Single‑cell multi‑omics advances in lymphoma research (Review)

  • Authors:
    • Chanjuan Jin
    • Di Zhou
    • Jun Li
    • Lintao Bi
    • Lisha Li
  • View Affiliations

  • Published online on: August 23, 2023     https://doi.org/10.3892/or.2023.8621
  • Article Number: 184
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The evolution of lymphoma is a multifactorial process that leads to unavoidable lymphoma heterogeneity in the form of genetic mutations, chromosomal translocations and other variations. Multi‑omics analyses based on single‑cell assays can reveal and characterize tumor components, enabling us to determine the timing of mutations and to profile disease progression. Increasing numbers of studies are using single‑cell transcriptomics to unravel the mechanisms of lymphoma evolution, drug resistance and therapeutic approaches. Various single‑cell multi‑omics measurements involving genomics, transcriptomics and epigenomics have improved knowledge of the complex lymphatic system and made it possible to obtain individualized and precise tumor biological characteristics, which cannot be accessed from bulk cell analysis, and this can facilitate individualized treatment. In the present review, the advances in multi‑omics analysis based on single‑cell assays of lymphoma specimens were systematically discussed, including the sequencing of the single‑cell from genomic and transcriptomic perspectives, the landscape of the lymphoma microenvironment, the development of single‑cell histology biomarkers, the identification of lymphoma origin and evolution, as well as the current challenges and future prospects of single‑cell multi‑omics. The authors' insights may contribute to the exploration of novel lymphoma biomarkers and the discovery of efficient treatment combinations that target immunological checkpoints and underlying molecular mechanisms.

Introduction

Lymphoma is a sizable collection of immunocyte-derived malignancies that exhibit molecular aberrations in various aspects (1). Variations in genotype, phenotype and metabolic events lead to changes in lymphoma cell morphology, progression rate and metastatic potential (2). However, conventional high-volume detection techniques, such as chromatin immunoprecipitation, real-time quantitative polymerase chain reaction, and western blotting, do not provide comprehensive analyses of the tumor cell landscape (3). Previously, novel techniques and analytic tools have been developed to promote research and understand lymphoma. These techniques and tools include capturing mRNAs from cell lysates using arrays of thousands of oligonucleotide probes (4), identifying differentially expressed genes and signaling pathways between two samples using Gene Set Enrichment Analysis (5), and inferring sample cell composition by deconvolution of the bulk transcriptome (6,7). Nevertheless, these techniques and tools fundamentally reflect the functions of lymphoma cells as a whole and do not describe the molecular mechanisms at the level of a single lymphoma cell. Presently, single-cell sequencing technologies involving genomics, transcriptomics, proteomics and epigenomics employ high-resolution characterization, which allows researchers to understand precisely tumor heterogeneity, the tumor microenvironment (TME), and tumor progression (8). Single-cell spatial transcriptomics is also used to understand these tumor events (9) (Fig. 1).

By utilizing the rapidly-evolving single-cell multi-omics techniques to identify the different types of lymphoma, it is possible to reveal heterogeneity (1,10,11) and drug resistance (12), identify presumptive precancerous groups (13) and evolutionary processes (14,15), identify new biomarkers (16,17), and explore TMEs (18,19). Lymphoma research has advanced to a more sophisticated level with the existence of single-cell multi-omics approaches. Using single-cell RNA sequencing (scRNA-seq) as an example, in Fig. 2 the differences and similarities between the workflow of scRNA-seq and bulk RNA seq are illustrated.

Presently, the generally acknowledged platforms of high-throughput single-cell sequencing are Drop-Seq (20), inDrop (21), and 10X Genomics (22). The scRNA-seq technologies commonly used in lymphoma analysis are listed in Table I.

Table I.

Common scRNA-seq techniques for lymphoma analysis.

Table I.

Common scRNA-seq techniques for lymphoma analysis.

TechniquesPlatformMeasure Characteristics(Refs.)
IFCsThe Fluidigm C1 SystemRNA sequence
  • produce high-quality gene expression readouts;

  • unstable capture efficiency;

  • unavoidable size bias;

(2326)
Seq-well, CytoseqMicrowell Platforms
  • cheap;

  • portable;

  • environmentally friendly;

(2730)
10X GenomicsChromium System by 10X Genomics
  • high efficiency;

  • prohibitive cost;

(31,32)
MARS-seq, SMART-seq3Fluorescence activating cell sorter (FACS)
  • large scale;

  • high sensitivity and precision;

(33)
CITE-seqExisting scRNA-seq platforms
  • not limited to a certain size or a certain number of cells to be tested;

  • high precision;

(34,35)

Single-cell multi-omics to characterize immune cells

The multiparametric classification approach from the World Health Organization and the revised European and American Lymphoma classification (36) have tremendously aided lymphoma clinical and translational research, which serves as a foundation for discovering the causes of molecular changes in these tumors (37). Although a diagnosis of Hodgkin's lymphoma according to the existence of Reed-Sternberg (R-S) cells has been proposed for a long time, the lymphomagenesis of Hodgkin's lymphoma remains unclear (38). Given that malignant R-S cells derived from germinal center (GC) B cells (39,40) represent a minority of lymphoma cells, an inadequate biopsy may fail to identify these cells and make an accurate diagnosis (41). Non-Hodgkin's lymphoma accounts for ~3% of all cancer occurrences and deaths worldwide, making it the most common malignant tumor in the hematological field (42). Each of its >40 subtypes has specific driver genetic mutations and distinct risk factors (43). Thus, it is the extensive and remarkable heterogeneity of lymphoma cells that requires to finely characterize lymphoma cell molecules.

Currently, there is rapid progress in the accurate characterization of single-cell immune cells, which allows for comprehensive detection of lymphoma and precise clinical diagnosis. For instance, phenotyping CD molecules (CD3+, CD4+ and CD8+) on T cells can help identify the specific cellular status and advance the development of immunotherapy (44). It has been shown that using just 6 biomarkers (CD95, CD73, RB, CD39, CD38 and CD27) can define tonsillar (and also lymph node) B cells and characterize B cells (45). Furthermore, single-cell sequencing analysis of NK cells from patients with acute myeloid leukemia (AML) revealed similar patient-specific patterns of NKp30 and CD27 expression, and downregulation of CD160 transcription on NK cells is related to the reduced survival of patients with AML, suggesting that CD160 is promising precision medicine biomarker (46). A single-cell histological assay of a subpopulation of T cells associated with Hodgkin's lymphoma revealed prominent expression of the immunosuppressor receptor LAG3, which offers a new approach targeting immune checkpoints in Hodgkin's lymphoma, second only to PD-1 (47). High-resolution clustering and combinatorial gene characterization using large scRNA-seq datasets of human γδ T cells allow in-depth characterization of these cells, which can specifically identify T cell receptor (TCR) Vδ1 and TCR Vδ2 subpopulations (48). Repertoire and Gene Expression by Sequencing (RAGE-seq), which combines short-read transcript analysis based on single-cell libraries of barcodes with long-read sequences and targeted capture of B cell receptor (BCR) or the TCR mRNA transcriptome, can identify the full-length antigen receptor profiles of B or T cells (49).

These single-cell multi-omics techniques have been applied to study various cells in the human immune cell population, and they have great potential in identifying new subpopulations of lymphoma cells or in exploring new biomarkers.

Single-cell multi-omics to define the transcriptomic, proteomic and epigenomic features of lymphoma

In the field of tumor research, single-cell multi-omics techniques have transformed the existing knowledge of the biological features of lymphoma lesions (3). Different technologies, such as scRNA-seq, scDNA-seq, single-cell chromatin immunoprecipitation-seq, and methylation sequencing, have been widely used in lymphoma research, and their specific features have been well characterized.

