Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jun 1.
Published in final edited form as: Nat Cell Biol. 2018 Nov 26;20(12):1349–1360. doi: 10.1038/s41556-018-0236-7

Tumour heterogeneity and metastasis at single-cell resolution

Devon A Lawson 1,2,*, Kai Kessenbrock 2,3,*, Ryan T Davis 1, Nicholas Pervolarakis 3,4, Zena Werb 5
PMCID: PMC6477686  NIHMSID: NIHMS1019608  PMID: 30482943

Abstract

Tumours comprise a heterogeneous collection of cells with distinct genetic and phenotypic properties that can differentially promote progression, metastasis and drug resistance. Emerging single-cell technologies provide a new opportunity to profile individual cells within tumours and investigate what roles they play in these processes. This Review discusses key technological considerations for single-cell studies in cancer, new findings using single-cell technologies and critical open questions for future applications.


Heterogeneity is pervasive in human cancer and manifests as morphological differences between cells or distinct karyotypic patterns, protein and biomarker expression levels and genetic profiles1,2. Tumours are complex ecosystems of malignant cells surrounded by non-malignant stroma, including fibroblasts, endothelial cells and infiltrating immune cells35. Intratumour heterogeneity arises through various mechanisms (Fig. 1). In clonal evolution models, stochastic accumulation of mutations through genomic instability results in increasing genetic diversity, with the tumour acquiring subclones with distinct genotypes over time6. Heterogeneity is also generated through cellular differentiation. In cancer stem cell (CSC) models, cancers are hierarchically organized with a stem cell-like population, sustaining tumour growth through self-renewal and differentiation7. The tumour microenvironment also generates intratumour heterogeneity by exerting different selective pressures in distinct regions of the tumour811. These models are not mutually exclusive and act together to create a complex system with multiple layers of heterogeneity established by the distinct genetic, epigenetic, transcriptomic, proteomic and functional properties of different cells.

Fig. 1 ∣. Common types of intratumour heterogeneity and its regulation by intrinsic and extrinsic factors.

Fig. 1 ∣

Tumours comprise a heterogeneous population of cells, which is regulated by both intrinsic and extrinsic factors. Tumour cells vary in biomarker expression, epigenetic landscape, hypoxic state, metabolic state, stage of differentiation, invasive potential and genotype due to genomic instability. The tumour microenvironment can also be heterogeneous, in which different types of fibroblasts, pro-tumour and anti-tumour immune infiltrate, vascular and lymphatic vessel density and extracellular matrix (ECM) composition affect tumour cell heterogeneity and function.

Nevertheless, most cancer research and therapy decisions are carried out at the whole-population level. Standard treatment strategies target a single receptor or pathway, treating cancer as a homogenous disease. Even new precision medicine programmes, such as the NCI-MATCH (National Cancer Institute—Molecular Analysis for Therapy Choice) trial, which genetically profiles individual patient tumours to determine the most appropriate targeted therapy, do not consider the number of cells that express the targeted variant and only require it to be detectable above background12. This therapeutic approach may fail for many reasons: if the variant is not critical to drive tumour growth or not expressed in the tumour-promoting cell populations; if some cell populations have additional driver or resistance mutations; or if tumour growth, viability or resistance is encoded at the non-genetic level.

However, technologies for interrogating the whole genome, transcriptome, epigenome and proteome in single cells are maturing. Advances in accuracy, throughput, automation, computational analysis and cost provide the potential to profile thousands of cells from an individual tumour. A first goal in cancer is to characterize the extent of intratumour heterogeneity in individual tumours, at various regulatory levels, from genotype to phenotype, and to spatially localize cell populations within tumours. Subsequently, understanding the function and effect of different cell populations on tumorigenesis, including which features promote tumour initiation, progression or drug resistance, will also be key. Functional characterization will be particularly challenging, as there is no clear method for extrapolating cell function from large-scale ‘omics’ data aside from traditional experimental interrogation. In the long term, new insights may be translated to the clinic, for example, to enable tumour composition analysis for diagnostics and therapeutic assignment, or to identify pre-existing drug-resistant subclones prior to treatment. In this Review, we discuss important technological considerations for experimental designs in cancer research, review single-cell studies that have provided new insights in tumour biology and present open questions for future single-cell applications.

Technological considerations for single-cell studies of cancer

Single-cell technologies have advanced rapidly in the past several years. Currently available protocols vary in cell capture method, library preparation chemistry and throughput (Table 1) (reviewed in refs 1316). Most protocols require single-cell suspension, so the first critical consideration is optimizing tumour dissociation to generate a cell suspension that is fully representative of the intact tumour in terms of cell populations, their frequencies and expression programmes. Digestion of solid tumours eliminates spatial information and can obscure the true programme of individual cells17,18. Although there is no consensus for how to measure these profound effects, cellular diversity after dissociation can be analysed by flow cytometry for known cell types or markers for the specific tumour type. Populations are typically validated by follow-up analyses in situ, but this approach only confirms their existence and does not determine whether all cell populations in the tumour were accounted for after cell dissociation. Ultimately, identification of the same populations using different protocols would increase confidence in the results.

Table 1 ∣.

Technical characteristics, advantages and limitations of single-cell technologies

Transcriptome
Protocol Library type Throughput Advantages Limitations Ref.
SMART-seq Full-length transcriptome Low •Ability to profile rare cell populations directly
•Cell capture visualization
•Splicing variant analysis and SNV profiles possible
•Limited scalability
•Higher degree of manual inputs
•Limited multiplexing possibilities
25,69,77, 111-113
RamDA-seq Full-length transcriptome Low •Ability to profile rare cell populations directly
•Splicing variant analysis and SNV profiles possible
•Ability to capture non-poly(A) transcripts
•Limited scalability
•Higher degree of manual inputs
•Limited multiplexing possibilities
114
Droplet based 3′ Transcriptome High •Reduction in PCR amplification bias
•High cell yield
•Decreased gene coverage per cell
•Sequencing required to estimate cell capture
•No splicing or full-transcript SNV information
•High cell input volumes are not suitable to rarer cell populations
23,110,
115-118
Microwell based 3′ Transcriptome Medium •Cell capture visualization
•Reduction in PCR amplification bias
•Parallel processing of experimental conditions on the same chip
•No splicing or full-transcript SNV information
•High cell input volumes are not suitable to rarer cell populations
119,120
Combinatorial indexing 3′ Transcriptome High •High cell yield
•Ability to run multiple conditions or cell types while maintaining identity
•Reduction in PCR amplification bias
•Decreased gene coverage per cell
•No splicing or full-transcript SNV information
121,122
Single-cell qPCR Targeted transcriptome Medium •Analysis of ‘small RNA’ targets (miRNA, snoRNA and piRNA, among others)
•High sensitivity for lowly expressed transcripts
•Ability to selectively probe for targets of interest
•Limited cell numbers per run
•Limited target transcripts per run
100,123
FISSEQ In situ sequencing High •Spatial localization of transcripts in tissue •High imaging system costs
•Extended protocol length (owing to imaging requirements)
•No rRNA depletion
124
seqFISH In situ hybridization Medium •Spatial localization of transcripts in tissue
•Exact transcript counts per cell
•High imaging system costs
•Limited scalability of transcript targets
125
MERFISH In situ hybridization Medium •Spatial localization of transcripts in tissue
•Reduced misidentification rate owing to a unique encoding scheme
•High imaging system costs
•Potentially high imaging times, resulting in sample degradation
126
Single -nucleus-seq 3′ Nuclear transcriptome High •Fragile cell or tissue processing •Potential bias of nuclear-retained transcripts over exported transcripts 28,127-129
Small RNA-seq 3’ Small RNA Transcriptome Low •Analysis of ‘small RNA’ targets (miRNA, snoRNA and piRNA, among others)
•Reduction in PCR amplification bias
•Limited scalability
•Potential 3′ end bias
130
Genome
Amplification
protocol
Library type Amplification Advantages Limitations Ref.
Ampli1 WGA Ligation-mediated PCR following a site-specific DNA digestion Exponential •Use of non-random primers results in a more even coverage •Long and time-consuming protocol 131
DOP-PCR Primer-based amplification Exponential •Accurate detection of CNVs •The polymerase used has a higher error rate
•Difficult to identify SNVs
89
MDA Phi29 looping amplification Exponential •High-fidelity polymerase
•Suitable for SNV analysis
•Uneven coverage of the total genome
•Difficult to identify CNVs
30,132
MALBAC Combination of looping and primer based Quasi-linear •Accurate enough for SNVs and large CNVs
•Most even read distribution
•Lower confidence SNV identification than MDA 133
LIANTI Linear amplification by transposon insertion Linear High genome coverage, reduced amplification bias and errors
•Increased accuracy for SNVs and CNVs
•Detection of micro CNV at kilobyte resolution
•Despite reduced errors, the false-positive rate (1.7 × 10−6) still prevents exact detection of SNVs 134
Capture method Library type Cost per cell Advantages Limitations Ref.
Whole genome Coverage of the entire genome $$$ •Unbiased
•Powerful for phylogenetic analysis
•Highly dimensional data
•Expensive
•Computationally intense
131,135
Whole exome Coverage of exonic and some regulatory regions $$ •Emphasis on actionable mutations in protein-coding regions •Potential dropout of exonic regions that are relevant owing to inefficient capture 132
Targeted Coverage of specific genomic sites of interest $ •Focused analysis on highly relevant regions
•Cheaper
•Biased and potentially restrictive
•Sacrifice of information
30

