RAMS11 promotes metastatic colorectal cancer progression. Nat Commun. 2020;11:2156.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Yamada A, Yu P, Lin W, Okugawa Y, Boland CR, Goel A. A RNA-Sequencing approach for the identification of novel long non-coding RNA biomarkers in colorectal cancer. Sci Rep. 2018;8:575.PubMedPubMedCentralCrossRef Yamada A, Yu P, Lin W, Okugawa Y, Boland CR, Goel A. A RNA-Sequencing approach for the identification of novel long non-coding RNA biomarkers in colorectal cancer. Sci Rep. 2018;8:575.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Bo H, Fan L, Li J, Liu Z, Zhang S, Shi L, et al. High Expression of lncRNA AFAP1-AS1 Promotes the Progression of Colon Cancer and Predicts Poor Prognosis. J Cancer. 2018;9:4677–83.PubMedPubMedCentralCrossRef Bo H, Fan L, Li J, Liu Z, Zhang S, Shi L, et al. High Expression of lncRNA AFAP1-AS1 Promotes the Progression of Colon Cancer and Predicts Poor Prognosis. J Cancer. 2018;9:4677–83.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Chen Q, Hu L, Chen K. Construction of a nomogram based on a hypoxia-related lncRNA signature to improve the prediction of gastric cancer prognosis. Front Genet. 2020;11:570325.PubMedPubMedCentralCrossRef Chen Q, Hu L, Chen K. Construction of a nomogram based on a hypoxia-related lncRNA signature to improve the prediction of gastric cancer prognosis. Front Genet. 2020;11:570325.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Guo YZ, Sun HH, Wang XT, Wang MT. Transcriptomic analysis reveals key lncRNAs associated with ribosomal biogenesis and epidermis differentiation in head and neck squamous cell carcinoma. J Zhejiang Univ Sci B. 2018;19:674–88.PubMedPubMedCentralCrossRef Guo YZ, Sun HH, Wang XT, Wang MT. Transcriptomic analysis reveals key lncRNAs associated with ribosomal biogenesis and epidermis differentiation in head and neck squamous cell carcinoma. J Zhejiang Univ Sci B. 2018;19:674–88.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Yao Y, Chen X, Lu S, Zhou C, Xu G, Yan Z, et al. Circulating long noncoding RNAs as biomarkers for predicting head and neck squamous cell carcinoma. Cell Physiol Biochem. 2018;50:1429–40.PubMedCrossRef Yao Y, Chen X, Lu S, Zhou C, Xu G, Yan Z, et al. Circulating long noncoding RNAs as biomarkers for predicting head and neck squamous cell carcinoma. Cell Physiol Biochem. 2018;50:1429–40.PubMedCrossRef
106.
Zurück zum Zitat Gong X, Siprashvili Z, Eminaga O, Shen Z, Sato Y, Kume H, et al. Novel lincRNA SLINKY is a prognostic biomarker in kidney cancer. Oncotarget. 2017;8:18657–69.PubMedPubMedCentralCrossRef Gong X, Siprashvili Z, Eminaga O, Shen Z, Sato Y, Kume H, et al. Novel lincRNA SLINKY is a prognostic biomarker in kidney cancer. Oncotarget. 2017;8:18657–69.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat James AR, Schroeder MP, Neumann M, Bastian L, Eckert C, Gökbuget N, et al. Long non-coding RNAs defining major subtypes of B cell precursor acute lymphoblastic leukemia. J Hematol Oncol. 2019;12:8.PubMedPubMedCentralCrossRef James AR, Schroeder MP, Neumann M, Bastian L, Eckert C, Gökbuget N, et al. Long non-coding RNAs defining major subtypes of B cell precursor acute lymphoblastic leukemia. J Hematol Oncol. 2019;12:8.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Li S, Ma Y, Tan Y, Ma X, Zhao M, Chen B, et al. Profiling and functional analysis of circular RNAs in acute promyelocytic leukemia and their dynamic regulation during all-trans retinoic acid treatment. Cell Death Dis. 2018;9:651.PubMedPubMedCentralCrossRef Li S, Ma Y, Tan Y, Ma X, Zhao M, Chen B, et al. Profiling and functional analysis of circular RNAs in acute promyelocytic leukemia and their dynamic regulation during all-trans retinoic acid treatment. Cell Death Dis. 2018;9:651.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Tirosh I, Izar B, Prakadan SM, Wadsworth MH 2nd, Treacy D, Trombetta JJ, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016;352:189–96.PubMedPubMedCentralCrossRef Tirosh I, Izar B, Prakadan SM, Wadsworth MH 2nd, Treacy D, Trombetta JJ, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016;352:189–96.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Maynard A, McCoach CE, Rotow JK, Harris L, Haderk F, Kerr DL, et al. Therapy-induced evolution of human lung cancer revealed by single-cell RNA sequencing. Cell. 2020;182(1232–51):e22. Maynard A, McCoach CE, Rotow JK, Harris L, Haderk F, Kerr DL, et al. Therapy-induced evolution of human lung cancer revealed by single-cell RNA sequencing. Cell. 2020;182(1232–51):e22.
112.
Zurück zum Zitat Nam AS, Chaligne R, Landau DA. Integrating genetic and non-genetic determinants of cancer evolution by single-cell multi-omics. Nat Rev Genet. 2020. Nam AS, Chaligne R, Landau DA. Integrating genetic and non-genetic determinants of cancer evolution by single-cell multi-omics. Nat Rev Genet. 2020.
113.