At the genetic level, the rather mature single-cell combinatorial indexing RNA sequencing can provide information on lymphoma copy number variation (50). In addition, genomic mutations can also be identified in different types of lymphoma with single-cell precision. Stratified clustering of NK cells showed that a total of 56 genomic mutations based on the JAK-STAT pathway and TP53 were present in 102 EBV patient samples (51), which is in line with previous bulk analysis. Single-cell exome sequencing and protein profiling (SNV and small insertions) of GI-DLBCL and non-GI-DLBCL exhibited changes in gene mutation frequency, among which ID3, CCDN3 and TP53 were increased, while BCL2, CREBBP and MYD88 were significantly decreased (52). In an exploration of lymphoma molecular drivers, researchers identified signature mutations in CD79B and MYD88 L265P as well as a CDKN2A deletion in BCR and NF-kB pathways (53). As for the clinical application, based on gene mutation data from the NF-κB pathway, CCND1, as well as ATM according to scRNA-seq, researchers classified mantle cell lymphoma into four genetic subgroups as C1-C4 for the first time (54). In another study, single-cell genome and exome sequencing showed that CDKN1B, SMARCB1 and DAZAP1 expression has a negative impact on patient prognosis, suggesting that they could be used as biomarkers for patient prognosis (55). Lymphoma heterogeneity has been investigated among primary BM specimens by applying single-cell targeted DNA sequencing through the Fluidigm C1 system (56). As for the molecular mechanism, single-cell gene transcription analysis of IG-non MYC translocations in Burkitt's lymphoma, the most common type of childhood lymphoma, revealed MYC oncogene dysregulation and demonstrated that at least one BL subgroup precursor could be expressed in each patient (57). Furthermore, KIR2DL4 was reported to be overexpressed in malignant NK cells by scRNA-seq, and KIR2DL4 promotes lymphoma pathogenesis by mediating NK cell proliferation and apoptosis, which are associated with certain signaling pathways such as AKT and NF-κB (58). After analyzing the transcriptome of tens of thousands single tumor cells from 6 primary FL patients, the results revealed that malignant B cells showed more pronounced expression of the BCL2 gene and absence of expression of MHC-II and CD52 genes. Moreover, the results also identified the co-expression of B2M and CEBPA genes in Treg cells with immune checkpoint molecules, which helps further map the network of genes included within the immunoregulatory mechanism (59). Thus, single-cell analysis could help expand tumor genomic studies in different classes of lymphoma and accelerate the transition from genetic analysis to pharmacodynamic therapy in the future.

Targeted and functional proteomic analysis enables bulk and thorough studies of proteins, particularly for high-throughput screening of promising biomarkers from complex tumor biological microenvironments. Currently, proteomics has become one of the key fields of lymphoma research, and it is considered as the most suitable approach to discovering new biomarkers and personalized therapies, as well as it being a high-throughput technique for revealing genotype-phenotype uncoupling in lymphoma and tracking proteomic dynamics in relapsed patients (60). For example, through the E1A-binding protein p300, mice with knockdown BCL6 expression on the surface of lymphomas can block oncogenic transformation by inhibiting the acetylation of Lys132 in the p53 gene, which upregulates cystein-1 to reduce BCL6 stability (61).

Single-cell epigenomic analysis of lymphoma has also greatly aided the analysis lymphoma diagnostic and prognostic processes. The epigenome of lymphoma is defined by the regulation of normal gene expression in cells, modified by histone proteins and DNA, plus the action of non-coding RNA (62). Previous studies have demonstrated strong associations between the differentiating expression of miRNAs and the pathogenic progression of NKTCL tumors by single-cell techniques targeting P53, cell-cycle related genes and MAPK pathways (63,64). The reduced functions of miR-26 and miR-101 cause EZH2 overexpression, whereas the over-activation of miR-223 contributes to PRDM1 suppression (65,66). Hypermethylation in the promoter region was studied by whole methylation assay and methylation site-specific validation, and the results demonstrated that the functions of TET2, PTPRK, SOCS6 and PTPN6 can increase the expression of methylated genes (67,68). Functionally, TET2 inactivation may lead to hypermethylation of lymphoma promoters, and the negative regulation of JAK-STAT by PTPRK, SOCS6 and PTPN6 suggests an alternative mechanism associated with the activation of the JAK-STAT signaling pathway (67).

Besides gene transcription, protein expression and epigenetic inheritance, single-cell technology also has roles in other areas. For example, scATAC-seq can identify specific chromatin motifs (69), whereas scNGS can provide information about somatic mutations and cellular heterogeneity (70). It is even possible to reveal the mutation profiles of B cells in the entire life cycle of a human by single-cell whole genome sequencing, thus identifying potential cancer-causing mutations (71)

Taken together, single-cell multi-omics techniques provide the productive characterization of the biological features and intrinsic dynamics of lymphoma, which may widen our knowledge of lymphoma and accelerate the pace of clinical diagnosis and treatment.

Single-cell multi-omics reveal lymphomagenesis and clonal evolution

A comprehensive understanding of tumorigenesis and clonal evolution can help to unravel tumor heterogeneity as well as targeted therapies for tumors. Although protein-altering lesions have always been detected in lymphoma diagnosis, the tumor's clonal structures and the initial progress remain unclear. Quantitative single-cell genomics based on mass spectrometry is used specifically to understand the pathogenesis of lymphomas, and its incremental value in deciphering the complexity of lymphoma entities to regulate the heterogeneity of molecular mechanisms is currently a popular diagnostic approach and therapeutic strategy (72). Single-cell multi-omics techniques can reveal the clonal specificity of lymphoma and establish the order of genetic events in lymphoma.

Lymphoma cells may have translocation mutations in gene bases, although few types of lymphoma can result from recurrent translocations involving specific genes. For example, the IGH translocation on 14q32 is responsible for a certain proportion of mature B-cell tumors (73). Meanwhile, regulatory chromatin genes (CREBBP, EZH2 and KMT2D) have also been shown to mutate in the early drivers in lymphoma (74). These mutations indicate the initiation phase of lymphoma development.

The clonal evolution of lymphoma is inevitably accompanied by changes in the transcriptome of genes, and to clarify genotyping, numerous experiments have been performed using single-cell sequencing techniques to describe the clonal progression of lymphoma. Fluorescence in situ hybridization results identified that lymphoma cells had MYC rearrangements that were not BCL2 or BCL6, but involved the immunoglobulin light or heavy chain gene and the 8q24 region, which are markers of lymphoma lesions (75). This finding is remarkable because the prognosis of lymphoma with MYC translocation is often worse than that of lymphoma alone (76). However, this is a limited ordering study of lymphoma using bulk samples, and it did not explore the tumor evolution. Single-cell capture and WGA of microfluidic specimens have proved RAG-mediated structural vibrations to precede lymphoma (77). The acquisition of fusion genes and the loss of 9p21 (CDKN2A/B) accounts for the intermediated clonal evolution of lymphoma (56). Gene rearrangements and chromosomal translocations such as these play a non-negligible role in promoting lymphoma development.

Multi-step mutations in NOTCH, the STAT3 signaling pathway, and TCR are always late events in the evolution of lymphoma and can further accelerate the proliferative potential of tumor cells, leading to the development of highly malignant clones that can lead to disease onset and progression (9).

Single-cell sequencing techniques allow the complete clonal evolutionary structure of tumor cells to be reconstructed, thus providing insights into the evolution of lymphoma and revealing synergistic combinations that promote clonal expansion and dominant mutations, which offers information for therapeutic decisions (Fig. 3). For example, according to 2017 World Health Organization criteria, lymphomas with mutations in MYC and BCL2 or BCL6 during clonal evolution, as mentioned above, and with concurrent DLBCL histological features are no longer classified as DLBCL, but are referred to as advanced B cell lymphomas with MYC and BCL2 or BCL6 rearrangements (41). These sub-entities tend to have a worse prognosis for common DLBCL and often require the application of different treatments (7880).

To further dissect the heterogeneity of the origin of lymphomas, the studies on the origin of lymphoma at the cellular level were also summarized, as detailed below (Single-cell multi-omics for modeling B-cell GCs).

Single-cell multi-omics to explore the mechanism of drug resistance in lymphoma

Drug resistance in lymphoma can be broadly divided into the development of resistance at genetic and transcriptional levels within cancer cells and the development of resistance at tissue and cellular levels. According to the widely accepted theory of clonal evolution, the pre-emptive generation of heterogeneous drug-resistant clonal mutations through therapeutic selection is the most critical biological process (81). The ability to detect populations of cells that survive under a period of anticancer treatment is essential for drug resistance prediction or even reversal. It is difficult for conventional genetic studies of bulk samples to identify small numbers of drug-resistant cells. Today, through gene expression analysis under single-cell resolution, these cells and their mechanisms of drug resistance can be characterized (82).

Changes in gene levels are responsible for drug resistance in certain cancers, including lymphoma. Transcriptome sequencing analysis targeting the BTK gene [encodes a signaling protein vital for B cell development, differentiation and signaling (83)] found that after the inhibition of BTK, the FC receptors (FCGR2A, FCGR2B and FCGR3A) on the surface of B cells exhibited a higher level of expression (84), and the FC receptors promoted the internalization of the CD20 monoclonal antibody rituximab [core drug against malignant B lymphocyte proliferation (85)], thus reducing its clinical efficacy (86). In addition, through mutation or deletion, TP53, ATM, CDKN2A, KMT2D, and other key lymphoma suppressor genes can be inactivated, leading to genomic instability and promoting secondary mutations and drug resistance (87).