DOP-PCR, degenerate oligonucleotide-primed PCR; FISSEQ, fluorescent in situ sequencing; MALBAC, multiple annealing and looping-based amplification cycle; MDA, multiple displacement amplification; MERFISH, multiplexed error-robust fluorescence in situ hybridization; miRNA, microRNA; piRNA, Piwi-interacting RNA; RamDA-seq, random displacement amplification sequencing; seqFISH, sequential fluorescence in situ hybridization; snoRNA, small nucleolar RNA; WGA, whole-genome amplification.

Technologies for transcriptome analysis are the most advanced and have been used to profile CSCs, map differentiation trajectories, describe drug resistance programmes and define the immune infiltrate in tumours1921. The first single-cell transcriptome technologies utilized microfluidics to capture cells followed by multiplex quantitative PCR (qPCR) for selected genes, but most recent studies favour single-cell RNA sequencing (scRNA-seq) to enable assessment of the entire transcriptome. Selection of the most appropriate scRNA-seq protocol depends on the sample size, the number of cells to be sequenced and whether transcript counting or full-length mRNA sequencing is desired (Table 1). When large cell numbers must be sequenced, high-throughput, semi-automated droplet-based approaches (for example, inDrop22 and Dropseq23) are optimal. However, these approaches typically achieve lower transcriptome coverage and detect fewer lowly expressed genes, and there is very limited transcript sequence coverage due to 3′ end counting, precluding single-nucleotide variant (SNV) and splicing analyses17 (Table 1). Droplet-based protocols are also less amenable to studying small cell numbers, for instance, circulating tumour cells (CTCs), disseminated tumour cells or micrometastases. For such samples, cell isolation by flow cytometry or micromanipulation followed by manual library preparation in microwell plates is more tractable. These protocols typically amplify full-length mRNA by switching mechanism at 5′ end of RNA template (SMART) or alternative chemistries, which enable full-length mRNA sequencing for SNV, splicing and deeper transcriptome analyses2427. For tissues that are preserved or cannot be readily dissociated, single-nucleus RNA-seq approaches, such as DroNc-seq28 or microwell-based single-nucleus RNA-seq29, may be optimal (Table 1).

Although less widely utilized, single-cell genome analysis has been used to track clonal dynamics, infer evolutionary trajectories and compare paired primary and metastatic tumours (as detailed below). A major challenge is that the DNA must be massively amplified with minimal error. Several alternative chemistries have been developed for whole-genome amplification, which are alternatively better suited for SNV or copy number variant (CNV) analyses (Table 1). Another major issue for single-cell genome studies is that, unlike whole-transcriptome approaches in which lower sequencing depth can still provide robust information about cell identity, the genome has a fixed, large size and there is effectively no cheap way to sequence it. Many single-cell studies are beginning to opt for lower breadth and sequence either the whole exome or a targeted panel of genes (reviewed in ref. 15). Analysis of single-cell genomic data is also more challenging and less standardized owing to technical errors, such as uneven amplification and allelic dropout. Allelic dropout, in which a particular region of one chromosome is not amplified, is the main technical and analytic challenge and requires imputation of the missing data. Imputation of variants based on probability can be incorrect and confound conclusions. Experimental methods that increase chromosomal compliment, such as in vitro clonal amplification or selection of cells undergoing mitosis, may enable more accurate genotyping30,31. Comparison of whole-genome amplification approaches and selected analytic methods can be found in refs 15,3236.

Protocols to measure other regulatory levels, such as the epigenome and proteome, are developing rapidly, as are multiscale approaches to analyse multiple regulatory levels in the same cell (that is, multi-omics) and methods for spatial analysis in intact tissues. Selected methods are highlighted in Box 1.

Box 1 ∣. Emerging technologies for single-cell analyses in cancer.

Single-cell transcriptome and genome studies have emerged as vital tools for investigating mechanisms of cancer, and more technologies are being adapted to operate in single-cell resolution to explore the disease in ways that were previously unavailable. This is exemplified by several common protocols for epigenetic analysis that now can be performed at the single-cell level, including DNase-seq136 and ATAC-seq137,138 for defining regions of open chromatin, Hi-C for investigation of chromosomal contacts139, ChIP–seq for histone position mapping140, bisulfite sequencing (single-cell bisulfite-seq)141 or bisulfite-free methods (single-cell CGI-seq)142 for measuring DNA methylation state, and CLEVER-seq143 and scAba-seq144 to measure active DNA demethylation by 5-fluorocytosine sequencing and 5-hydroxymethylcytosine sequencing, respectively. Multiscale analysis of multiple regulatory levels in the same cell is also an area of rapid development and provides the potential to comprehensively understand how and why malignant cells produce a particular phenotype, function or behaviour. Combinatorial methods currently exist for analysis of the genome and transcriptome (G&T-seq and DR-seq)145147, epigenome and transcriptome (scMT-seq, scTrio-seq and scNMT-seq)148150, and techniques for studying the proteome and transcriptome are in development151,152. Single-cell proteomics are another area of paramount interest as protein expression is the ultimate functional output of the cell. Although there are no commercial methods for whole-proteome level analysis, several technologies for high-parameter protein analysis in single cells have been developed. Time of flight mass cytometry (CyTOF), which utilizes heavy metal-conjugated antibodies to quantify protein expression by time-of-flight inductively coupled plasma mass spectrometry, can theoretically multiplex up to 135 parameters in single cells153155. The co-detection by indexing (CODEX) platform also enables high-parameter protein expression analysis using an in situ polymerization-based indexing procedure and fluorescently barcoded antibodies156. This provides the added advantage of spatial localization of cell populations within the native tissue context. Spatial genomics technologies for single-cell analysis, such as FISSEQ157, seqFISH158 or MERFISH126, are another area of rapid development, which reveal cellular neighbourhoods that are specific to the tumour microenvironment. Ultimately, spatial approaches will allow investigators to define changes in cell populations that associate with specific histological and pathological tissue phenotypes.