Zurück zum Zitat Berglund E, Maaskola J, Schultz N, Friedrich S, Marklund M, Bergenstråhle J, et al. Spatial maps of prostate cancer transcriptomes reveal an unexplored landscape of heterogeneity. Nat Commun. 2018;9:2419.PubMedPubMedCentralCrossRef Berglund E, Maaskola J, Schultz N, Friedrich S, Marklund M, Bergenstråhle J, et al. Spatial maps of prostate cancer transcriptomes reveal an unexplored landscape of heterogeneity. Nat Commun. 2018;9:2419.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Bakken TE, Hodge RD, Miller JA, Yao Z, Nguyen TN, Aevermann B, et al. Single-nucleus and single-cell transcriptomes compared in matched cortical cell types. PLoS ONE. 2018;13:e0209648.PubMedPubMedCentralCrossRef Bakken TE, Hodge RD, Miller JA, Yao Z, Nguyen TN, Aevermann B, et al. Single-nucleus and single-cell transcriptomes compared in matched cortical cell types. PLoS ONE. 2018;13:e0209648.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Selewa A, Dohn R, Eckart H, Lozano S, Xie B, Gauchat E, et al. Systematic comparison of high-throughput single-cell and single-nucleus transcriptomes during cardiomyocyte differentiation. Sci Rep. 2020;10:1535.PubMedPubMedCentralCrossRef Selewa A, Dohn R, Eckart H, Lozano S, Xie B, Gauchat E, et al. Systematic comparison of high-throughput single-cell and single-nucleus transcriptomes during cardiomyocyte differentiation. Sci Rep. 2020;10:1535.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Lim ZF, Ma PC. Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy. J Hematol Oncol. 2019;12:134.PubMedPubMedCentralCrossRef Lim ZF, Ma PC. Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy. J Hematol Oncol. 2019;12:134.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Wang Q, Guldner IH, Golomb SM, Sun L, Harris JA, Lu X, et al. Single-cell profiling guided combinatorial immunotherapy for fast-evolving CDK4/6 inhibitor-resistant HER2-positive breast cancer. Nat Commun. 2019;10:3817.PubMedPubMedCentralCrossRef Wang Q, Guldner IH, Golomb SM, Sun L, Harris JA, Lu X, et al. Single-cell profiling guided combinatorial immunotherapy for fast-evolving CDK4/6 inhibitor-resistant HER2-positive breast cancer. Nat Commun. 2019;10:3817.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Tanaka N, Katayama S, Reddy A, Nishimura K, Niwa N, Hongo H, et al. Single-cell RNA-seq analysis reveals the platinum resistance gene COX7B and the surrogate marker CD63. Cancer Med. 2018;7:6193–204.PubMedPubMedCentralCrossRef Tanaka N, Katayama S, Reddy A, Nishimura K, Niwa N, Hongo H, et al. Single-cell RNA-seq analysis reveals the platinum resistance gene COX7B and the surrogate marker CD63. Cancer Med. 2018;7:6193–204.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Debruyne DN, Dries R, Sengupta S, Seruggia D, Gao Y, Sharma B, et al. BORIS promotes chromatin regulatory interactions in treatment-resistant cancer cells. Nature. 2019;572:676–80.PubMedPubMedCentralCrossRef Debruyne DN, Dries R, Sengupta S, Seruggia D, Gao Y, Sharma B, et al. BORIS promotes chromatin regulatory interactions in treatment-resistant cancer cells. Nature. 2019;572:676–80.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Sharma A, Cao EY, Kumar V, Zhang X, Leong HS, Wong AML, et al. Longitudinal single-cell RNA sequencing of patient-derived primary cells reveals drug-induced infidelity in stem cell hierarchy. Nat Commun. 2018;9:4931.PubMedPubMedCentralCrossRef Sharma A, Cao EY, Kumar V, Zhang X, Leong HS, Wong AML, et al. Longitudinal single-cell RNA sequencing of patient-derived primary cells reveals drug-induced infidelity in stem cell hierarchy. Nat Commun. 2018;9:4931.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Shao F, Huang M, Meng F, Huang Q. Circular RNA SIgnature predicts gemcitabine resistance of pancreatic ductal adenocarcinoma. Front Pharmacol. 2018;9:584.PubMedPubMedCentralCrossRef Shao F, Huang M, Meng F, Huang Q. Circular RNA SIgnature predicts gemcitabine resistance of pancreatic ductal adenocarcinoma. Front Pharmacol. 2018;9:584.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Wang WT, Han C, Sun YM, Chen TQ, Chen YQ. Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol. 2019;12:55.PubMedPubMedCentralCrossRef Wang WT, Han C, Sun YM, Chen TQ, Chen YQ. Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol. 2019;12:55.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Shang J, Chen WM, Liu S, Wang ZH, Wei TN, Chen ZZ, et al. CircPAN3 contributes to drug resistance in acute myeloid leukemia through regulation of autophagy. Leuk Res. 2019;85:106198.PubMedCrossRef Shang J, Chen WM, Liu S, Wang ZH, Wei TN, Chen ZZ, et al. CircPAN3 contributes to drug resistance in acute myeloid leukemia through regulation of autophagy. Leuk Res. 2019;85:106198.PubMedCrossRef
124.
Zurück zum Zitat Suzuki K, Okuno Y, Kawashima N, Muramatsu H, Okuno T, Wang X, et al. MEF2D-BCL9 fusion gene is associated with high-risk acute B-cell precursor lymphoblastic leukemia in adolescents. J Clin Oncol. 2016;34:3451–9.PubMedCrossRef Suzuki K, Okuno Y, Kawashima N, Muramatsu H, Okuno T, Wang X, et al. MEF2D-BCL9 fusion gene is associated with high-risk acute B-cell precursor lymphoblastic leukemia in adolescents. J Clin Oncol. 2016;34:3451–9.PubMedCrossRef
125.