At the cellular level, lymphoma resistance in the macroscopic sense is mostly manifested by the presence of inherent molecular mechanisms of genotoxic drug resistance (88), hypoxia (89) and DNA damage (90). Previous studies have used single-cell transcriptomics techniques to deeply explore the mechanisms of acquired drug resistance and the differential alterations in the lymphoma microenvironment. Early experimental manipulations of whole-genome sequencing and DNA microarray of murine lymphoma have been performed to analyze CpG methylation, mRNA expression and DNA sequences that are related to increased drug resistance (91). Another transcriptional analysis revealed that lymphomas with low GAPDH expression predominantly adopt bovine/phosphorus metabolism and are dependent on mTORC1 signaling and glutaminyl solubilization to generate ATP (92), which indicates a large metabolic reprogramming response and reflects the acquisition mechanism of drug resistance (93). In another pan-cancer analysis of lymphoma, the results revealed differences in mRNA expression using data from The Cancer Genome Atlas and concluded that targeting the high expression of TRPV channel-associated genes can enhance drug resistance, and they may be promising biomarkers of patient prognosis (94). In addition to lymphoma, there is a single-cell muti-omics study showing that the levels of MSH2, MSH6 and MLH1, together with homologous recombination effectors such as BRCA2 and RAD51, are reduced in rectal cancer, suggesting that just like single-cell organisms, tumor cells can also promote drug-resistant persistent cells through continuous mutation derivation (95).

Although lymphoma has multiple mechanisms of drug resistance development, targeted single-cell studies have been used for molecular sequencing and outcome assessment of drug resistance in lymphoma. The single-cell multi-omics techniques not only explore and detect, but also show therapeutic potential to drug resistance in lymphoma. According to a single-cell analysis, KP772 can induce apoptosis of BCL-2-independent cells and upregulate the Harakri gene, making KP772 a promising candidate for anti-multidrug resistance of malignant lymphoma (96). Meanwhile, splicing modulators have been clinically tested for the treatment of drug resistance in lymphoma given their ability to interfere with drug metabolism or absorption of gene expression such as FPGS, dCK and SLC29A1 (97).

In addition to changes at the genetic and cellular levels, the mechanisms by which lymphomas develop drug resistance are complex and also include changes in the TME.

Single-cell multi-omics to profile the lymphoma microenvironment

The heterogeneity of lymphoma cells includes malignant cells and the TME (98). Although the relationship between cellular subpopulations and the TME remains not fully understood, it is most likely associated with lymphoma drug resistance and recurrence (99). As a result, research has focused on targeting the lymphoma microenvironment to achieve research breakthroughs.

Transcriptomic analyses from multiple independent lymphoma cohorts were performed to describe four microenvironment subtypes based on clinical behaviors and biological aberrations, and this approach identified the ECM proteins DCN and BGN as novel potential therapeutic targets for the TME (100). In another assessment of gene expression profiles, it was revealed that SPARC expression in the TME has fair inter-observer reproducibility and is a strong prognostic reference (101). In terms of treatment, the increased expression of CD8+ co-receptors (PTRPC and FYB) was found to enhance LCK and FYN expression, thereby activating the signaling of T cells and greatly contributing to immunocidal effects (84). A comprehensive transcriptomic atlas from over 100,000 lymphoma non-hematopoietic cells (NHCs; mesenchymal stromal and endothelial cells) at single-cell resolution demonstrated that NHC heterogeneity in LNs can be detected even in aggressive lymphomas, indicating the powerful practicability of single-cell analysis of the NHC profile in characterizing various TME subtypes (102).

It is well established that angiogenesis contributes to the inevitable progression of lymphoma through signaling transducers, cytokines and other components in the TME (103,104). Studies have shown that STAT3 is overexpressed in primary central nervous system lymphoma tissues vs. normal brain cells and vessels (105), and chromatin modifications occur when PCNSL forms (106). Single-cell histological analysis have shown that ROBO1, KAT2B and KMT2D regulate the lymphoma microenvironment by disrupting the functions of keratinocytes and stromal cells via non-histone acetylation (107), including histone deacetylase and DNA methyltransferase inhibitors are under investigation, suggesting they may be a new therapeutic option for lymphoma (108). Lysine acetyltransferase inhibitors enable the activation of KAT2B transcription to affect lymphoma angiogenesis (109). Recently, the study of gene impact associated with epigenetic modifications and angiogenetic events to fully profile the lymphoma microenvironment has gained research momentum.

The TME is also closely related to immune escape and drug resistance development in lymphoma, and studies at single-cell resolution reveal several novel mechanisms, suggesting that the microenvironment can suppress the host's antitumor immune activity (110). The immune escape of tumors is also a topic of research and an approach of clinical care.

Taken together, single-cell multi-omics techniques allow for the comprehensive characterization of the lymphoma microenvironment and the application of the gained information to a wider range of research.

Single-cell multi-omics to model B-cell GCs

GCs formed in secondary lymphatic organs undertake the binding of antigens to mature primitive B cells. GC B cells can produce numerous types of non-Hodgkin's lymphoma, such as DLBCL and Burkitt's lymphoma, which develop through different pathogenic mechanisms due to their origin in different stages of GC B cells (111). Transcriptional profiling revealed that DZ and LZ cells express RNA and the surface proteins CXCR4 and CD83, respectively, reflecting a heterogeneity that distinguishes them from other types of cells (112).

Single-cell multi-omics analysis revealed that reduced TNFRSF14 expression and STAT6 activation improves the inhibitory effect on GC B-cell signaling (113,114). The results showed that the epigenetic modifiers EZH2 and CREBBP can directly impact on GC B-cell's function. For example, upon CREBBP inactivation, MHC-II and CD40 are suppressed, which disturbs the presentation of antigens from lymphoma cells to CD4+ T cells (115).

Utilizing single-cell multi-omics techniques to model GCs can improved the characterization of the origin of lymphoma and provide more constructive insight into lymphomagenesis.

Single-cell multi-omics contribute to lymphoma treatment

The extensive heterogeneity of lymphoma is a major obstacle to the clinical management and the pharmacological treatment of this disease. As aforementioned in the sections pertaining to the development of novel biomarkers and the investigation of drug resistance and immune mechanisms in lymphoma, single-cell technology is rapidly evolving and may be of great value in the diagnosis and treatment of lymphoma. For example, scRNA-seq can identify several suitable entities for targeted therapy. Combined with capture technology, scRNA-seq can identify transcriptomic profiles related to the efficacy and toxicity of certain drugs, which has also been used to study the biomarkers of toxicity (116). Similarly, another study used scRNA-seq data to identify CD19 expression in brain wall cells and reveal the mechanism of neurotoxicity in CD19-targeted therapy (117). And it is promising that combining single-cell multi-omics techniques would be beneficial in the diagnosis of lymphoma, especially if the pathology is poorly characterized, as scRNA-seq identified immune cell biomarkers and determined essential biomarkers such as CD31, CD84 and CD226, which were hugely over-regulated and are promising to be the diagnostic biomarkers for lymphoma (118).

At the same time, numerous new assays based on single-cell sequencing are emerging, and investigators worldwide are working toward an easier and a more accurate application of single-cell technologies for lymphoma detection, which may offer constructive options for lymphoma treatment. For example, transient transfection of short barcode oligonucleotides allows the estimation of drug targets by transcriptional patterns of multiple scRNA-sequences to assess the efficacy and cytotoxicity of different drugs (119). Currently, a study is underway on the identification of targeted neoantigens by applying scRNA-seq combined with TCR sequencing for single-cell genomics (120). In the sequencing of paired single-cell RNA and TCR in patient tumors before and after PD-1 immunotherapy, the researchers identified the co-expression of biomarkers of chronic T cell apoptosis (121). The profiles of various T cells and TCR populations from normal adjacent tissues of tumors and peripheral blood was investigated by in-depth scRNA-seq and TCR sequencing from common types of tumors (122). RNA velocity can predict the future changes of each cell on a timescale of hours through the direct estimation of unsprayed and spliced mRNA from common scRNA-seq protocols, and it will greatly assist in the analysis of developmental lineages and cell dynamics in the future, which has been shown experimentally to be also applicable to human lymphoma (123). Mostly natural sequencing-by-synthesis for scRNA-seq is a novel approach applicable to the Ultima genomics platform, which has been benchmarked against scRNA-seq technologies and has a broader application potential (124).

In addition to the aforementioned novel single-cell technologies that have been applied to lymphoma, numerous other technologies in the field of oncology may have application potential in lymphoma, including CITE-seq, which enables simultaneous comparison of protein abundance and mRNA expression in specimens (34). In addition, SUPeR-seq synchronously detects linear and circ-rRNA expression levels in the tumor cell and the other control cell (125), whereas G&T-seq describes the whole genome and transcriptome of any sample cell (126). scTrio-seq simultaneously profiles the genomic, transcriptomic and epigenomic status of a single tumor cell (127), whereas INS-seq, a synthesis technique that enables large-scale parallel collection of scRNA-seq data and detection of intracellular protein activity, holds promise in exploring new immune subpopulations by analyzing different intracellular immune signaling signatures as well as metabolic activity and transcription factor binding (128). Currently, SCT provides a favorable platform for cancer diagnosis through the development of specific tumor biomarkers and individualized tumor therapy, and this technology will revolutionize the prognosis and treatment of all types of cancer on a global scale (129,130). MULTI-seq involves the multiplexed analysis of scRNA-sequences and mononuclear RNA sequences via lipid-labelled indexing, which can barcode all cells and nuclei worldwide through accessible plasma membranes. This approach is currently being used in triple-negative breast cancer (131).