Genetic heterogeneity and subclonal dynamics

Most tumours comprise subpopulations of cells with distinct genotypes called subclones. Next-generation sequencing data have revealed that a tumour possesses on average over 10,000 somatic mutations: ~2–8 in ‘driver’ genes that confer a selective growth advantage, and ~30–60 protein-coding changes in ‘passenger’ genes that may alter other cellular functions37. Subclonal diversification arises through genomic instability and numerous mechanisms, including homologous recombination deficiency, chromosome instability, chromothripsis, misregulation of APOBEC enzyme activity and drug treatment6,3846. Several models for subclonal diversification have been proposed based on next-generation sequencing data of bulk human tumour samples, the prevailing being linear, branching, neutral and punctuated models (reviewed in ref. 47). Most next-generation sequencing studies report branching evolution in human cancers, including leukaemia, breast and liver cancers, colorectal cancer, ovarian cancer, prostate cancer, kidney cancer, melanoma and brain cancer (reviewed in ref. 47).

Although sophisticated statistical and mathematical models have been developed to infer subclonal dynamics and tumour evolution from bulk data, they rely on major assumptions and cannot extrapolate single-cell genotypes because overlapping mutation frequencies cannot be assigned to the same or different cells, technical errors may yield imprecise mutation frequencies and detection limits preclude identification of rare subclones15 (Fig. 2). Single-cell genome analyses mitigate many of these limitations and, most importantly, can determine whether mutations are in the same or different cells. This can help to address important questions about subclonal dynamics, such as how specific subclones interact (for example, collaborate versus compete), which subclones can invade and metastasize, how subclonal composition affects clinical outcome and how drug resistance evolves (for example, from pre-existing clones versus the acquisition of new mutations).

Fig. 2 ∣. Deciphering subclonal composition and cell types and states in single-cell omics data.

Fig. 2 ∣

a, Inferring clonal trajectories and subclonal heterogeneity from bulk primary tumour genome sequencing data. In this example experiment, a tumour is sampled at a single time point (dotted lines). The table shows the frequency of each detected mutation. The panels show three (of many) possible clonal trajectories that can be inferred. The nodes represent points at which a mutation occurred, and overlapping coloured regions indicate that each of the mutations is present within any cell that is part of that population. b, Challenges associated with deciphering the genotype of a metastatic founder clone and subclonal trajectories from bulk genome sequencing of paired metastatic and primary tumours. The table shows the observed mutation frequencies in an example experiment in which a metastatic tumour from the individual in a was sequenced. The panels show three possible explanations for the observed frequencies. c, Cell types and states found in normal tissues. Tissues comprise different mature ‘cell types’ (labelled 1–5), which carry out specified functions. Cells within a ‘type’ can exist in a spectrum of allowable ‘cell states’ depending on the physiological status of the tissue. Mature cell types are derived from stem cells through a series of discrete differentiation intermediates or progenitors. The circles represent single cells, and the colour clouds represent the spectrum of allowable states. The density of circles represents the probability of observing a cell with that phenotype in a scRNA-seq experiment. d, Tumour cell types and states differ from normal tissue. Single-cell studies have shown that tumours contain stem-like cells (CSCs) and that differentiation is often noisy, skewed towards specific cell lineages.

Single-cell studies of subclonal heterogeneity have produced new details about subclonal frequencies and their evolution during tumour progression. Single-nucleus sequencing of breast tumours showed that copy number evolution occurred in short bursts early in tumour evolution, whereas point mutations evolved gradually over time to produce more extensive clonal diversity48. This provides support for a combined punctuated–branched model for tumour evolution in breast cancer. Genetically distinct clones with unique biological and clinical properties have been identified in acute lymphoblastic leukaemia, colon and breast tumours by single-cell genome analysis49,50. For example, targeted single-cell SNV analysis of patients with acute lymphoblastic leukaemia revealed codominant clones, and showed that KRAS mutations occurred late in disease development but were not sufficient for clonal dominance49. In a case study of colon cancer, the dominant clone possessed APC and TP53 mutations typical of colon cancers, but a rare subclone with CDC27 and PABPC1 mutations was also identified50. These studies suggest that combinatorial therapies that target multiple subclones may produce better results against polyclonal disease.

A study investigating clonal evolution during breast cancer invasion showed a direct lineage relationship between non-invasive ductal carcinoma in situ and adjacent invasive ductal carcinoma lesions in individual patients with breast cancer using topographical single-cell sequencing51. These results indicate that most mutations and CNVs evolved within the ducts prior to invasion, with multiple clones escaping from the ducts and co-migrating into the adjacent tissues to establish invasive carcinomas. These findings contrast with models for cancer cell invasion proposing that distinct clones give rise to in situ and invasive tumour cells5254, and argue against the notion that extrinsic stimuli (that is, ‘field effects’) cause multifocal disease.

Single-cell genetic analysis has also provided new insights into the longstanding debate over whether drug resistance is caused by the selection of rare pre-existing clones or through acquired resistance by induction of new mutations. Single-cell DNA sequencing of longitudinal samples from patients with breast cancer before and after neoadjuvant chemotherapy showed that resistant genotypes are pre-existing and selected by chemotherapy55. Interestingly, scRNA-seq of the same samples showed that the transcriptome profiles of cells pre-treatment and post-treatment were entirely distinct. Cells with resistant programmes were undetectable before treatment, although subsets of cells expressing individual genes associated with chemoresistance could be identified. These data indicate that chemoresistance is conferred through both genetic selection and induction of new transcriptome programmes.

Single-cell analyses of CTCs have also revealed genetic mechanisms of drug resistance. Studies of CTCs from patients with breast cancer have shown heterogeneity for genes important for diagnosis and therapy response, such as ERBB2 and PIK3CA56,57. Analysis of CTCs from patients with small-cell lung cancer revealed distinct molecular mechanisms for resistance to chemotherapy58. CTCs from chemoresistant patients displayed different CNV profiles than CTCs from chemorefractory patients, suggesting a different genetic basis for immediate resistance (chemorefractory) versus delayed resistance (chemoresistant). A classifier was also developed to distinguish between the two types of patients, supporting the application of single-cell genomics for CTC-based diagnostics58.

There are still several limitations to single-cell genomic analyses that prevent their more general use in experimental and clinical practice. Technical limitations that hinder confident SNV calling due to allelic dropout are a major issue. Understanding of the biological function and clinical importance of specific genetic clones is also lacking. It will be critical to decipher how a single-cell genotype translates into cellular function, how individual clones affect tumour behaviour and how clones interact to promote tumour progression, metastasis and drug resistance. Innovative strategies are needed to address the lack of a universal approach for the isolation of specific genetic clones and experimental interrogation of their function.