Zurück zum Zitat Pallarès V, Unzueta U, Falgàs A, Sánchez-García L, Serna N, Gallardo A, et al. An Auristatin nanoconjugate targeting CXCR4+ leukemic cells blocks acute myeloid leukemia dissemination. J Hematol Oncol. 2020;13:36.PubMedPubMedCentralCrossRef Pallarès V, Unzueta U, Falgàs A, Sánchez-García L, Serna N, Gallardo A, et al. An Auristatin nanoconjugate targeting CXCR4+ leukemic cells blocks acute myeloid leukemia dissemination. J Hematol Oncol. 2020;13:36.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Ding Y, Gao H, Zhang Y, Li Y, Vasdev N, Gao Y, et al. Alantolactone selectively ablates acute myeloid leukemia stem and progenitor cells. J Hematol Oncol. 2016;9:93.PubMedPubMedCentralCrossRef Ding Y, Gao H, Zhang Y, Li Y, Vasdev N, Gao Y, et al. Alantolactone selectively ablates acute myeloid leukemia stem and progenitor cells. J Hematol Oncol. 2016;9:93.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Bill M, Papaioannou D, Karunasiri M, Kohlschmidt J, Pepe F, Walker CJ, et al. Expression and functional relevance of long non-coding RNAs in acute myeloid leukemia stem cells. Leukemia. 2019;33:2169–82.PubMedCrossRef Bill M, Papaioannou D, Karunasiri M, Kohlschmidt J, Pepe F, Walker CJ, et al. Expression and functional relevance of long non-coding RNAs in acute myeloid leukemia stem cells. Leukemia. 2019;33:2169–82.PubMedCrossRef
129.
Zurück zum Zitat Burrell RA, Swanton C. Re-evaluating clonal dominance in cancer evolution. Trends Cancer. 2016;2:263–76.PubMedCrossRef Burrell RA, Swanton C. Re-evaluating clonal dominance in cancer evolution. Trends Cancer. 2016;2:263–76.PubMedCrossRef
130.
Zurück zum Zitat Shalek AK, Satija R, Adiconis X, Gertner RS, Gaublomme JT, Raychowdhury R, et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature. 2013;498:236–40.PubMedPubMedCentralCrossRef Shalek AK, Satija R, Adiconis X, Gertner RS, Gaublomme JT, Raychowdhury R, et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature. 2013;498:236–40.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Pan Y, Lu F, Fei Q, Yu X, Xiong P, Yu X, et al. Single-cell RNA sequencing reveals compartmental remodeling of tumor-infiltrating immune cells induced by anti-CD47 targeting in pancreatic cancer. J Hematol Oncol. 2019;12:124.PubMedPubMedCentralCrossRef Pan Y, Lu F, Fei Q, Yu X, Xiong P, Yu X, et al. Single-cell RNA sequencing reveals compartmental remodeling of tumor-infiltrating immune cells induced by anti-CD47 targeting in pancreatic cancer. J Hematol Oncol. 2019;12:124.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 2015;161:1202–14.PubMedPubMedCentralCrossRef Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 2015;161:1202–14.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Azizi E, Carr AJ, Plitas G, Cornish AE, Konopacki C, Prabhakaran S, et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell. 2018;174:1293–308.PubMedPubMedCentralCrossRef Azizi E, Carr AJ, Plitas G, Cornish AE, Konopacki C, Prabhakaran S, et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell. 2018;174:1293–308.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell. 2017;169:1342–56.PubMedCrossRef Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell. 2017;169:1342–56.PubMedCrossRef
135.
Zurück zum Zitat Cunha LD, Yang M, Carter R, Guy C, Harris L, Crawford JC, et al. LC3-associated phagocytosis in myeloid cells promotes tumor immune tolerance. Cell. 2018;175:429–41.PubMedPubMedCentralCrossRef Cunha LD, Yang M, Carter R, Guy C, Harris L, Crawford JC, et al. LC3-associated phagocytosis in myeloid cells promotes tumor immune tolerance. Cell. 2018;175:429–41.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat van Galen P, Hovestadt V, Wadsworth Ii MH, Hughes TK, Griffin GK, Battaglia S, et al. Single-cell RNA-seq reveals AML hierarchies relevant to disease progression and immunity. Cell. 2019;176:1265–81.PubMedPubMedCentralCrossRef van Galen P, Hovestadt V, Wadsworth Ii MH, Hughes TK, Griffin GK, Battaglia S, et al. Single-cell RNA-seq reveals AML hierarchies relevant to disease progression and immunity. Cell. 2019;176:1265–81.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Ren X, Zhong G, Zhang Q, Zhang L, Sun Y, Zhang Z. Reconstruction of cell spatial organization from single-cell RNA sequencing data based on ligand-receptor mediated self-assembly. Cell Res. 2020;30:763–78.PubMedPubMedCentralCrossRef Ren X, Zhong G, Zhang Q, Zhang L, Sun Y, Zhang Z. Reconstruction of cell spatial organization from single-cell RNA sequencing data based on ligand-receptor mediated self-assembly. Cell Res. 2020;30:763–78.PubMedPubMedCentralCrossRef
138.
139.
Zurück zum Zitat Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16:431–46.PubMedCrossRef Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16:431–46.PubMedCrossRef
140.