Future prospects

The future of single-cell technology is unlimited, as evidenced by the increasing number of new techniques and improvements that are emerging (132). For example, RAGE-seq reconstructs highly diverse sequences (49). To advance lymphoma research and address lymphoma heterogeneity, additional integrated single-cell multi-omics technologies are essential to understand the cellular (tumor, stromal and immune) information about transcriptomes, proteomes and epigenomes, and even predict the state of each cell via pseudo-time detection of abundant patient data. In the near future, it may be possible to routinely analyze millions of cells; as a case in point, a pilot study to characterize a human cell atlas with 35 trillion human cells has reportedly begun (133).

Through multiple fusion approaches and technological innovations, single-cell transcriptomics techniques can assess how individual genetic drivers differently contribute to the fitness of lymphoma cells and investigate the mechanisms of cellular mutational symbiosis, multiple lesion interactions and clonal evolution, as revealed by spatial transcriptomics and spatial proteomics (134). Single-cell spatial transcription technologies have confirmed the upregulated expression of the major ligands CCR4, CCL17 and CCL22, and the downregulated expression of certain biomarkers in NK cells and CTL cells (135). Single-cell spatial analysis was applied to the DZ/LZ region of GCs in the tested lymph nodes, showing that MHC I/II expression and EZH2 mutation frequency were different within this microenvironment, which could assist in the diagnosis of GC lymphoma (136). And to reveal deeper analyses of tumor-environment interactions, the spatial analysis identified CXCR3 as biomarkers suggesting immune desert region, and also indicated therapeutic targets such as CCR4 and TIM-3, which are associated with combination treatment assays of lymphoma cellular therapies (137). Although knowledge of lymphoma spatial histology is currently in a state of infancy, there already exists a glimpse of the great potential of the combined application of single-cell multi-omics. Convergence and cross-analysis of multi-omics on top of different histologic assays may be a more important direction of development, and it is expected that multi-omics research will change the treatment paradigm of lymphoma in the future, and improve guiding the choice of clinical treatments. With the help of single-cell technology, this revolution is expanding throughout the field of immunology, not just lymphoma (34,138).

Feedback on the developed single-cell multi-omics platforms will be provided clinically through advanced cancer risk stratification, which is likely to deliver accurate lymphoma prognosis and detailed individual treatment.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Jilin (grant no. YDZJ202201ZYTS117).

Availability of data and materials

Not applicable.

Authors' contributions

CJ and DZ authored or reviewed drafts of the paper, and approved the final draft. JL provided figures and helped with proofreading of draft. LB and LL prepared tables, and approved the final draft. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ysebaert L, Quillet-Mary A, Tosolini M, Pont F, Laurent C and Fournié JJ: Lymphoma heterogeneity unraveled by single-cell transcriptomics. Front Immunol. 12:5976512021. View Article : Google Scholar : PubMed/NCBI

2 

Bocci F, Gearhart-Serna L, Boareto M, Ribeiro M, Ben-Jacob E, Devi GR, Levine H, Onuchic JN and Jolly MK: Toward understanding cancer stem cell heterogeneity in the tumor microenvironment. Proc Natl Acad Sci USA. 116:148–157. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Lei Y, Tang R, Xu J, Wang W, Zhang B, Liu J, Yu X and Shi S: Applications of single-cell sequencing in cancer research: Progress and perspectives. J Hematol Oncol. 14:912021. View Article : Google Scholar : PubMed/NCBI

4 

Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X, et al: Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 403:503–511. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M and Alizadeh AA: Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 12:453–457. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Gentles AJ, Newman AM, Liu CL, Bratman SV, Feng W, Kim D, Nair VS, Xu Y, Khuong A, Hoang CD, et al: The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med. 21:938–945. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Kothalawala WJ, Barták BK, Nagy ZB, Zsigrai S, Szigeti KA, Valcz G, Takács I, Kalmár A and Molnár B: A detailed overview about the single-cell analyses of solid tumors focusing on colorectal cancer. Pathol Oncol Res. 28:16103422022. View Article : Google Scholar : PubMed/NCBI

9 

Bingham GC, Lee F, Naba A and Barker TH: Spatial-omics: Novel approaches to probe cell heterogeneity and extracellular matrix biology. Matrix Biol. 91–92. 152–166. 2020.

10 

Borcherding N, Voigt AP, Liu V, Link BK, Zhang W and Jabbari A: Single-Cell profiling of cutaneous T-Cell lymphoma reveals underlying heterogeneity associated with disease progression. Clin Cancer Res. 25:2996–3005. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Gaydosik AM, Tabib T, Geskin LJ, Bayan CA, Conway JF, Lafyatis R and Fuschiotti P: Single-Cell lymphocyte heterogeneity in advanced cutaneous T-cell lymphoma skin tumors. Clin Cancer Res. 25:4443–4454. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Zhang W, Yang B, Weng L, Li J, Bai J, Wang T, Wang J, Ye J, Jing H, Jiao Y, et al: Single cell sequencing reveals cell populations that predict primary resistance to imatinib in chronic myeloid leukemia. Aging (Albany NY). 12:25337–25355. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Ren J, Qu R, Rahman NT, Lewis JM, King ALO, Liao X, Mirza FN, Carlson KR, Huang Y, Gigante S, et al: Integrated transcriptome and trajectory analysis of cutaneous T-cell lymphoma identifies putative precancer populations. Blood Adv. 7:445–457. 2023. View Article : Google Scholar : PubMed/NCBI

14 

Yamagishi M, Kubokawa M, Kuze Y, Suzuki A, Yokomizo A, Kobayashi S, Nakashima M, Makiyama J, Iwanaga M, Fukuda T, et al: Chronological genome and single-cell transcriptome integration characterizes the evolutionary process of adult T cell leukemia-lymphoma. Nat Commun. 12:48212021. View Article : Google Scholar : PubMed/NCBI

15 

Haebe S, Shree T, Sathe A, Day G, Czerwinski DK, Grimes SM, Lee H, Binkley MS, Long SR, Martin B, et al: Single-cell analysis can define distinct evolution of tumor sites in follicular lymphoma. Blood. 137:2869–2880. 2021. View Article : Google Scholar : PubMed/NCBI

16 

Borcherding N, Severson KJ, Henderson N, Ortolan LS, Rosenthal AC, Bellizzi AM, Liu V, Link BK, Mangold AR and Jabbari A: Single-cell analysis of Sézary syndrome reveals novel markers and shifting gene profiles associated with treatment. Blood Adv. 7:321–335. 2023. View Article : Google Scholar : PubMed/NCBI

17 

Valentin Hansen S, Høy Hansen M, Cédile O, Møller MB, Haaber J, Abildgaard N and Guldborg Nyvold C: Detailed characterization of the transcriptome of single B cells in mantle cell lymphoma suggesting a potential use for SOX4. Sci Rep. 11:190922021. View Article : Google Scholar : PubMed/NCBI

18 

Pritchett JC, Yang ZZ, Kim HJ, Villasboas JC, Tang X, Jalali S, Cerhan JR, Feldman AL and Ansell SM: High-dimensional and single-cell transcriptome analysis of the tumor microenvironment in angioimmunoblastic T cell lymphoma (AITL). Leukemia. 36:165–176. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Wei B, Liu Z, Fan Y, Wang S, Dong C, Rao W, Yang F, Cheng G and Zhang J: Analysis of cellular heterogeneity in immune microenvironment of primary central nervous system lymphoma by single-cell sequencing. Front Oncol. 11:6830072021. View Article : Google Scholar : PubMed/NCBI

20 

Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, et al: Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 161:1202–1214. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L, Weitz DA and Kirschner MW: Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell. 161:1187–1201. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Zheng GX, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, Ziraldo SB, Wheeler TD, McDermott GP, Zhu J, et al: Massively parallel digital transcriptional profiling of single cells. Nat Commun. 8:140492017. View Article : Google Scholar : PubMed/NCBI

23 

Gong H, Do D and Ramakrishnan R: Single-Cell mRNA-Seq using the fluidigm C1 system and integrated fluidics circuits. Methods Mol Biol. 1783:193–207. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Han X, Chen H, Huang D, Chen H, Fei L, Cheng C, Huang H, Yuan GC and Guo G: Mapping human pluripotent stem cell differentiation pathways using high throughput single-cell RNA-sequencing. Genome Biol. 19:472018. View Article : Google Scholar : PubMed/NCBI