Non-genetic heterogeneity and cellular differentiation

ScRNA-seq studies have shown that many normal tissues are maintained by a pool of adult stem cells that differentiate into multiple ‘cell types’ with distinct ‘cell states’ distinguished by more subtle differences in differentiation, activation, metabolic state or stage of the cell cycle5965 (Fig. 2c). Similarly, tumours also contain CSCs, which behave like normal stem cells in their capacity to self-renew, differentiate and propagate the tumour upon transplant (reviewed in ref. 66). However, some tumours do not follow this model67,68. One outstanding question is to what degree CSCs and other tumour cell populations maintain the developmental programme of their normal cell counterparts, versus dedifferentiating or assuming an aberrant cell state (Fig. 2d). Single-cell technologies provide the opportunity to measure cell states in individual cells and map tumour cells onto the normal spectrum of allowable cell types and states of their tissue of origin.

One of the first single-cell studies in cancer used a single-cell multiplex qPCR approach to show that human colon cancers contain distinct cell populations that mirror the cellular lineages of the normal colon19. This transcriptional heterogeneity was not due to underlying genetic heterogeneity, as injection of single CSCs into immune-deficient mice gave rise to monoclonal tumours as heterogeneous as the parental one. A study of human oligodendroglioma using scRNA-seq20 showed that tumours were composed of astrocyte-like and oligodendrocyte-like cells and a rare subpopulation of undifferentiated cells resembling neural stem cells. Similar to the colon cancer study, CNV and SNV analysis showed that each subclonal lineage displayed similar hierarchies. Both studies found differences in the tumour cells relative to their normal counterparts. In the oligodendroglioma study, the two glial lineages seemed to originate from the stem cell-like population, without the discrete differentiation intermediates observed in the normal tissue. Rather, a continuum of differentiation profiles was observed along each lineage, indicating that tumour cells may exist in more dynamic and less discrete differentiation states than normal cells20 (Fig. 2d). Another study of oligodendroglioma showed that progression to more advanced stages was associated with expansion of primitive cells, with tumours bearing less similarity to the cellular composition of normal tissues69. However, the lack of normal cell population profiling in these studies makes it difficult to discern how similar each population was to its healthy counterpart. Likewise, in the colon cancer study19, several cell subpopulations typical of the normal colon were absent in the tumours, suggesting that there was a skewing or block in normal differentiation19 (Fig. 2d).

Single-cell studies of acute myelogenous leukaemia (AML) have shown similar alterations in normal differentiation. A mouse model of AML was shown to contain two cell types: one resembling granulocyte/monocyte progenitors and the other macrophage and dendritic cells70. Normal and leukaemic counterparts showed significantly different gene network modules, suggesting an aberrant programme in leukaemic cells. Altered myeloid differentiation has also been observed in human paediatric AML by single-cell mass cytometry. In one report, two alternative approaches, PhenoGraph and statistical analysis of perturbation response (SARA), were used to compare signalling programmes in normal haematopoietic and leukaemic cells71. Although both primitive and mature monocytelike cells were identified in all patients, they displayed different degrees of aberrant myeloid differentiation, which did not correlate with the cell-surface marker phenotype. In AML, enumeration of primitive CD34+/CD45low blasts is often used for clinical diagnosis and classification of leukaemias72. This study showed that intrinsic signalling programmes were often decoupled from the cell-surface phenotype, challenging the standard diagnostic approach. Many patients possessed CD34 leukaemic cells displaying primitive signalling programmes, and the percentage of primitive cells rather than cell-surface phenotype was more predictive of overall survival71.

These studies suggest that cancer cells often resemble normal cell types and states, but acquire aberrant programmes and display skewed differentiation towards more restricted lineages than their tissue of origin. Defining the aberrant programmes in single cells with relevance to stability and plasticity may reveal new mechanistic insight and therapeutic targeting strategies.

Heterogeneity in diagnostics and therapy response

Mechanisms of drug response have also been studied at the single-cell level. CTCs have been a major focus as they offer a non-invasive window into tumour response. One study reveals that the majority of patients with prostate cancer with varying stages of disease and resistance to androgen deprivation therapies harboured CTCs with at least one type of androgen receptor alteration73. Furthermore, patients on the second-line androgen receptor inhibitor enzalutamide (administered after the development of resistance to androgen deprivation therapy) often had CTCs with activated non-canonical Wnt signalling, showcasing how different mechanisms for generating non-genetic heterogeneity can produce drug resistance, and that tracking this through assaying CTCs is clinically feasible.

Single-cell mass cytometry has also been used to measure differential responses to chemotherapy in patients with AML. The percentage of proliferating stem and progenitor-like leukaemic cells was shown to be significantly correlated with therapy response, in addition to signalling abnormalities in primitive AML cell populations74. Patients with clinically favourable subtypes of AML demonstrated a higher fraction of cells in S-phase than other subtypes. This suggests that the quiescent state facilitates cancer cell protection and survival during therapy. In another study, chemotherapy increased phenotypic diversity away from traditional stem cell phenotypes in patients with AML75. This finding contrasts with classic resistance models that suggest selection of a more primitive cell population during treatment results in outgrowth of a more uniform population.

ScRNA-seq has also shown how standard diagnostic bulk tumour profiling can mask clinically relevant heterogeneity. Glioblastomas can be stratified into four distinct subtypes based on their mRNA expression signature: proneural, neural, classical and mesenchymal76. Interestingly, scRNA-seq analysis showed that glioblastomas contain cells that resemble each of the four subtypes, even though this is not observed in bulk analyses. Increased intratumour heterogeneity was also associated with decreased survival, perhaps because of improper patient stratification and treatment assignment, or enhanced genomic instability, tumour evolution or resistance77.

Single-cell analyses may also eventually be valuable for predictive diagnostics. Single-cell analysis of targeted transcriptome (SCATTome), which predicts drug response for individual cells based on transcriptome signatures using an assortment of machine learning methods, was able to predict response to proteasome inhibitor treatment in patients with multiple myeloma78.

Despite these new discoveries, several limitations of single-cell analyses are worth further consideration. As in single-cell genome studies, assigning function to cell populations is the biggest challenge for most non-genetic single-cell studies. Gene ontology and gene set enrichment analyses can provide clues for signalling pathways that are overrepresented in a cell population, but most of these tools are developed for bulk transcriptome analysis and handle missing data poorly (reviewed in refs 79,80). As most scRNA-seq protocols only detect 10–40% of the transcriptome, they are not optimal for studying lowly expressed genes, such as those encoding transcription factors, or for the investigation of specific signalling pathways. Dropout of signalling intermediates makes determining whether a pathway is activated challenging. It is also often difficult to determine what constitutes a true cell population, as slight changes in clustering parameters can produce different results. How to decide what represents a cell type versus a cell state continues to be a major debate in the field, particularly for rare cell populations, which may represent cells transiently switching from one state to another or true rare cell types. The only way to assign function to a cell population is through conventional experimentation, which most single-cell reports lack. It is also important to consider that tumour behaviour is probably more than just the sum of its individual cell parts, with different cell populations synergizing or collaborating to take on new macroscale functions and behaviours that are not observable using reductionist approaches. If so, new integrated informatic, experimental and modelling approaches for analysing single cells will be necessary to truly understand tumour behaviour.