Zurück zum Zitat Singhal S, Bhojnagarwala PS, O’Brien S, Moon EK, Garfall AL, Rao AS, et al. Origin and role of a subset of tumor-associated neutrophils with antigen-presenting cell features in early-stage human lung cancer. Cancer Cell. 2016;30:120–35.PubMedPubMedCentralCrossRef Singhal S, Bhojnagarwala PS, O’Brien S, Moon EK, Garfall AL, Rao AS, et al. Origin and role of a subset of tumor-associated neutrophils with antigen-presenting cell features in early-stage human lung cancer. Cancer Cell. 2016;30:120–35.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Massara M, Bonavita O, Savino B, Caronni N, Mollica Poeta V, Sironi M, et al. ACKR2 in hematopoietic precursors as a checkpoint of neutrophil release and anti-metastatic activity. Nat Commun. 2018;9:676.PubMedPubMedCentralCrossRef Massara M, Bonavita O, Savino B, Caronni N, Mollica Poeta V, Sironi M, et al. ACKR2 in hematopoietic precursors as a checkpoint of neutrophil release and anti-metastatic activity. Nat Commun. 2018;9:676.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Ponzetta A, Carriero R, Carnevale S, Barbagallo M, Molgora M, Perucchini C, et al. Neutrophils driving unconventional T cells mediate resistance against murine sarcomas and selected human tumors. Cell. 2019;178(346–60):e24. Ponzetta A, Carriero R, Carnevale S, Barbagallo M, Molgora M, Perucchini C, et al. Neutrophils driving unconventional T cells mediate resistance against murine sarcomas and selected human tumors. Cell. 2019;178(346–60):e24.
143.
Zurück zum Zitat Janiszewska M, Tabassum DP, Castaño Z, Cristea S, Yamamoto KN, Kingston NL, et al. Subclonal cooperation drives metastasis by modulating local and systemic immune microenvironments. Nat Cell Biol. 2019;21:879–88.PubMedPubMedCentralCrossRef Janiszewska M, Tabassum DP, Castaño Z, Cristea S, Yamamoto KN, Kingston NL, et al. Subclonal cooperation drives metastasis by modulating local and systemic immune microenvironments. Nat Cell Biol. 2019;21:879–88.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Rath J, Bajwa G, Carreres B, Hoyer E, Gruber I, Martínez-Paniagua M, et al. Single-cell transcriptomics identifies multiple pathways underlying antitumor function of TCR- and CD8αβ-engineered human CD4 T cells. Sci Adv. 2020;6:eaaz7809. Rath J, Bajwa G, Carreres B, Hoyer E, Gruber I, Martínez-Paniagua M, et al. Single-cell transcriptomics identifies multiple pathways underlying antitumor function of TCR- and CD8αβ-engineered human CD4 T cells. Sci Adv. 2020;6:eaaz7809.
145.
Zurück zum Zitat Nirschl CJ, Suárez-Fariñas M, Izar B, Prakadan S, Dannenfelser R, Tirosh I, et al. IFNγ-dependent tissue-immune homeostasis is co-opted in the tumor microenvironment. Cell. 2017;170(127–41):e15. Nirschl CJ, Suárez-Fariñas M, Izar B, Prakadan S, Dannenfelser R, Tirosh I, et al. IFNγ-dependent tissue-immune homeostasis is co-opted in the tumor microenvironment. Cell. 2017;170(127–41):e15.
146.
Zurück zum Zitat Wang D, Lin J, Yang X, Long J, Bai Y, Yang X, et al. Combination regimens with PD-1/PD-L1 immune checkpoint inhibitors for gastrointestinal malignancies. J Hematol Oncol. 2019;12:42.PubMedPubMedCentralCrossRef Wang D, Lin J, Yang X, Long J, Bai Y, Yang X, et al. Combination regimens with PD-1/PD-L1 immune checkpoint inhibitors for gastrointestinal malignancies. J Hematol Oncol. 2019;12:42.PubMedPubMedCentralCrossRef
147.
148.
Zurück zum Zitat Zhao Z, Zheng L, Chen W, Weng W, Song J, Ji J. Delivery strategies of cancer immunotherapy: recent advances and future perspectives. J Hematol Oncol. 2019;12:126.PubMedPubMedCentralCrossRef Zhao Z, Zheng L, Chen W, Weng W, Song J, Ji J. Delivery strategies of cancer immunotherapy: recent advances and future perspectives. J Hematol Oncol. 2019;12:126.PubMedPubMedCentralCrossRef
149.
150.
Zurück zum Zitat Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015;14:561–84.PubMedCrossRef Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015;14:561–84.PubMedCrossRef
151.
152.
Zurück zum Zitat Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99.PubMedPubMedCentralCrossRef Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99.PubMedPubMedCentralCrossRef
153.
Zurück zum Zitat Li Z, Song W, Rubinstein M, Liu D. Recent updates in cancer immunotherapy: a comprehensive review and perspective of the 2018 China Cancer Immunotherapy Workshop in Beijing. J Hematol Oncol. 2018;11:142.PubMedPubMedCentralCrossRef Li Z, Song W, Rubinstein M, Liu D. Recent updates in cancer immunotherapy: a comprehensive review and perspective of the 2018 China Cancer Immunotherapy Workshop in Beijing. J Hematol Oncol. 2018;11:142.PubMedPubMedCentralCrossRef
154.
Zurück zum Zitat Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.PubMedPubMedCentralCrossRef Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16:275–87.PubMedPubMedCentralCrossRef Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16:275–87.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 2018;24:1550–8.PubMedPubMedCentralCrossRef Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 2018;24:1550–8.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Zilionis R, Engblom C, Pfirschke C, Savova V, Zemmour D, Saatcioglu HD, et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity. 2019;50:1317–34.PubMedPubMedCentralCrossRef Zilionis R, Engblom C, Pfirschke C, Savova V, Zemmour D, Saatcioglu HD, et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity. 2019;50:1317–34.PubMedPubMedCentralCrossRef
159.