25 

Ziegenhain C, Vieth B, Parekh S, Reinius B, Guillaumet-Adkins A, Smets M, Leonhardt H, Heyn H, Hellmann I and Enard W: Comparative analysis of single-cell RNA sequencing methods. Mol Cell. 65:631–643.e4. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Xin Y, Kim J, Ni M, Wei Y, Okamoto H, Lee J, Adler C, Cavino K, Murphy AJ, Yancopoulos GD, et al: Use of the Fluidigm C1 platform for RNA sequencing of single mouse pancreatic islet cells. Proc Natl Acad Sci USA. 113:3293–3298. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Gierahn TM, Wadsworth MH II, Hughes TK, Bryson BD, Butler A, Satija R, Fortune S, Love JC and Shalek AK: Seq-Well: Portable, low-cost RNA sequencing of single cells at high throughput. Nat Methods. 14:395–398. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Aicher TP, Carroll S, Raddi G, Gierahn T, Wadsworth MH II, Hughes TK, Love C and Shalek AK: Seq-Well: A sample-efficient, portable picowell platform for massively parallel single-cell RNA sequencing. Methods Mol Biol. 1979:111–132. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Han X, Wang R, Zhou Y, Fei L, Sun H, Lai S, Saadatpour A, Zhou Z, Chen H, Ye F, et al: Mapping the mouse cell atlas by microwell-seq. Cell. 173:13072018. View Article : Google Scholar : PubMed/NCBI

30 

Lai S, Huang W, Xu Y, Jiang M, Chen H, Cheng C, Lu Y, Huang H, Guo G and Han X: Comparative transcriptomic analysis of hematopoietic system between human and mouse by Microwell-seq. Cell Discov. 4:342018. View Article : Google Scholar : PubMed/NCBI

31 

Briggs JA, Weinreb C, Wagner DE, Megason S, Peshkin L, Kirschner MW and Klein AM: The dynamics of gene expression in vertebrate embryogenesis at single-cell resolution. Science. 360:eaar57802018. View Article : Google Scholar : PubMed/NCBI

32 

Tosches MA, Yamawaki TM, Naumann RK, Jacobi AA, Tushev G and Laurent G: Evolution of pallium, hippocampus, and cortical cell types revealed by single-cell transcriptomics in reptiles. Science. 360:881–888. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Jaitin DA, Kenigsberg E, Keren-Shaul H, Elefant N, Paul F, Zaretsky I, Mildner A, Cohen N, Jung S, Tanay A and Amit I: Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science. 343:776–779. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H, Satija R and Smibert P: Simultaneous epitope and transcriptome measurement in single cells. Nat Methods. 14:865–868. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Mimitou EP, Cheng A, Montalbano A, Hao S, Stoeckius M, Legut M, Roush T, Herrera A, Papalexi E, Ouyang Z, et al: Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat Methods. 16:409–412. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Harris NL, Jaffe ES, Stein H, Banks PM, Chan JK, Cleary ML, Delsol G, De Wolf-Peeters C, Falini B, Gatter KC, et al: A revised European-American classification of lymphoid neoplasms: A proposal from the International Lymphoma Study Group. Blood. 84:1361–1392. 1994. View Article : Google Scholar : PubMed/NCBI

37 

Barbui T, Thiele J, Gisslinger H, Kvasnicka HM, Vannucchi AM, Guglielmelli P, Orazi A and Tefferi A: The 2016 WHO classification and diagnostic criteria for myeloproliferative neoplasms: Document summary and in-depth discussion. Blood Cancer J. 8:152018. View Article : Google Scholar : PubMed/NCBI

38 

Glaser SL and Jarrett RF: The epidemiology of Hodgkin's disease. Baillieres Clin Haematol. 9:401–416. 1996. View Article : Google Scholar : PubMed/NCBI

39 

Marafioti T, Hummel M, Foss HD, Laumen H, Korbjuhn P, Anagnostopoulos I, Lammert H, Demel G, Theil J, Wirth T and Stein H: Hodgkin and reed-sternberg cells represent an expansion of a single clone originating from a germinal center B-cell with functional immunoglobulin gene rearrangements but defective immunoglobulin transcription. Blood. 95:1443–1450. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Kanzler H, Küppers R, Hansmann ML and Rajewsky K: Hodgkin and Reed-Sternberg cells in Hodgkin's disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells. J Exp Med. 184:1495–1505. 1996. View Article : Google Scholar : PubMed/NCBI

41 

Grimm KE and O'Malley DP: Aggressive B cell lymphomas in the 2017 revised WHO classification of tumors of hematopoietic and lymphoid tissues. Ann Diagn Pathol. 38:6–10. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Thandra KC, Barsouk A, Saginala K, Padala SA, Barsouk A and Rawla P: Epidemiology of Non-Hodgkin's Lymphoma. Med Sci (Basel). 9:52021.PubMed/NCBI

43 

de Leval L and Jaffe ES: Lymphoma Classification. Cancer J. 26:176–185. 2020. View Article : Google Scholar : PubMed/NCBI

44 

García-Sanz R and Jiménez C: Time to move to the single-cell level: Applications of single-cell multi-omics to hematological malignancies and Waldenström's Macroglobulinemia-A particularly heterogeneous lymphoma. Cancers (Basel). 13:15412021. View Article : Google Scholar : PubMed/NCBI

45 

Glass DR, Tsai AG, Oliveria JP, Hartmann FJ, Kimmey SC, Calderon AA, Borges L, Glass MC, Wagar LE, Davis MM and Bendall SC: An Integrated Multi-omic Single-cell atlas of human B cell identity. Immunity. 53:217–232.e5. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Crinier A, Dumas PY, Escalière B, Piperoglou C, Gil L, Villacreces A, Vély F, Ivanovic Z, Milpied P, Narni-Mancinelli É and Vivier É: Single-cell profiling reveals the trajectories of natural killer cell differentiation in bone marrow and a stress signature induced by acute myeloid leukemia. Cell Mol Immunol. 18:1290–1304. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Aoki T, Chong LC, Takata K, Milne K, Hav M, Colombo A, Chavez EA, Nissen M, Wang X, Miyata-Takata T, et al: Single-Cell transcriptome analysis reveals disease-defining T-cell subsets in the tumor microenvironment of classic Hodgkin Lymphoma. Cancer Discov. 10:406–421. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Pizzolato G, Kaminski H, Tosolini M, Franchini DM, Pont F, Martins F, Valle C, Labourdette D, Cadot S, Quillet-Mary A, et al: Single-cell RNA sequencing unveils the shared and the distinct cytotoxic hallmarks of human TCRVδ1 and TCRVδ2 γδ T lymphocytes. Proc Natl Acad Sci USA. 116:11906–11915. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Singh M, Al-Eryani G, Carswell S, Ferguson JM, Blackburn J, Barton K, Roden D, Luciani F, Giang Phan T, Junankar S, et al: High-throughput targeted long-read single cell sequencing reveals the clonal and transcriptional landscape of lymphocytes. Nat Commun. 10:31202019. View Article : Google Scholar : PubMed/NCBI

50 

Vitak SA, Torkenczy KA, Rosenkrantz JL, Fields AJ, Christiansen L, Wong MH, Carbone L, Steemers FJ and Adey A: Sequencing thousands of single-cell genomes with combinatorial indexing. Nat Methods. 14:302–328. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Xiong J, Cui BW, Wang N, Dai YT, Zhang H, Wang CF, Zhong HJ, Cheng S, Ou-Yang BS, Hu Y, et al: Genomic and transcriptomic characterization of natural killer T cell lymphoma. Cancer Cell. 37:403–419.e6. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Li P, Chai J, Chen Z, Liu Y, Wei J, Liu Y, Zhao D, Ma J, Wang K, Li X, et al: Genomic mutation profile of primary gastrointestinal diffuse large B-Cell Lymphoma. Front Oncol. 11:6226482021. View Article : Google Scholar : PubMed/NCBI

53 

Radke J, Ishaque N, Koll R, Gu Z, Schumann E, Sieverling L, Uhrig S, Hübschmann D, Toprak UH, López C, et al: The genomic and transcriptional landscape of primary central nervous system lymphoma. Nat Commun. 13:25582022. View Article : Google Scholar : PubMed/NCBI

54 

Yi S, Yan Y, Jin M, Bhattacharya S, Wang Y, Wu Y, Yang L, Gine E, Clot G, Chen L, et al: Genomic and transcriptomic profiling reveals distinct molecular subsets associated with outcomes in mantle cell lymphoma. J Clin Invest. 132:e1532832022. View Article : Google Scholar : PubMed/NCBI

55 

Nadeu F, Martin-Garcia D, Clot G, Díaz-Navarro A, Duran-Ferrer M, Navarro A, Vilarrasa-Blasi R, Kulis M, Royo R, Gutiérrez-Abril J, et al: Genomic and epigenomic insights into the origin, pathogenesis, and clinical behavior of mantle cell lymphoma subtypes. Blood. 136:1419–1432. 2020. View Article : Google Scholar : PubMed/NCBI