Heterogeneity and metastasis

Metastasis remains the cause of most patient mortality and continues to be challenging to treat clinically and investigate experimentally. The general dogma is that metastasis is carried out by rare cells with unique cellular and molecular properties81. Single-cell investigations now enable the identification and characterization of such cells, including their localisation in primary tumours, and the effect of genetic versus non-genetic and intrinsic versus extrinsic factors on metastasis (Fig. 3).

Fig. 3 ∣. Genetic and phenotypic properties of metastasis-initiating cells at the single-cell level.

Fig. 3 ∣

Metastasis is a rare event, in which most cancer cells cannot progress through major bottlenecks associated with invasion, intravasation, extravasation, seeding and colonization to produce a malignant macrometastatic tumour. In this model, cancer cells are heterogeneous in genotype (nuclei) and phenotype (cytoplasm), and metastasis-initiating cells possess a distinct combination of both. Dashed arrow indicates that cancer cells within micrometastases can die. Death rates within micrometastases can balance proliferation rates, and thereby prevent progression to macrometastasis by the failure to produce net positive growth.

Next-generation sequencing studies of bulk tumour samples indicate that metastasis is initiated by a subclone of the primary tumour. In one study, whole-genome sequencing identified numerous point mutations and small indels represented in higher frequencies in the brain metastatic tumour than in the paired primary breast tumour82. Similar disparities in mutation frequencies have been reported in paired tumours and metastases from patients with pancreatic and renal cancers, in which the metastasis founder subclones localized to a specific region in the primary tumours, supporting a subclonal model for metastasis initiation83,84. One confounding issue in these studies is that the metastases often possessed unique mutations that were not found in paired primary tumours83. This makes it challenging to infer the genotype of the original metastasis founder cells, because it is not clear whether the metastasis-exclusive mutations were present in the primary tumour below the limit of detection or whether they arose after metastatic seeding via parallel evolution (Fig. 2b).

Single-cell genome studies of metastasis are beginning to resolve these challenges. Early single-cell studies of metastasis focused on disseminated tumour cells in the bone marrow of patients with breast cancer, and found that these cells disseminate early in tumour evolution by karyotype and subchromosomal variant analyses85,86. This is corroborated by reports showing that metastatic lesions similarly derive from early dissemination events87,88. Other single-cell genetic methods have generated contradictory findings. A single-cell CNV analysis of breast cancer metastasis showed that a single clonal expansion formed the patient primary tumour and seeded its metastasis89. Single-cell CNV and SNV analysis in patients with colon cancer showed monoclonal metastatic seeding in one patient and polyclonal in another90. The authors of that study also concluded that metastasis occurred late in primary tumour evolution in both patients, as the metastases harboured all of the trunk mutations that were present in the primary tumour cells8588,9193.

Single-cell genomic analyses also provide evidence for collective cell migration as a mechanism for cancer cell invasion. Topographical single-cell sequencing analysis showed that multiple clones co-migrated through the basement membrane of breast ducts and into adjacent tissues to establish invasive breast carcinomas51. This is consistent with previous work showing collective cell migration at the invasive front of tumours, as well as observations that CTC clusters in the bloodstream are more effective than single CTCs for seeding metastasis9496.

Single-cell genome analyses of patient CTCs also provide insight into the genotype of potential metastasis-initiating cells. CTCs display substantial subclonal diversity, suggesting that cells of various genotypes are capable of entering the circulation97. This includes very rare subclones, as CTCs from patients with colon cancer were reported to carry SNVs that were only present in the paired primary tumours at very low frequencies98. Another study found that CTCs in patients with breast cancer often possess variants that are not found in the primary tumour, indicating that they either represent a rare subclone or occur after dissemination57. Importantly, the CTCs were heterogeneous for mutations in two common breast cancer drug targets, ERBB2 and PIK3CA, so identifying which CTCs subsequently produce metastases has direct clinical relevance.

As it is clear that metastatic propensity is not encoded exclusively at the genetic level (reviewed in ref. 99), it will be necessary to investigate other programmes (for example, transcriptomic and epigenetic) driving metastatic progression at single-cell resolution. Single-cell multiplex qPCR technology has shown that metastasis is initiated by cells with stem cell and epithelial–mesenchymal transition-like characteristics in patient-derived xenograft models of breast cancer100. This is consistent with results from the MMTV-PyMT breast cancer mouse model showing CSCs as the origin of metastasis101, as well as reports implicating stem cell and epithelial–mesenchymal transition programmes in other breast cancer models93,102. A recent scRNA-seq study of human head-and-neck cancers further implicated epithelial–mesenchymal transition in metastasis103. Future studies should utilize single-cell technologies as a new opportunity to investigate other major outstanding questions about metastatic progression, such as what drives metastatic latency and reawakening, how metastatic cells interact with their microenvironment and how they develop resistance to anti-tumour immunity.

Heterogeneity in the microenvironment

The cellular composition of the tumour microenvironment is critical for disease progression and patient prognosis104 and changes dramatically during tumorigenesis due to increased angiogenesis, tumour-associated inflammation and fibrosis8. Emerging single-cell tools bear great potential to profile the individual cell types comprising the tumour ecosystem, to determine how they differ from normal homeostasis and to reconstruct feedback mechanisms mediating cell-to-cell communications between tumour and stromal cells (Fig. 1).

A fundamental caveat in bulk cancer genomic analyses (for example, The Cancer Genome Atlas) is that the differences in transcriptomic signatures between tumours may arise due to differential infiltration of immune and other stromal cell types rather than differences in the tumour cells. This can be addressed with analyses at single-cell resolution. Indeed, scRNA-seq analysis has revealed that the bulk transcriptome differences between oligodendroglioma and astrocytoma driven by mutant IDH are primarily due to alterations within stromal macrophage and microglial populations69. Another study matched the transcriptomes of non-malignant cells to cell-type-specific expression profiles and inferred the signalling dialogue between cancer cells and their microenvironment21. This revealed distinct patterns of exhaustion or activation of tumour-infiltrating T cells in some tumours, suggesting they may exhibit differential responses to immunotherapies21.

Breakthroughs in cancer immunotherapy have sparked intensive research into tumour–immune cell interactions using genomic tools, and single-cell analysis pipelines will be instrumental in these approaches105. ScRNA-seq analysis of human metastatic melanoma revealed specific homeostatic modules in monocytes and dendritic cells within the tumour microenvironment106. A recent systems-level approach using single-cell mass cytometry demonstrated that engagement of systemic and peripheral immunity is critical for tumour rejection after immunotherapy107. Future approaches would need to dissect the specificity of anti-tumour immunity before and after checkpoint inhibition, for example, using single-cell V(D)J sequencing to identify the T cell and B cell clones associated with anti-tumour immunity108. Direct comparison of single-cell expression signatures from stromal cells in the tumour microenvironment to those in other physiologically similar conditions (for example, inflammation or wound healing) will also determine whether the tumour context induces changes within the normal spectrum of states that a cell may adopt under different conditions or whether cells assume an aberrant tumour microenvironment-specific programme that is not found in normal physiological contexts (Fig. 3c,d).