Zurück zum Zitat Zhang L, Li Z, Skrzypczynska KM, Fang Q, Zhang W, O’Brien SA, et al. Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell. 2020;181:442–59.PubMedCrossRef Zhang L, Li Z, Skrzypczynska KM, Fang Q, Zhang W, O’Brien SA, et al. Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell. 2020;181:442–59.PubMedCrossRef
160.
Zurück zum Zitat Katzenelenbogen Y, Sheban F, Yalin A, Yofe I, Svetlichnyy D, Jaitin DA, et al. Coupled scRNA-Seq and intracellular protein activity reveal an immunosuppressive role of TREM2 in cancer. Cell. 2020;182:872–85.PubMedCrossRef Katzenelenbogen Y, Sheban F, Yalin A, Yofe I, Svetlichnyy D, Jaitin DA, et al. Coupled scRNA-Seq and intracellular protein activity reveal an immunosuppressive role of TREM2 in cancer. Cell. 2020;182:872–85.PubMedCrossRef
161.
Zurück zum Zitat Jiang T, Shi T, Zhang H, Hu J, Song Y, Wei J, et al. Tumor neoantigens: from basic research to clinical applications. J Hematol Oncol. 2019;12:93.PubMedPubMedCentralCrossRef Jiang T, Shi T, Zhang H, Hu J, Song Y, Wei J, et al. Tumor neoantigens: from basic research to clinical applications. J Hematol Oncol. 2019;12:93.PubMedPubMedCentralCrossRef
162.
Zurück zum Zitat McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–9.PubMedPubMedCentralCrossRef McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–9.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat Hacohen N, Fritsch EF, Carter TA, Lander ES, Wu CJ. Getting personal with neoantigen-based therapeutic cancer vaccines. Cancer Immunol Res. 2013;1:11–5.PubMedPubMedCentralCrossRef Hacohen N, Fritsch EF, Carter TA, Lander ES, Wu CJ. Getting personal with neoantigen-based therapeutic cancer vaccines. Cancer Immunol Res. 2013;1:11–5.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217–21.PubMedPubMedCentralCrossRef Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217–21.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Nathanson T, Ahuja A, Rubinsteyn A, Aksoy BA, Hellmann MD, Miao D, et al. Somatic mutations and neoepitope homology in melanomas treated with CTLA-4 blockade. Cancer Immunol Res. 2017;5:84–91.PubMedCrossRef Nathanson T, Ahuja A, Rubinsteyn A, Aksoy BA, Hellmann MD, Miao D, et al. Somatic mutations and neoepitope homology in melanomas treated with CTLA-4 blockade. Cancer Immunol Res. 2017;5:84–91.PubMedCrossRef
166.
Zurück zum Zitat Safonov A, Jiang T, Bianchini G, Győrffy B, Karn T, Hatzis C, et al. Immune gene expression is associated with genomic aberrations in breast cancer. Cancer Res. 2017;77:3317–24.PubMedCrossRef Safonov A, Jiang T, Bianchini G, Győrffy B, Karn T, Hatzis C, et al. Immune gene expression is associated with genomic aberrations in breast cancer. Cancer Res. 2017;77:3317–24.PubMedCrossRef
167.
Zurück zum Zitat Karasaki T, Nagayama K, Kuwano H, Nitadori JI, Sato M, Anraku M, et al. Prediction and prioritization of neoantigens: integration of RNA sequencing data with whole-exome sequencing. Cancer Sci. 2017;108:170–7.PubMedPubMedCentralCrossRef Karasaki T, Nagayama K, Kuwano H, Nitadori JI, Sato M, Anraku M, et al. Prediction and prioritization of neoantigens: integration of RNA sequencing data with whole-exome sequencing. Cancer Sci. 2017;108:170–7.PubMedPubMedCentralCrossRef
168.
Zurück zum Zitat Nejo T, Matsushita H, Karasaki T, Nomura M, Saito K, Tanaka S, et al. Reduced neoantigen expression revealed by longitudinal multiomics as a possible immune evasion mechanism in glioma. Cancer Immunol Res. 2019;7:1148–61.PubMedCrossRef Nejo T, Matsushita H, Karasaki T, Nomura M, Saito K, Tanaka S, et al. Reduced neoantigen expression revealed by longitudinal multiomics as a possible immune evasion mechanism in glioma. Cancer Immunol Res. 2019;7:1148–61.PubMedCrossRef
169.
Zurück zum Zitat Lv JW, Zheng ZQ, Wang ZX, Zhou GQ, Chen L, Mao YP, et al. Pan-cancer genomic analyses reveal prognostic and immunogenic features of the tumor melatonergic microenvironment across 14 solid cancer types. J Pineal Res. 2019;66:e12557.PubMedCrossRef Lv JW, Zheng ZQ, Wang ZX, Zhou GQ, Chen L, Mao YP, et al. Pan-cancer genomic analyses reveal prognostic and immunogenic features of the tumor melatonergic microenvironment across 14 solid cancer types. J Pineal Res. 2019;66:e12557.PubMedCrossRef
170.
Zurück zum Zitat Linxweiler M, Kuo F, Katabi N, Lee M, Nadeem Z, Dalin MG, et al. The immune microenvironment and neoantigen landscape of aggressive salivary gland carcinomas differ by subtype. Clin Cancer Res. 2020;26:2859–70.PubMedCrossRefPubMedCentral Linxweiler M, Kuo F, Katabi N, Lee M, Nadeem Z, Dalin MG, et al. The immune microenvironment and neoantigen landscape of aggressive salivary gland carcinomas differ by subtype. Clin Cancer Res. 2020;26:2859–70.PubMedCrossRefPubMedCentral
171.