56 

De Bie J, Demeyer S, Alberti-Servera L, Geerdens E, Segers H, Broux M, De Keersmaecker K, Michaux L, Vandenberghe P, Voet T, et al: Single-cell sequencing reveals the origin and the order of mutation acquisition in T-cell acute lymphoblastic leukemia. Leukemia. 32:1358–1369. 2018. View Article : Google Scholar : PubMed/NCBI

57 

López C, Kleinheinz K, Aukema SM, Rohde M, Bernhart SH, Hübschmann D, Wagener R, Toprak UH, Raimondi F, Kreuz M, et al: Genomic and transcriptomic changes complement each other in the pathogenesis of sporadic Burkitt lymphoma. Nat Commun. 10:14592019. View Article : Google Scholar : PubMed/NCBI

58 

Küçük C, Hu X, Gong Q, Jiang B, Cornish A, Gaulard P, McKeithan T and Chan WC: Diagnostic and biological significance of KIR EXPRESSION PROFILE DETErmined by RNA-Seq in Natural Killer/T-Cell Lymphoma. Am J Pathol. 186:1435–1441. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Andor N, Simonds EF, Czerwinski DK, Chen J, Grimes SM, Wood-Bouwens C, Zheng GXY, Kubit MA, Greer S, Weiss WA, et al: Single-cell RNA-Seq of follicular lymphoma reveals malignant B-cell types and coexpression of T-cell immune checkpoints. Blood. 133:1119–1129. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Huang Z, Ma L, Huang C, Li Q and Nice EC: Proteomic profiling of human plasma for cancer biomarker discovery. Proteomics. 17((6))2017.

61 

Kim MK, Song JY, Koh DI, Kim JY, Hatano M, Jeon BN, Kim MY, Cho SY, Kim KS and Hur MW: Reciprocal negative regulation between the tumor suppressor protein p53 and B cell CLL/lymphoma 6 (BCL6) via control of caspase-1 expression. J Biol Chem. 294:299–313. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Daniunaite K, Jarmalaite S and Kriukiene E: Epigenomic technologies for deciphering circulating tumor DNA. Curr Opin Biotechnol. 55:23–29. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Ng SB, Yan J, Huang G, Selvarajan V, Tay JL, Lin B, Bi C, Tan J, Kwong YL, Shimizu N, et al: Dysregulated microRNAs affect pathways and targets of biologic relevance in nasal-type natural killer/T-cell lymphoma. Blood. 118:4919–4929. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Zhang X, Ji W, Huang R, Li L, Wang X, Li L, Fu X, Sun Z, Li Z, Chen Q and Zhang M: MicroRNA-155 is a potential molecular marker of natural killer/T-cell lymphoma. Oncotarget. 7:53808–53819. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Yan J, Ng SB, Tay JL, Lin B, Koh TL, Tan J, Selvarajan V, Liu SC, Bi C, Wang S, et al: EZH2 overexpression in natural killer/T-cell lymphoma confers growth advantage independently of histone methyltransferase activity. Blood. 121:4512–4520. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Liang L, Nong L, Zhang S, Zhao J, Ti H, Dong Y, Zhang B and Li T: The downregulation of PRDM1/Blimp-1 is associated with aberrant expression of miR-223 in extranodal NK/T-cell lymphoma, nasal type. J Exp Clin Cancer Res. 33:72014. View Article : Google Scholar : PubMed/NCBI

67 

Küçük C, Hu X, Jiang B, Klinkebiel D, Geng H, Gong Q, Bouska A, Iqbal J, Gaulard P, McKeithan TW and Chan WC: Global promoter methylation analysis reveals novel candidate tumor suppressor genes in natural killer cell lymphoma. Clin Cancer Res. 21:1699–1711. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Chen YW, Guo T, Shen L, Wong KY, Tao Q, Choi WW, Au-Yeung RK, Chan YP, Wong ML, Tang JC, et al: Receptor-type tyrosine-protein phosphatase κ directly targets STAT3 activation for tumor suppression in nasal NK/T-cell lymphoma. Blood. 125:1589–1600. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Ranzoni AM, Tangherloni A, Berest I, Riva SG, Myers B, Strzelecka PM, Xu J, Panada E, Mohorianu I, Zaugg JB and Cvejic A: Integrative Single-Cell RNA-Seq and ATAC-seq analysis of human developmental hematopoiesis. Cell Stem Cell. 28:472–487.e7. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Yu J, Gemenetzis G, Kinny-Köster B, Habib JR, Groot VP, Teinor J, Yin L, Pu N, Hasanain A, van Oosten F, et al: Pancreatic circulating tumor cell detection by targeted single-cell next-generation sequencing. Cancer Lett. 493:245–253. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Zhang L, Dong X, Lee M, Maslov AY, Wang T and Vijg J: Single-cell whole-genome sequencing reveals the functional landscape of somatic mutations in B lymphocytes across the human lifespan. Proc Natl Acad Sci USA. 116:9014–9019. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Psatha K, Kollipara L, Voutyraki C, Divanach P, Sickmann A, Rassidakis GZ, Drakos E and Aivaliotis M: Deciphering lymphoma pathogenesis via state-of-the-art mass spectrometry-based quantitative proteomics. J Chromatogr B Analyt Technol Biomed Life Sci. 1047:2–14. 2017. View Article : Google Scholar : PubMed/NCBI

73 

Bacher U, Haferlach T, Alpermann T, Kern W, Schnittger S and Haferlach C: Several lymphoma-specific genetic events in parallel can be found in mature B-cell neoplasms. Genes Chromosomes Cancer. 50:43–50. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Okosun J, Bödör C, Wang J, Araf S, Yang CY, Pan C, Boller S, Cittaro D, Bozek M, Iqbal S, et al: Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet. 46:176–181. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Sewastianik T, Prochorec-Sobieszek M, Chapuy B and Juszczyński P: MYC deregulation in lymphoid tumors: Molecular mechanisms, clinical consequences and therapeutic implications. Biochim Biophys Acta. 1846:457–467. 2014.PubMed/NCBI

76 

Rosenthal A and Rimsza L: Genomics of aggressive B-cell lymphoma. Hematology Am Soc Hematol Educ Program. 2018:69–74. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Gawad C, Koh W and Quake SR: Dissecting the clonal origins of childhood acute lymphoblastic leukemia by single-cell genomics. Proc Natl Acad Sci USA. 111:17947–17952. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Snuderl M, Kolman OK, Chen YB, Hsu JJ, Ackerman AM, Dal Cin P, Ferry JA, Harris NL, Hasserjian RP, Zukerberg LR, et al: B-cell lymphomas with concurrent IGH-BCL2 and MYC rearrangements are aggressive neoplasms with clinical and pathologic features distinct from Burkitt lymphoma and diffuse large B-cell lymphoma. Am J Surg Pathol. 34:327–340. 2010. View Article : Google Scholar : PubMed/NCBI

79 

Huang W, Medeiros LJ, Lin P, Wang W, Tang G, Khoury J, Konoplev S, Yin CC, Xu J, Oki Y and Li S: MYC/BCL2/BCL6 triple hit lymphoma: A study of 40 patients with a comparison to MYC/BCL2 and MYC/BCL6 double hit lymphomas. Mod Pathol. 31:1470–1478. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Moore EM, Aggarwal N, Surti U and Swerdlow SH: Further exploration of the complexities of large B-Cell Lymphomas With MYC abnormalities and the importance of a blastoid morphology. Am J Surg Pathol. 41:1155–1166. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Jiang Y, Redmond D, Nie K, Eng KW, Clozel T, Martin P, Tan LH, Melnick AM, Tam W and Elemento O: Deep sequencing reveals clonal evolution patterns and mutation events associated with relapse in B-cell lymphomas. Genome Biol. 15:4322014. View Article : Google Scholar : PubMed/NCBI

82 

Ding S, Chen X and Shen K: Single-cell RNA sequencing in breast cancer: Understanding tumor heterogeneity and paving roads to individualized therapy. Cancer Commun (Lond). 40:329–344. 2020. View Article : Google Scholar : PubMed/NCBI

83 

Kim E, Hurtz C, Koehrer S, Wang Z, Balasubramanian S, Chang BY, Müschen M, Davis RE and Burger JA: Ibrutinib inhibits pre-BCR(+) B-cell acute lymphoblastic leukemia progression by targeting BTK and BLK. Blood. 129:1155–1165. 2017. View Article : Google Scholar : PubMed/NCBI