Future directions

Given its effect on tumour behaviour and clinical outcome, measurements of intratumour heterogeneity should be increasingly incorporated into standard clinical practice. Single-cell targeted DNA sequencing has been commercialized for cancer diagnostics in AML109. In addition, commercial scRNA-seq platforms are simple, automated and fast enough to be immediately amenable for diagnostic applications. The continuous release of higher-throughput sequencers will also continue to drive cost down110. Thus, the main hurdles to clinical implementation will probably be upstream and downstream of the single-cell technology itself. First, analysis of single-cell sequencing data requires computational skill, so pipelines for demultiplexing, alignment and cell population analysis should be packaged into software and integrated into instrumentation in a single workflow. Furthermore, we lack automated instrumentation or consensus protocols to generate single-cell suspensions from an excised tumour without human intervention. Typical laboratory workflows are manual, low-throughput and not sufficiently reproducible for clinical application. Third, sampling bias is a notable hindrance and it may be necessary to catalogue whole tumours for accurate assessment of heterogeneity. In many cases, only small biopsies are available or patients are pre-treated with neoadjuvant therapies before sample procurement, which may alter the cellular composition of the tumour. However, the main hurdle to clinical implementation will be data interpretation. To harness the power of single-cell assessment of intratumour heterogeneity beyond descriptive cataloguing, we need to first elucidate the biological and clinical functions of different cell populations and develop new ways to specifically target them.

Acknowledgements

We thank those whose work informed the writing of this manuscript and apologize to those authors whose elegant studies we were unable to acknowledge in this Review. We thank K. Blake and J. Wu for thoughtful discussion and suggestions regarding the content of this Review. This work was supported by NIH grants (U01CA199315 to Z.W., K22 CA190511 to D.A.L. and R00 CA181490 to K.K.) and the Chan/Zuckerberg Initiative (HCA-A-1704-01668 to K.K. and D.A.L.). N.P was supported by the National Institute of Biomedical Imaging and Bioengineering, National Research Service Award T32 EB009418 from the University of California, Irvine, Center for Complex Biological Systems. R.T.D. was supported by the NIH, NCI Award T32CA009054 through matched funds.

Footnotes

Competing interests

The authors declare no competing interests.