Zurück zum Zitat Kim S, Kim HS, Kim E, Lee MG, Shin EC, Paik S, et al. Neopepsee: accurate genome-level prediction of neoantigens by harnessing sequence and amino acid immunogenicity information. Ann Oncol. 2018;29:1030–6.PubMedCrossRef Kim S, Kim HS, Kim E, Lee MG, Shin EC, Paik S, et al. Neopepsee: accurate genome-level prediction of neoantigens by harnessing sequence and amino acid immunogenicity information. Ann Oncol. 2018;29:1030–6.PubMedCrossRef
172.
Zurück zum Zitat Wang TY, Wang L, Alam SK, Hoeppner LH, Yang R. ScanNeo: identifying indel-derived neoantigens using RNA-Seq data. Bioinformatics. 2019;35:4159–61.PubMedCrossRef Wang TY, Wang L, Alam SK, Hoeppner LH, Yang R. ScanNeo: identifying indel-derived neoantigens using RNA-Seq data. Bioinformatics. 2019;35:4159–61.PubMedCrossRef
173.
Zurück zum Zitat Zhang Z, Zhou C, Tang L, Gong Y, Wei Z, Zhang G, et al. ASNEO: Identification of personalized alternative splicing based neoantigens with RNA-seq. Aging (Albany NY). 2020;12:14633–48.CrossRef Zhang Z, Zhou C, Tang L, Gong Y, Wei Z, Zhang G, et al. ASNEO: Identification of personalized alternative splicing based neoantigens with RNA-seq. Aging (Albany NY). 2020;12:14633–48.CrossRef
175.
Zurück zum Zitat Lu YC, Zheng Z, Robbins PF, Tran E, Prickett TD, Gartner JJ, et al. An efficient single-cell RNA-seq approach to identify neoantigen-specific T cell receptors. Mol Ther. 2018;26:379–89.PubMedCrossRef Lu YC, Zheng Z, Robbins PF, Tran E, Prickett TD, Gartner JJ, et al. An efficient single-cell RNA-seq approach to identify neoantigen-specific T cell receptors. Mol Ther. 2018;26:379–89.PubMedCrossRef
176.
Zurück zum Zitat Engström P, Steijger T, Sipos B, Grant G, Kahles A, Rätsch G, et al. Systematic evaluation of spliced alignment programs for RNA-seq data. Nat Methods. 2013;10:1185–91.PubMedPubMedCentralCrossRef Engström P, Steijger T, Sipos B, Grant G, Kahles A, Rätsch G, et al. Systematic evaluation of spliced alignment programs for RNA-seq data. Nat Methods. 2013;10:1185–91.PubMedPubMedCentralCrossRef
177.
178.
Zurück zum Zitat Quail MA, Smith M, Coupland P, Otto TD, Harris SR, Connor TR, et al. A tale of three next generation sequencing platforms: comparison of Ion Torrent, Pacific Biosciences and Illumina MiSeq sequencers. BMC Genomics. 2012;13:341.PubMedPubMedCentralCrossRef Quail MA, Smith M, Coupland P, Otto TD, Harris SR, Connor TR, et al. A tale of three next generation sequencing platforms: comparison of Ion Torrent, Pacific Biosciences and Illumina MiSeq sequencers. BMC Genomics. 2012;13:341.PubMedPubMedCentralCrossRef
179.
Zurück zum Zitat Tilgner H, Grubert F, Sharon D, Snyder MP. Defining a personal, allele-specific, and single-molecule long-read transcriptome. Proc Natl Acad Sci USA. 2014;111:9869–74.PubMedCrossRefPubMedCentral Tilgner H, Grubert F, Sharon D, Snyder MP. Defining a personal, allele-specific, and single-molecule long-read transcriptome. Proc Natl Acad Sci USA. 2014;111:9869–74.PubMedCrossRefPubMedCentral
180.
Zurück zum Zitat Stark R, Grzelak M, Hadfield J. RNA sequencing: the teenage years. Nat Rev Genet. 2019;20:631–56.PubMedCrossRef Stark R, Grzelak M, Hadfield J. RNA sequencing: the teenage years. Nat Rev Genet. 2019;20:631–56.PubMedCrossRef
181.
Zurück zum Zitat Antipov D, Korobeynikov A, McLean J, Pevzner P. hybridSPAdes: an algorithm for hybrid assembly of short and long reads. Bioinformatics. 2016;32:1009–15.PubMedCrossRef Antipov D, Korobeynikov A, McLean J, Pevzner P. hybridSPAdes: an algorithm for hybrid assembly of short and long reads. Bioinformatics. 2016;32:1009–15.PubMedCrossRef
182.
Zurück zum Zitat Boluki S, Zamani Dadaneh S, Qian X, Dougherty E. Optimal clustering with missing values. BMC Bioinform. 2019;20:321.CrossRef Boluki S, Zamani Dadaneh S, Qian X, Dougherty E. Optimal clustering with missing values. BMC Bioinform. 2019;20:321.CrossRef
183.
Zurück zum Zitat Hicks S, Townes F, Teng M, Irizarry R. Missing data and technical variability in single-cell RNA-sequencing experiments. Biostatistics. 2018;19:562–78.PubMedCrossRef Hicks S, Townes F, Teng M, Irizarry R. Missing data and technical variability in single-cell RNA-sequencing experiments. Biostatistics. 2018;19:562–78.PubMedCrossRef
185.