84 

Wang L, Mo S, Li X, He Y and Yang J: Single-cell RNA-seq reveals the immune escape and drug resistance mechanisms of mantle cell lymphoma. Cancer Biol Med. 17:726–739. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Marcus R, Imrie K, Solal-Celigny P, Catalano JV, Dmoszynska A, Raposo JC, Offner FC, Gomez-Codina J, Belch A, Cunningham D, et al: Phase III study of R-CVP compared with cyclophosphamide, vincristine, and prednisone alone in patients with previously untreated advanced follicular lymphoma. J Clin Oncol. 26:4579–4586. 2008. View Article : Google Scholar : PubMed/NCBI

86 

Lim SH, Vaughan AT, Ashton-Key M, Williams EL, Dixon SV, Chan HT, Beers SA, French RR, Cox KL, Davies AJ, et al: Fc gamma receptor IIb on target B cells promotes rituximab internalization and reduces clinical efficacy. Blood. 118:2530–2540. 2011. View Article : Google Scholar : PubMed/NCBI

87 

Zhang J, Dominguez-Sola D, Hussein S, Lee JE, Holmes AB, Bansal M, Vlasevska S, Mo T, Tang H, Basso K, et al: Disruption of KMT2D perturbs germinal center B cell development and promotes lymphomagenesis. Nat Med. 21:1190–1198. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M and Vardiman JW: The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 127:2391–2405. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Swerdlow SH and Cook JR: As the world turns, evolving lymphoma classifications-past, present and future. Hum Pathol. 95:55–77. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Takagi M: DNA damage response and hematological malignancy. Int J Hematol. 106:345–356. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Flinders C, Lam L, Rubbi L, Ferrari R, Fitz-Gibbon S, Chen PY, Thompson M, Christofk H, B Agus D, Ruderman D, et al: Epigenetic changes mediated by polycomb repressive complex 2 and E2a are associated with drug resistance in a mouse model of lymphoma. Genome Med. 8:542016. View Article : Google Scholar : PubMed/NCBI

92 

Chiche J, Reverso-Meinietti J, Mouchotte A, Rubio-Patiño C, Mhaidly R, Villa E, Bossowski JP, Proics E, Grima-Reyes M, Paquet A, et al: GAPDH expression predicts the response to R-CHOP, the tumor metabolic status, and the response of DLBCL patients to metabolic inhibitors. Cell Metab. 29:1243–1257.e10. 2019. View Article : Google Scholar : PubMed/NCBI

93 

Klener P and Klanova M: Drug Resistance in Non-Hodgkin Lymphomas. Int J Mol Sci. 21:20812020. View Article : Google Scholar : PubMed/NCBI

94 

Wang X, Li G, Zhang Y, Li L, Qiu L, Qian Z, Zhou S, Wang X, Li Q and Zhang H: Pan-Cancer analysis reveals genomic and clinical characteristics of TRPV Channel-related genes. Front Oncol. 12:8131002022. View Article : Google Scholar : PubMed/NCBI

95 

Russo M, Crisafulli G, Sogari A, Reilly NM, Arena S, Lamba S, Bartolini A, Amodio V, Magrì A, Novara L, et al: Adaptive mutability of colorectal cancers in response to targeted therapies. Science. 366:1473–1480. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Kater L, Kater B, Jakupec MA, Keppler BK and Prokop A: KP772 overcomes multiple drug resistance in malignant lymphoma and leukemia cells in vitro by inducing Bcl-2-independent apoptosis and upregulation of Harakiri. J Biol Inorg Chem. 26:897–907. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Sciarrillo R, Wojtuszkiewicz A, Assaraf YG, Jansen G, Kaspers GJL, Giovannetti E and Cloos J: The role of alternative splicing in cancer: From oncogenesis to drug resistance. Drug Resist Updat. 53:1007282020. View Article : Google Scholar : PubMed/NCBI

98 

Roider T, Seufert J, Uvarovskii A, Frauhammer F, Bordas M, Abedpour N, Stolarczyk M, Mallm JP, Herbst SA, Bruch PM, et al: Dissecting intratumour heterogeneity of nodal B-cell lymphomas at the transcriptional, genetic and drug-response levels. Nat Cell Biol. 22:896–906. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Wills QF, Livak KJ, Tipping AJ, Enver T, Goldson AJ, Sexton DW and Holmes C: Single-cell gene expression analysis reveals genetic associations masked in whole-tissue experiments. Nat Biotechnol. 31:748–752. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Kotlov N, Bagaev A, Revuelta MV, Phillip JM, Cacciapuoti MT, Antysheva Z, Svekolkin V, Tikhonova E, Miheecheva N, Kuzkina N, et al: Clinical and Biological Subtypes of B-cell lymphoma revealed by microenvironmental signatures. Cancer Discov. 11:1468–1489. 2021. View Article : Google Scholar : PubMed/NCBI

101 

Croci GA, Au-Yeung RKH, Reinke S, Staiger AM, Koch K, Oschlies I, Richter J, Poeschel V, Held G, Loeffler M, et al: SPARC-positive macrophages are the superior prognostic factor in the microenvironment of diffuse large B-cell lymphoma and independent of MYC rearrangement and double-/triple-hit status. Ann Oncol. 32:1400–1409. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Abe Y, Sakata-Yanagimoto M, Fujisawa M, Miyoshi H, Suehara Y, Hattori K, Kusakabe M, Sakamoto T, Nishikii H, Nguyen TB, et al: A single-cell atlas of non-haematopoietic cells in human lymph nodes and lymphoma reveals a landscape of stromal remodelling. Nat Cell Biol. 24:565–578. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Ferreri AJ, Cwynarski K, Pulczynski E, Ponzoni M, Deckert M, Politi LS, Torri V, Fox CP, Rosée PL, Schorb E, et al: Chemoimmunotherapy with methotrexate, cytarabine, thiotepa, and rituximab (MATRix regimen) in patients with primary CNS lymphoma: Results of the first randomisation of the International Extranodal Lymphoma Study Group-32 (IELSG32) phase 2 trial. Lancet Haematol. 3:e217–e227. 2016. View Article : Google Scholar : PubMed/NCBI

104 

Bromberg JEC, Issa S, Bakunina K, Minnema MC, Seute T, Durian M, Cull G, Schouten HC, Stevens WBC, Zijlstra JM, et al: Rituximab in patients with primary CNS lymphoma (HOVON 105/ALLG NHL 24): A randomised, open-label, phase 3 intergroup study. Lancet Oncol. 20:216–228. 2019. View Article : Google Scholar : PubMed/NCBI

105 

Ruggieri S, Tamma R, Resta N, Albano F, Coccaro N, Loconte D, Annese T, Errede M, Specchia G, Senetta R, et al: Stat3-positive tumor cells contribute to vessels neoformation in primary central nervous system lymphoma. Oncotarget. 8:31254–31269. 2017. View Article : Google Scholar : PubMed/NCBI

106 

Zhou Y, Liu W, Xu Z, Zhu H, Xiao D, Su W, Zeng R, Feng Y, Duan Y, Zhou J and Zhong M: Analysis of genomic alteration in primary central nervous system lymphoma and the expression of some related genes. Neoplasia. 20:1059–1069. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Ribatti D, Nico B, Ranieri G, Specchia G and Vacca A: The role of angiogenesis in human non-Hodgkin lymphomas. Neoplasia. 15:231–238. 2013. View Article : Google Scholar : PubMed/NCBI

108 

Clozel T, Yang S, Elstrom RL, Tam W, Martin P, Kormaksson M, Banerjee S, Vasanthakumar A, Culjkovic B, Scott DW, et al: Mechanism-based epigenetic chemosensitization therapy of diffuse large B-cell lymphoma. Cancer Discov. 3:1002–1019. 2013. View Article : Google Scholar : PubMed/NCBI

109 

Hazar B, Paydas S, Zorludemir S, Sahin B and Tuncer I: Prognostic significance of microvessel density and vascular endothelial growth factor (VEGF) expression in non-Hodgkin's lymphoma. Leuk Lymphoma. 44:2089–2093. 2003. View Article : Google Scholar : PubMed/NCBI

110 

Carlo-Stella C and Santoro A: Microenvironment-related biomarkers and novel targets in classical Hodgkin's lymphoma. Biomark Med. 9:807–817. 2015. View Article : Google Scholar : PubMed/NCBI

111 

Holmes AB, Corinaldesi C, Shen Q, Kumar R, Compagno N, Wang Z, Nitzan M, Grunstein E, Pasqualucci L, Dalla-Favera R and Basso K: Single-cell analysis of germinal-center B cells informs on lymphoma cell of origin and outcome. J Exp Med. 217:e202004832020. View Article : Google Scholar : PubMed/NCBI

112 

Mintz MA and Cyster JG: T follicular helper cells in germinal center B cell selection and lymphomagenesis. Immunol Rev. 296:48–61. 2020. View Article : Google Scholar : PubMed/NCBI

113 

Turqueti-Neves A, Otte M, Prazeres da Costa O, Höpken UE, Lipp M, Buch T and Voehringer D: B-cell-intrinsic STAT6 signaling controls germinal center formation. Eur J Immunol. 44:2130–2138. 2014. View Article : Google Scholar : PubMed/NCBI

114 

Mintz MA, Felce JH, Chou MY, Mayya V, Xu Y, Shui JW, An J, Li Z, Marson A, Okada T, et al: The HVEM-BTLA Axis Restrains T cell help to germinal center B cells and functions as a cell-extrinsic suppressor in lymphomagenesis. Immunity. 51:310–323.e7. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Hashwah H, Schmid CA, Kasser S, Bertram K, Stelling A, Manz MG and Müller A: Inactivation of CREBBP expands the germinal center B cell compartment, down-regulates MHCII expression and promotes DLBCL growth. Proc Natl Acad Sci USA. 114:9701–9706. 2017. View Article : Google Scholar : PubMed/NCBI

116 

Deng Q, Han G, Puebla-Osorio N, Ma MCJ, Strati P, Chasen B, Dai E, Dang M, Jain N, Yang H, et al: Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat Med. 26:1878–1887. 2020. View Article : Google Scholar : PubMed/NCBI

117 

Parker KR, Migliorini D, Perkey E, Yost KE, Bhaduri A, Bagga P, Haris M, Wilson NE, Liu F, Gabunia K, et al: Single-Cell analyses identify brain mural cells expressing CD19 as potential off-tumor targets for CAR-T immunotherapies. Cell. 183:126–142.e17. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Shi Y, Ding W, Gu W, Shen Y, Li H, Zheng Z, Zheng X, Liu Y and Ling Y: Single-cell phenotypic profiling to identify a set of immune cell protein biomarkers for relapsed and refractory diffuse large B cell lymphoma: A single-center study. J Leukoc Biol. 112:1633–1648. 2022. View Article : Google Scholar : PubMed/NCBI

119 

Shin D, Lee W, Lee JH and Bang D: Multiplexed single-cell RNA-seq via transient barcoding for simultaneous expression profiling of various drug perturbations. Sci Adv. 5:eaav22492019. View Article : Google Scholar : PubMed/NCBI

120 

Krieg C, Nowicka M, Guglietta S, Schindler S, Hartmann FJ, Weber LM, Dummer R, Robinson MD, Levesque MP and Becher B: High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 24:144–153. 2018. View Article : Google Scholar : PubMed/NCBI

121 

Yost KE, Satpathy AT, Wells DK, Qi Y, Wang C, Kageyama R, McNamara KL, Granja JM, Sarin KY, Brown RA, et al: Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 25:1251–1259. 2019. View Article : Google Scholar : PubMed/NCBI

122 

Wu TD, Madireddi S, de Almeida PE, Banchereau R, Chen YJ, Chitre AS, Chiang EY, Iftikhar H, O'Gorman WE, Au-Yeung A, et al: Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature. 579:274–278. 2020. View Article : Google Scholar : PubMed/NCBI

123 

La Manno G, Soldatov R, Zeisel A, Braun E, Hochgerner H, Petukhov V, Lidschreiber K, Kastriti ME, Lönnerberg P, Furlan A, et al: RNA velocity of single cells. Nature. 560:494–498. 2018. View Article : Google Scholar : PubMed/NCBI

124 

Simmons SK, Lithwick-Yanai G, Adiconis X, Oberstrass F, Iremadze N, Geiger-Schuller K, Thakore PI, Frangieh CJ, Barad O, Almogy G, et al: Mostly natural sequencing-by-synthesis for scRNA-seq using Ultima sequencing. Nat Biotechnol. 41:204–211. 2023. View Article : Google Scholar : PubMed/NCBI

125 

Fan X, Zhang X, Wu X, Guo H, Hu Y, Tang F and Huang Y: Single-cell RNA-seq transcriptome analysis of linear and circular RNAs in mouse preimplantation embryos. Genome Biol. 16:1482015. View Article : Google Scholar : PubMed/NCBI

126 

Macaulay IC, Haerty W, Kumar P, Li YI, Hu TX, Teng MJ, Goolam M, Saurat N, Coupland P, Shirley LM, et al: G&T-seq: Parallel sequencing of single-cell genomes and transcriptomes. Nat Methods. 12:519–522. 2015. View Article : Google Scholar : PubMed/NCBI

127 

Hou Y, Guo H, Cao C, Li X, Hu B, Zhu P, Wu X, Wen L, Tang F, Huang Y and Peng J: Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res. 26:304–319. 2016. View Article : Google Scholar : PubMed/NCBI

128 

Katzenelenbogen Y, Sheban F, Yalin A, Yofe I, Svetlichnyy D, Jaitin DA, Bornstein C, Moshe A, Keren-Shaul H, Cohen M, et al: Coupled scRNA-Seq and intracellular protein activity reveal an immunosuppressive role of TREM2 in cancer. Cell. 182:872–885.e19. 2020. View Article : Google Scholar : PubMed/NCBI

129 

Svensson V, Vento-Tormo R and Teichmann SA: Exponential scaling of single-cell RNA-seq in the past decade. Nat Protoc. 13:599–604. 2018. View Article : Google Scholar : PubMed/NCBI

130 

Bai X, Li Y, Zeng X, Zhao Q and Zhang Z: Single-cell sequencing technology in tumor research. Clin Chim Acta. 518:101–109. 2021. View Article : Google Scholar : PubMed/NCBI

131 

McGinnis CS, Patterson DM, Winkler J, Conrad DN, Hein MY, Srivastava V, Hu JL, Murrow LM, Weissman JS, Werb Z, et al: MULTI-seq: Sample multiplexing for single-cell RNA sequencing using lipid-tagged indices. Nat Methods. 16:619–626. 2019. View Article : Google Scholar : PubMed/NCBI

132 

Kashima Y, Sakamoto Y, Kaneko K, Seki M, Suzuki Y and Suzuki A: Single-cell sequencing techniques from individual to multiomics analyses. Exp Mol Med. 52:1419–1427. 2020. View Article : Google Scholar : PubMed/NCBI

133 

Ando Y, Kwon AT and Shin JW: An era of single-cell genomics consortia. Exp Mol Med. 52:1409–1418. 2020. View Article : Google Scholar : PubMed/NCBI

134 

Landeira-Viñuela A, Díez P, Juanes-Velasco P, Lécrevisse Q, Orfao A, De Las Rivas J and Fuentes M: Deepening into intracellular signaling landscape through integrative spatial proteomics and transcriptomics in a lymphoma model. Biomolecules. 11:17762021. View Article : Google Scholar : PubMed/NCBI

135 

Du J, Qiu C, Li WS, Wang B, Han XL, Lin SW, Fu XH, Hou J and Huang ZF: Spatial transcriptomics analysis reveals that CCL17 and CCL22 are robust indicators of a suppressive immune environment in angioimmunoblastic T cell lymphoma (AITL). Front Biosci (Landmark Ed). 27:2702022. View Article : Google Scholar : PubMed/NCBI

136 

Tripodo C, Zanardi F, Iannelli F, Mazzara S, Vegliante M, Morello G, Di Napoli A, Mangogna A, Facchetti F, Sangaletti S, et al: A spatially resolved dark-versus light-zone microenvironment signature subdivides germinal center-related aggressive B cell lymphomas. iScience. 23:1015622020. View Article : Google Scholar : PubMed/NCBI

137 

Colombo AR, Hav M, Singh M, Xu A, Gamboa A, Lemos T, Gerdtsson E, Chen D, Houldsworth J, Shaknovich R, et al: Single-cell spatial analysis of tumor immune architecture in diffuse large B-cell lymphoma. Blood Adv. 6:4675–4690. 2022. View Article : Google Scholar : PubMed/NCBI

138 

Efremova M, Vento-Tormo R, Park JE, Teichmann SA and James KR: Immunology in the Era of single-cell technologies. Annu Rev Immunol. 38:727–757. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2023
Volume 50 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jin C, Zhou D, Li J, Bi L and Li L: Single‑cell multi‑omics advances in lymphoma research (Review). Oncol Rep 50: 184, 2023.
APA
Jin, C., Zhou, D., Li, J., Bi, L., & Li, L. (2023). Single‑cell multi‑omics advances in lymphoma research (Review). Oncology Reports, 50, 184. https://doi.org/10.3892/or.2023.8621
MLA
Jin, C., Zhou, D., Li, J., Bi, L., Li, L."Single‑cell multi‑omics advances in lymphoma research (Review)". Oncology Reports 50.4 (2023): 184.
Chicago
Jin, C., Zhou, D., Li, J., Bi, L., Li, L."Single‑cell multi‑omics advances in lymphoma research (Review)". Oncology Reports 50, no. 4 (2023): 184. https://doi.org/10.3892/or.2023.8621