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Heppner GH Tumor heterogeneity. Cancer Res. 44, 2259–2265 (1984). [PubMed] [Google Scholar]
  • 2.Welch DR Tumor heterogeneity∔a ‘contemporary concept’ founded on historical insights and predictions. Cancer Res. 76, 4–6 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mroz EA et al. High intratumor genetic heterogeneity is related to worse outcome in patients with head and neck squamous cell carcinoma. Cancer 119, 3034–3042 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Marusyk A, Almendro V & Polyak K Intra-tumour heterogeneity: a looking glass for cancer? Nat. Rev. Cancer 12, 323–334 (2012). [DOI] [PubMed] [Google Scholar]
  • 5.McGranahan N et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.McGranahan N & Swanton C Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 27, 15–26 (2015). [DOI] [PubMed] [Google Scholar]
  • 7.Kreso A & Dick JE Evolution of the cancer stem cell model. Cell Stem Cell 14, 275–291 (2014). [DOI] [PubMed] [Google Scholar]
  • 8.Quail DF & Joyce JA Microenvironmental regulation of tumor progression and metastasis. Nat. Med 19, 1423–1437 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Pistollato F et al. Intratumoral hypoxic gradient drives stem cells distribution and MGMT expression in glioblastoma. Stem Cells 28, 851–862 (2010). [DOI] [PubMed] [Google Scholar]
  • 10.Widmer DS et al. Hypoxia contributes to melanoma heterogeneity by triggering HIF1α-dependent phenotype switching. J. Invest. Dermatol 133, 2436–2443 (2013). [DOI] [PubMed] [Google Scholar]
  • 11.Black JC et al. Hypoxia drives transient site-specific copy gain and drug-resistant gene expression. Genes Dev. 29, 1018–1031 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lih CJ et al. Analytical validation of the next-generation sequencing assay for a nationwide signal-finding clinical trial: Molecular Analysis for Therapy Choice Clinical Trial. J. Mol. Diagn 19, 313–327 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Nguyen QH, Pervolarakis N, Nee K & Kessenbrock K Experimental considerations for single-cell RNA sequencing approaches. Front. Cell Dev. Biol 6, 108 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kalisky T et al. A brief review of single-cell transcriptomic technologies. Brief Funct. Genomics 17, 64–76 (2018). [DOI] [PubMed] [Google Scholar]
  • 15.Gawad C, Koh W & Quake SR Single-cell genome sequencing: current state of the science. Nat. Rev. Genet 17, 175–188 (2016). [DOI] [PubMed] [Google Scholar]
  • 16.Van Loo P & Voet T Single cell analysis of cancer genomes. Curr. Opin. Genet. Dev 24, 82–91 (2014). [DOI] [PubMed] [Google Scholar]
  • 17.Svensson V et al. Power analysis of single-cell RNA-sequencing experiments. Nat. Methods 14, 381–387 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.van den Brink SC et al. Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations. Nat Methods 14, 935–936 (2017). [DOI] [PubMed] [Google Scholar]
  • 19.Dalerba P et al. Single-cell dissection of transcriptional heterogeneity in human colon tumors. Nat. Biotechnol 29, 1120–1127 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tirosh I et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature 539, 309–313 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tirosh I et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Klein AM et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Macosko EZ et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Picelli S et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013) [DOI] [PubMed] [Google Scholar]
  • 25.Picelli S et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc 9, 171–181 (2014). [DOI] [PubMed] [Google Scholar]
  • 26.Bhargava V, Head SR, Ordoukhanian P, Mercola M & Subramaniam S Technical variations in low-input RNA-seq methodologies. Sci. Rep 4, 3678 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bhargava V, Ko P, Willems E, Mercola M & Subramaniam S Quantitative transcriptomics using designed primer-based amplification. Sci. Rep 3, 1740 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Habib N et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat Methods 14, 955–958 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gao R et al. Nanogrid single-nucleus RNA sequencing reveals phenotypic diversity in breast cancer. Nat. Commun 8, 228 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Leung ML et al. Highly multiplexed targeted DNA sequencing from single nuclei. Nat. Protoc 11, 214–235 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Leung ML, Wang Y, Waters J & Navin NE SNES: single nucleus exome sequencing. Genome Biol. 16, 55 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Huang L, Ma F, Chapman A, Lu S & Xie XS Single-cell whole-genome amplification and sequencing: methodology and applications. Annu. Rev. Genomics Hum. Genet 16, 79–102 (2015). [DOI] [PubMed] [Google Scholar]
  • 33.Borgstrom E, Paterlini M, Mold JE, Frisen J & Lundeberg J Comparison of whole genome amplification techniques for human single cell exome sequencing. PLoS ONE 12, e0171566 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Navin NE & Chen K Genotyping tumor clones from single-cell data. Nat. Methods 13, 555–556 (2016). [DOI] [PubMed] [Google Scholar]
  • 35.Schwartz R & Schaffer AA The evolution of tumour phylogenetics: principles and practice. Nat. Rev. Genet 18, 213–229 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Salehi S et al. ddClone: joint statistical inference of clonal populations from single cell and bulk tumour sequencing data. Genome Biol. 18, 44 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vogelstein B et al. Cancer genome landscapes. Science 339, 1546–1558 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Alexandrov LB et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Burns MB, Temiz NA & Harris RS Evidence for APOBEC3B mutagenesis in multiple human cancers. Nat. Genet. 45, 977–983 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Roberts SA et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers. Nat. Genet 45, 970–976 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Abkevich V et al. Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer. Br. J. Cancer 107, 1776–1782 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Popova T et al. Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation. Cancer Res. 72, 5454–5462 (2012). [DOI] [PubMed] [Google Scholar]
  • 43.Stephens PJ et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Zack TI et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet 45, 1134–1140 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Meier B et al. C. elegans whole-genome sequencing reveals mutational signatures related to carcinogens and DNA repair deficiency. Genome Res. 24, 1624–1636 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Johnson BE et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 343, 189–193 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Davis A, Gao R & Navin N Tumor evolution: Linear, branching, neutral or punctuated? Biochim. Biophys. Acta 1867, 151–161 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang Y et al. Clonal evolution in breast cancer revealed by single nucleus genome sequencing. Nature 512, 155–160 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gawad C, Koh W & Quake SR Dissecting the clonal origins of childhood acute lymphoblastic leukemia by single-cell genomics. Proc. Natl Acad. Sci. USA 111, 17947–17952 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yu C et al. Discovery of biclonal origin and a novel oncogene SLC12A5 in colon cancer by single-cell sequencing. Cell Res. 24, 701–712 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Casasent AK et al. Multiclonal invasion in breast tumors identified by topographic single cell sequencing. Cell 172, 205–217.e12 (2018) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Gerlinger M et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet 46,225–233 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Miron A et al. PIK3CA mutations in in situ and invasive breastcarcinomas. Cancer Res. 70, 5674–5678 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yates LR et al. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat. Med 21, 751–759 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kim C et al. Chemoresistance evolution in triple-negative breast cancer delineated by single-cell sequencing. Cell 173, 879–893.e13 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pestrin M et al. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Mol. Oncol 9, 749–757 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Polzer B et al. Molecular profiling of single circulating tumor cells with diagnostic intention. EMBO Mol. Med 6, 1371–1386 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Carter L et al. Molecular analysis of circulating tumor cells identifies distinct copy-number profiles in patients with chemosensitive and chemorefractory small-cell lung cancer. Nat. Med 23, 114–119 (2017). [DOI] [PubMed] [Google Scholar]
  • 59.Trapnell C Defining cell types and states with single-cell genomics. Genome Res. 25, 1491–1498 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Treutlein B et al. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature 509, 371–375 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Pollen AA et al. Low-coverage single-cell mRNA sequencing reveals cellular heterogeneity and activated signaling pathways in developing cerebral cortex. Nat. Biotechnol 32, 1053–1058 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ahn RS et al. Transcriptional landscape of epithelial and immune cell populations revealed through FACS-seq of healthy human skin. Sci. Rep 7, 1343 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Jaitin DA et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bjorklund AK et al. The heterogeneity of human CD127+ innate lymphoid cells revealed by single-cell RNA sequencing. Nat. Immunol 17, 451–460 (2016). [DOI] [PubMed] [Google Scholar]
  • 65.Chen L et al. Transcriptomes of major renal collecting duct cell types in mouse identified by single-cell RNA-seq. Proc. Natl Acad. Sci. USA 114, E9989–E9998 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Batlle E & Clevers H Cancer stem cells revisited. Nat. Med 23, 1124–1134 (2017). [DOI] [PubMed] [Google Scholar]
  • 67.Quintana E et al. Efficient tumour formation by single human melanoma cells. Nature 456, 593–598 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ball CR et al. Succession of transiently active tumor-initiating cell clones in human pancreatic cancer xenografts. EMBO Mol. Med 9, 918–932 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Venteicher AS et al. Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science 355, eaai8478 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Saadatpour A, Guo G, Orkin SH & Yuan GC Characterizing heterogeneity in leukemic cells using single-cell gene expression analysis. Genome Biol. 15, 525 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Levine JH et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell 162, 184–197 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Craig FE & Foon KA Flow cytometric immunophenotyping for hematologic neoplasms. Blood 111, 3941–3967 (2008). [DOI] [PubMed] [Google Scholar]
  • 73.Miyamoto DT et al. RNA-seq of single prostate CTCs implicates noncanonical Wnt signaling in antiandrogen resistance. Science 349, 1351–1356 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Behbehani GK et al. Mass cytometric functional profiling of acute myeloid leukemia defines cell-cycle and immunophenotypic properties that correlate with known responses to therapy. Cancer Discov. 5, 988–1003 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ferrell PB Jr et al. High-dimensional analysis of acute myeloid leukemia reveals phenotypic changes in persistent cells during induction therapy. PLoS ONE 11, e0153207 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Verhaak RG et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17, 98–110 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Patel AP et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Mitra AK et al. Single-cell analysis of targeted transcriptome predicts drug sensitivity of single cells within human myeloma tumors. Leukemia 30, 1094–1102 (2016). [DOI] [PubMed] [Google Scholar]