Zurück zum Zitat Song F, Chan G, Wei Y. Flexible experimental designs for valid single-cell RNA-sequencing experiments allowing batch effects correction. Nat Commun. 2020;11:3274.PubMedPubMedCentralCrossRef Song F, Chan G, Wei Y. Flexible experimental designs for valid single-cell RNA-sequencing experiments allowing batch effects correction. Nat Commun. 2020;11:3274.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Buenrostro J, Giresi P, Zaba L, Chang H, Greenleaf W. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods. 2013;10:1213–8.PubMedPubMedCentralCrossRef Buenrostro J, Giresi P, Zaba L, Chang H, Greenleaf W. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods. 2013;10:1213–8.PubMedPubMedCentralCrossRef
187.
Zurück zum Zitat Yang C, Ma L, Xiao D, Ying Z, Jiang X, Lin Y. Sparassis latifolia Integration of ATAC-Seq and RNA-Seq Identifies Key Genes in Light-Induced Primordia Formation of. Int J Mol Sci. 2019;21. Yang C, Ma L, Xiao D, Ying Z, Jiang X, Lin Y. Sparassis latifolia Integration of ATAC-Seq and RNA-Seq Identifies Key Genes in Light-Induced Primordia Formation of. Int J Mol Sci. 2019;21.
188.
Zurück zum Zitat Wu X, Yang Y, Zhong C, Guo Y, Wei T, Li S, et al. Epinephelus coioides Integration of ATAC-seq and RNA-seq Unravels Chromatin Accessibility during Sex Reversal in Orange-Spotted Grouper (Epinephelus coioides). Int J Mol Sci. 2020;21. Wu X, Yang Y, Zhong C, Guo Y, Wei T, Li S, et al. Epinephelus coioides Integration of ATAC-seq and RNA-seq Unravels Chromatin Accessibility during Sex Reversal in Orange-Spotted Grouper (Epinephelus coioides). Int J Mol Sci. 2020;21.
189.
Zurück zum Zitat Simonis M, Kooren J, de Laat W. An evaluation of 3C-based methods to capture DNA interactions. Nat Methods. 2007;4:895–901.PubMedCrossRef Simonis M, Kooren J, de Laat W. An evaluation of 3C-based methods to capture DNA interactions. Nat Methods. 2007;4:895–901.PubMedCrossRef
190.
Zurück zum Zitat Lieberman-Aiden E, van Berkum N, Williams L, Imakaev M, Ragoczy T, Telling A, et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science. 2009;326:289–93.PubMedPubMedCentralCrossRef Lieberman-Aiden E, van Berkum N, Williams L, Imakaev M, Ragoczy T, Telling A, et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science. 2009;326:289–93.PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Mifsud B, Tavares-Cadete F, Young A, Sugar R, Schoenfelder S, Ferreira L, et al. Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C. Nat Genet. 2015;47:598–606.PubMedCrossRef Mifsud B, Tavares-Cadete F, Young A, Sugar R, Schoenfelder S, Ferreira L, et al. Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C. Nat Genet. 2015;47:598–606.PubMedCrossRef
192.
Zurück zum Zitat Crane E, Bian Q, McCord R, Lajoie B, Wheeler B, Ralston E, et al. Condensin-driven remodelling of X chromosome topology during dosage compensation. Nature. 2015;523:240–4.PubMedPubMedCentralCrossRef Crane E, Bian Q, McCord R, Lajoie B, Wheeler B, Ralston E, et al. Condensin-driven remodelling of X chromosome topology during dosage compensation. Nature. 2015;523:240–4.PubMedPubMedCentralCrossRef
193.
Zurück zum Zitat Chen H, Seaman L, Liu S, Ried T, Rajapakse I. Chromosome conformation and gene expression patterns differ profoundly in human fibroblasts grown in spheroids versus monolayers. Nucleus. 2017;8:383–91.PubMedPubMedCentralCrossRef Chen H, Seaman L, Liu S, Ried T, Rajapakse I. Chromosome conformation and gene expression patterns differ profoundly in human fibroblasts grown in spheroids versus monolayers. Nucleus. 2017;8:383–91.PubMedPubMedCentralCrossRef
194.
Zurück zum Zitat Vara C, Paytuví-Gallart A, Cuartero Y, Le Dily F, Garcia F, Salvà-Castro J, et al. Three-dimensional genomic structure and cohesin occupancy correlate with transcriptional activity during spermatogenesis. Cell Rep. 2019;28(352–67):e9. Vara C, Paytuví-Gallart A, Cuartero Y, Le Dily F, Garcia F, Salvà-Castro J, et al. Three-dimensional genomic structure and cohesin occupancy correlate with transcriptional activity during spermatogenesis. Cell Rep. 2019;28(352–67):e9.
197.
Zurück zum Zitat Parker M, Knop K, Sherwood A, Schurch N, Mackinnon K, Gould P, et al. Nanopore direct RNA sequencing maps the complexity of Arabidopsis mRNA processing and mA modification. eLife. 2020;9. Parker M, Knop K, Sherwood A, Schurch N, Mackinnon K, Gould P, et al. Nanopore direct RNA sequencing maps the complexity of Arabidopsis mRNA processing and mA modification. eLife. 2020;9.
198.
Zurück zum Zitat Zhang S, Li R, Zhang L, Chen S, Xie M, Yang L, et al. New insights into Arabidopsis transcriptome complexity revealed by direct sequencing of native RNAs. Nucleic Acids Res. 2020;48:7700–11.PubMedPubMedCentralCrossRef Zhang S, Li R, Zhang L, Chen S, Xie M, Yang L, et al. New insights into Arabidopsis transcriptome complexity revealed by direct sequencing of native RNAs. Nucleic Acids Res. 2020;48:7700–11.PubMedPubMedCentralCrossRef
199.
Zurück zum Zitat Pan X, Zhang H, Xu D, Chen J, Chen W, Gan S, et al. Identification of a novel cancer stem cell subpopulation that promotes progression of human fatal renal cell carcinoma by single-cell RNA-seq analysis. Int J Biol Sci. 2020;16:3149–62.PubMedPubMedCentralCrossRef Pan X, Zhang H, Xu D, Chen J, Chen W, Gan S, et al. Identification of a novel cancer stem cell subpopulation that promotes progression of human fatal renal cell carcinoma by single-cell RNA-seq analysis. Int J Biol Sci. 2020;16:3149–62.PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Li Y, Chen C, Chen J, Lai Y, Wang S, Jiang S, et al. Single-cell analysis reveals immune modulation and metabolic switch in tumor-draining lymph nodes. Oncoimmunology. 2020;9:1830513.PubMedPubMedCentralCrossRef Li Y, Chen C, Chen J, Lai Y, Wang S, Jiang S, et al. Single-cell analysis reveals immune modulation and metabolic switch in tumor-draining lymph nodes. Oncoimmunology. 2020;9:1830513.PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Giladi A, Cohen M, Medaglia C, Baran Y, Li B, Zada M, et al. Dissecting cellular crosstalk by sequencing physically interacting cells. Nat Biotechnol. 2020;38:629–37.PubMedCrossRef Giladi A, Cohen M, Medaglia C, Baran Y, Li B, Zada M, et al. Dissecting cellular crosstalk by sequencing physically interacting cells. Nat Biotechnol. 2020;38:629–37.PubMedCrossRef
202.
Zurück zum Zitat Caruso F, Garofano L, D'Angelo F, Yu K, Tang F, Yuan J, et al. A map of tumor-host interactions in glioma at single-cell resolution. GigaScience. 2020;9. Caruso F, Garofano L, D'Angelo F, Yu K, Tang F, Yuan J, et al. A map of tumor-host interactions in glioma at single-cell resolution. GigaScience. 2020;9.
203.
Zurück zum Zitat Jaitin D, Weiner A, Yofe I, Lara-Astiaso D, Keren-Shaul H, David E, et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell. 2016;167:1883–96.PubMedCrossRef Jaitin D, Weiner A, Yofe I, Lara-Astiaso D, Keren-Shaul H, David E, et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell. 2016;167:1883–96.PubMedCrossRef
204.
Zurück zum Zitat Datlinger P, Rendeiro A, Schmidl C, Krausgruber T, Traxler P, Klughammer J, et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat Methods. 2017;14:297–301.PubMedPubMedCentralCrossRef Datlinger P, Rendeiro A, Schmidl C, Krausgruber T, Traxler P, Klughammer J, et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat Methods. 2017;14:297–301.PubMedPubMedCentralCrossRef
205.
Zurück zum Zitat Genga R, Kernfeld E, Parsi K, Parsons T, Ziller M, Maehr R. Single-cell RNA-sequencing-based CRISPRi screening resolves molecular drivers of early human endoderm development. Cell Rep. 2019;27:708–18.PubMedPubMedCentralCrossRef Genga R, Kernfeld E, Parsi K, Parsons T, Ziller M, Maehr R. Single-cell RNA-sequencing-based CRISPRi screening resolves molecular drivers of early human endoderm development. Cell Rep. 2019;27:708–18.PubMedPubMedCentralCrossRef
206.
Zurück zum Zitat Qiu Q, Hu P, Qiu X, Govek K, Cámara P, Wu H. Massively parallel and time-resolved RNA sequencing in single cells with scNT-seq. Nat Methods. 2020;17:991–1001.PubMedCrossRefPubMedCentral Qiu Q, Hu P, Qiu X, Govek K, Cámara P, Wu H. Massively parallel and time-resolved RNA sequencing in single cells with scNT-seq. Nat Methods. 2020;17:991–1001.PubMedCrossRefPubMedCentral
207.
Zurück zum Zitat McFarland J, Paolella B, Warren A, Geiger-Schuller K, Shibue T, Rothberg M, et al. Multiplexed single-cell transcriptional response profiling to define cancer vulnerabilities and therapeutic mechanism of action. Nat commun. 2020;11:4296.PubMedPubMedCentralCrossRef McFarland J, Paolella B, Warren A, Geiger-Schuller K, Shibue T, Rothberg M, et al. Multiplexed single-cell transcriptional response profiling to define cancer vulnerabilities and therapeutic mechanism of action. Nat commun. 2020;11:4296.PubMedPubMedCentralCrossRef
208.
Zurück zum Zitat Sun W, Dong H, Balaz M, Slyper M, Drokhlyansky E, Colleluori G, et al. snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis. Nature. 2020;587:98–102.PubMedCrossRef Sun W, Dong H, Balaz M, Slyper M, Drokhlyansky E, Colleluori G, et al. snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis. Nature. 2020;587:98–102.PubMedCrossRef
Metadaten
Titel
RNA sequencing: new technologies and applications in cancer research
verfasst von
Mingye Hong
Shuang Tao
Ling Zhang
Li-Ting Diao
Xuanmei Huang
Shaohui Huang
Shu-Juan Xie
Zhen-Dong Xiao
Hua Zhang
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2020
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-01005-x

Weitere Artikel der Ausgabe 1/2020

Journal of Hematology & Oncology 1/2020 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.