  • 79.Poirion OB, Zhu X, Ching T & Garmire L Single-cell transcriptomics bioinformatics and computational challenges. Front. Genet 7, 163 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wagner A, Regev A & Yosef N Revealing the vectors of cellular identity with single-cell genomics. Nat. Biotechnol 34, 1145–1160 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Massague J & Obenauf AC Metastatic colonization by circulating tumour cells. Nature 529, 298–306 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Ding L et al. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature 464, 999–1005 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Gerlinger M et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med 366, 883–892 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yachida S et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467, 1114–1117 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Schardt JA et al. Genomic analysis of single cytokeratin-positive cells from bone marrow reveals early mutational events in breast cancer. Cancer Cell 8, 227–239 (2005). [DOI] [PubMed] [Google Scholar]
  • 86.Schmidt-Kittler O et al. From latent disseminated cells to overt metastasis: genetic analysis of systemic breast cancer progression. Proc. Natl Acad. Sci. USA 100, 7737–7742 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Husemann Y et al. Systemic spread is an early step in breast cancer. Cancer Cell 13, 58–68 (2008). [DOI] [PubMed] [Google Scholar]
  • 88.Hosseini H et al. Early dissemination seeds metastasis in breast cancer. Nature 540, 552–558 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Navin N et al. Tumour evolution inferred by single-cell sequencing. Nature 472, 90–94 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Leung ML et al. Single-cell DNA sequencing reveals a late-dissemination model in metastatic colorectal cancer. Genome Res. 27, 1287–1299 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Riethmuller G & Klein CA Early cancer cell dissemination and late metastatic relapse: clinical reflections and biological approaches to the dormancy problem in patients. Semin. Cancer Biol 11, 307–311 (2001). [DOI] [PubMed] [Google Scholar]
  • 92.Linde N et al. Macrophages orchestrate breast cancer early dissemination and metastasis. Nat. Commun 9, 21 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Harper KL et al. Mechanism of early dissemination and metastasis in Her2+ mammary cancer. Nature 540, 588–592 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Cheung KJ, Gabrielson E, Werb Z & Ewald AJ Collective invasion in breast cancer requires a conserved basal epithelial program. Cell 155, 1639–1651 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Liotta LA, Saidel MG & Kleinerman J The significance of hematogenous tumor cell clumps in the metastatic process. Cancer Res. 36, 889–894 (1976). [PubMed] [Google Scholar]
  • 96.Aceto N et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 158, 1110–1122 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ni X et al. Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients. Proc. Natl Acad. Sci. USA 110, 21083–21088 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Heitzer E et al. Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing. Cancer Res. 73, 2965–2975 (2013). [DOI] [PubMed] [Google Scholar]
  • 99.Nguyen DX, Bos PD & Massague J Metastasis: from dissemination to organ-specific colonization. Nat. Rev. Cancer 9, 274–284 (2009). [DOI] [PubMed] [Google Scholar]
  • 100.Lawson DA et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature 526, 131–135 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Malanchi I et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481, 85–89 (2012). [DOI] [PubMed] [Google Scholar]
  • 102.Malladi S et al. Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell 165, 45–60 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Puram SV et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171, 1611–1624.e24 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Fridman WH, Zitvogel L, Sautes-Fridman C & Kroemer G The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol 14, 717–734 (2017). [DOI] [PubMed] [Google Scholar]
  • 105.Hackl H, Charoentong P, Finotello F & Trajanoski Z Computational genomics tools for dissecting tumour–immune cell interactions. Nat. Rev. Genet 17, 441–458 (2016). [DOI] [PubMed] [Google Scholar]
  • 106.Nirschl CJ et al. IFNγ-dependent tissue-immune homeostasis is co-opted in the tumor microenvironment. Cell 170, 127–141.e15 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Spitzer MH et al. Systemic immunity is required for effective cancer immunotherapy. Cell 168, 487–502.e15 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Redmond D, Poran A & Elemento O Single-cell TCRseq: paired recovery of entire T-cell α and β chain transcripts in T-cell receptors from single-cell RNAseq. Genome Med. 8, 80 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Eastburn DJ et al. High-throughput single-cell DNA sequencing of AML tumors with droplet microfluidics. Blood 130, 3965 (2017). [Google Scholar]
  • 110.Zheng GX et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun 8, 14049 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tirosh I et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chung W et al. Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer. Nat. Commun. 8, 15081 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Darmanis S et al. Single-cell RNA-seq analysis of infiltrating neoplastic cells at the migrating front of human glioblastoma. Cell Rep. 21, 1399–1410 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Hayashi T et al. Single-cell full-length total RNA sequencing uncovers dynamics of recursive splicing and enhancer RNAs. Nat. Commun 9, 619 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Klein AM et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Muller S et al. Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome Biol. 18, 234 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Bell JM et al. Chromosome-scale mega-haplotypes enable digital karyotyping of cancer aneuploidy. Nucleic Acids Res. 45, e162 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Savage P et al. A targetable EGFR-dependent tumor-initiating program in breast cancer. Cell Rep. 21, 1140–1149 (2017). [DOI] [PubMed] [Google Scholar]
  • 119.Yuan J & Sims PA An automated microwell platform for large-scale single cell RNA-seq. Sci. Rep. 6, 33883 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Gierahn TM et al. Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput. Nat. Methods 14, 395–398 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Rosenberg AB et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 360, 176–182 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Cao J et al. Comprehensive single-cell transcriptional profiling of a multicellular organism. Science 357, 661–667 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Taniguchi K, Kajiyama T & Kambara H Quantitative analysis of gene expression in a single cell by qPCR. Nat. Methods 6, 503–506 (2009). [DOI] [PubMed] [Google Scholar]
  • 124.Lee JH et al. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat. Protoc 10, 442–458 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Shah S, Lubeck E, Zhou W & Cai L seqFISH accurately detects transcripts in single cells and reveals robust spatial organization in the hippocampus. Neuron 94, 752–758.e1 (2017). [DOI] [PubMed] [Google Scholar]
  • 126.Chen KH, Boettiger AN, Moffitt JR, Wang S & Zhuang X Spatially resolved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Habib N et al. Div-Seq: single-nucleus RNA-seq reveals dynamics of rare adult newborn neurons. Science 353, 925–928 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Lake BB et al. Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain. Science 352, 1586–1590 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Grindberg RV et al. RNA-sequencing from single nuclei. Proc. Natl Acad. Sci. USA 110, 19802–19807 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Faridani OR et al. Single-cell sequencing of the small-RNA transcriptome. Nat. Biotechnol 34, 1264–1266 (2016). [DOI] [PubMed] [Google Scholar]
  • 131.Klein CA et al. Comparative genomic hybridization, loss of heterozygosity, and DNA sequence analysis of single cells. Proc. Natl Acad. Sci. USA 96, 4494–4499 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Lohr JG et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat. Biotechnol 32, 479–484 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Lu S et al. Probing meiotic recombination and aneuploidy of single sperm cells by whole-genome sequencing. Science 338, 1627–1630 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Chen C et al. Single-cell whole-genome analyses by linear amplification via transposon insertion (LIANTI). Science 356, 189–194 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Lodato MA et al. Somatic mutation in single human neurons tracks developmental and transcriptional history. Science 350, 94–98 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Jin W et al. Genome-wide detection of DNase I hypersensitive sites in single cells and FFPE tissue samples. Nature 528, 142–146 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Buenrostro JD et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Cusanovich DA et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Ramani V et al. Massively multiplex single-cell Hi-C. Nat. Methods 14, 263–266 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Rotem A et al. Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state. Nat. Biotechnol 33, 1165–1172 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Smallwood SA et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Han L et al. Bisulfite-independent analysis of CpG island methylation enables genome-scale stratification of single cells. Nucleic Acids Res. 45, e77 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zhu C et al. Single-cell 5-formylcytosine landscapes of mammalian early embryos and ESCs at single-base resolution. Cell Stem Cell 20, 720–731.e5 (2017). [DOI] [PubMed] [Google Scholar]
  • 144.Mooijman D, Dey SS, Boisset JC, Crosetto N & van Oudenaarden A Single-cell 5hmC sequencing reveals chromosome-wide cell-to-cell variability and enables lineage reconstruction. Nat. Biotechnol 34, 852–856 (2016). [DOI] [PubMed] [Google Scholar]
  • 145.Macaulay IC et al. G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nat. Methods 12, 519–522 (2015). [DOI] [PubMed] [Google Scholar]
  • 146.Macaulay IC et al. Separation and parallel sequencing of the genomes and transcriptomes of single cells using G&T-seq. Nat. Protoc 11, 2081–2103 (2016). [DOI] [PubMed] [Google Scholar]
  • 147.Dey SS, Kester L, Spanjaard B, Bienko M & van Oudenaarden A Integrated genome and transcriptome sequencing of the same cell. Nat. Biotechnol 33, 285–289 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Angermueller C et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat. Methods 13, 229–232 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hou Y et al. Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res. 26, 304–319 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Clark SJ et al. scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat. Commun 9, 781 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Stoeckius M et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Peterson VM et al. Multiplexed quantification of proteins and transcripts in single cells. Nat. Biotechnol 35, 936–939 (2017). [DOI] [PubMed] [Google Scholar]
  • 153.Bandura DR et al. Mass cytometry: technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Anal. Chem 81, 6813–6822 (2009). [DOI] [PubMed] [Google Scholar]
  • 154.Ornatsky O et al. Highly multiparametric analysis by mass cytometry. J. Immunol. Methods 361, 1–20 (2010). [DOI] [PubMed] [Google Scholar]
  • 155.Bendall SC et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 332, 687–696 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Goltsev Y et al. Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Preprint at 10.1101/203166 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Lee JH et al. Highly multiplexed subcellular RNA sequencing in situ. Science 343, 1360–1363 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Coskun AF & Cai L Dense transcript profiling in single cells by image correlation decoding. Nat Methods 13, 657–660 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES