You are on page 1of 19

nature reviews genetics https://doi.org/10.

1038/s41576-022-00553-x

Review article Check for updates

Spatial biology of cancer evolution


Zaira Seferbekova , Artem Lomakin
1,4
, Lucy R. Yates
1,2,4 3,5
& Moritz Gerstung 1,5

Abstract Sections

The natural history of cancers can be understood through the lens Introduction

of evolution given that the driving forces of cancer development are Tissue organization controls
mutation and selection of fitter clones. Cancer growth and progression evolution

are spatial processes that involve the breakdown of normal tissue Mapping the cancer
ecosystem
organization, invasion and metastasis. For these reasons, spatial
patterns are an integral part of histological tumour grading and staging Evolution of the cancer
ecosystem
as they measure the progression from normal to malignant disease.
Translational opportunities
Furthermore, tumour cells are part of an ecosystem of tumour cells
and their surrounding tumour microenvironment. A range of new Conclusions and future
perspectives
spatial genomic, transcriptomic and proteomic technologies offers
new avenues for the study of cancer evolution with great molecular
and spatial detail. These methods enable precise characterizations
of the tumour microenvironment, cellular interactions therein and
micro-anatomical structures. In conjunction with spatial genomics,
it emerges that tumours and microenvironments co-evolve, which
helps explain observable patterns of heterogeneity and offers new
routes for therapeutic interventions.

Division of AI in Oncology, German Cancer Research Centre DKFZ, Heidelberg, Germany. 2European Molecular
1

Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK. 3Wellcome Sanger Institute,
Hinxton, UK. 4These authors contributed equally: Zaira Seferbekova, Artem Lomakin. 5These authors jointly
supervised this work: Lucy R. Yates, Moritz Gerstung. e-mail: moritz.gerstung@dkfz.de

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 295


Review article

Introduction transcriptomic and proteomic layers enables a rich characterization


The drivers of evolution are mutation and selection1,2. Mutation of of tumour ecosystems.
somatic cells is an inevitable and persistent consequence of life3–5. In this Review, we focus on the spatial aspects of cancer evolu-
In most normal tissues, mutations accumulate at a steady rate of around tion, summarize how spatial transcriptomics and proteomics reveal a
15–50 per cell per year of life, with only the germline known to exhibit complex landscape of the tumour ecosystem, discuss how interactions
lower rates4. Tumour cells often exhibit an elevated mutation burden with the TME are integral to cancer evolution, and propose potential
due to the variety of mutational processes they have been exposed to clinical applications and future directions. We focus particularly on
during life and, in some cases, due to acquired hypermutation6. The con- the role of tissue micro-anatomy in controlling the rate of evolution
tinuous accumulation of mutations inevitably leads to diversification as well as cellular interactions with the TME within which cancer cells
at the level of single cells both in tumours and normal tissues. evolve.
The second force of evolution is selection, which describes how a
fitter lineage outgrows its relatives. Selection operates at the level of Tissue organization controls evolution
a wide range of heritable phenotypes that derive from genomics and The rate at which malignant clones emerge and spread through
epigenomics. Clones with a selective advantage in the environment to the tissue is controlled by its organization. Levels of organization
which they are exposed will expand, while those with a disadvantage include the micro-anatomical tissue architecture as well as differen-
will tend to disappear (Fig. 1a). Advantageous variants are therefore tiation hierarchies with few stem cells feeding successively larger pools
enriched in genomes of aged normal tissues and cancers7,8. More than of differentiated cells. The breakdown of these protective principles
500 so-called cancer driver genes have been reported to date9–11, which is a key feature of cancer development. However, the resident tissue
are believed to cause different cancer hallmark traits and enabling char- structure, for example, the ductal system in the breast or epithelial
acteristics that involve cell-intrinsic mechanisms as well as interactions layers of the oesophagus, can also influence the rate of progression
with the tumour microenvironment (TME)12,13. at pre-invasive stages. The transformation ends with overwhelming
While mutation continuously generates subclonal diversity at the metastatic disease.
level of single cells, it is well documented that tumours are mosaics
of subclones each comprising hundreds of thousands of cells that have Normal tissue organization suppresses evolution
arisen from a shared ancestor14–18 (Fig. 1a); these patterns have been It has long been hypothesized that somatic tissues are structured to
further confirmed by single-cell studies19–24. The mechanisms of these suppress the rate of somatic evolution1. These ideas are based on fun-
expansions remain debated. It has been shown that cancer subclones damental theoretical considerations that the rate of evolution depends
exhibit signs of positive selection and driver gene mutations that can on population structure. Generally, the rate of evolution is determined
also be found clonally18. However, it is also conceivable that subclones by the rate at which new mutations are generated (a product of the
branching at early stages of tumour development reach considerable number of cells and the mutation rate per cell), the probability that
size without a selective advantage as they continue to expand with the new mutants sweep through the population48 and the time it takes to
same tumour, a phenomenon termed neutral evolution25. do so49,50. In addition to suppressing the mutation rate, differentiation
Subclonal mosaicism is often found to be spatially variegated hierarchies can reduce the chance of emergence and slow the spread
as demonstrated in diverse studies based on tumour macrodissec- of mutant lineages. The haematopoietic system constitutes a basic
tion26–33 and laser-capture microdissection21,34–40 (Fig. 1b). These clonal example51: its 1013 cells derive from around 100,000 haematopoietic
variegation patterns were shown to be accompanied by differential stem cells52,53, which themselves divide slowly and produce a range
gene expression in multiple cancer types, including renal27, colorec- of faster-dividing differentiated cells54. This hierarchy reduces the
tal41, lung42 and breast cancer43, using spatially resolved genomic and rate of evolution, as only mutations in the comparably small number
transcriptomic analyses, indicating the presence of subclone-specific of stem cells and potentially the first progenitors, or mutations that
gene expression and associations between certain subclones and lead to a differentiation blockage, will remain in the population. The
characteristic TMEs. colon extends these organizational principles by its compartmen-
The exact mechanisms creating these phenomena are not fully talization into micro-anatomical crypts; in each crypt, a similar dif-
understood, largely owing to a dearth of methods that can molecularly ferentiation hierarchy operates in which a very small number of stem
characterize whole tumour sections with spatial resolution. A variety cells replenishes the colonic epithelium54. As differentiating cells are
of non-exclusive explanations have been proposed. A rapid expansion fated to die within ~5 days, only mutations arising in the stem cells per-
could lead to a fragmentation of closely related clones44 even though sist1,55,56. Furthermore, micro-anatomy largely constrains the fixation of
it has also been argued that cell dispersal is a crucial element shaping stem-cell mutations to individual crypts (Fig. 2a).
tumour structure45,46. However, it is also possible that tissue micro- Systematic studies of normal tissues confirm that, in tissues with
anatomy, such as the ductal system of the breast, provides a template a more complex architecture, clones typically remain small in size
for clonal segregation (discussed further in the section ‘Tissue organi- and develop independently in comparison to less structured tissues
zation controls evolution’). Lastly, it is conceivable that locally distinct where clonal expansions tend to be larger5. In a recent investigation of
microenvironments or niches favour the selection of particular clones. 517 normal colonic crypts, only 1% harboured cancer driver gene muta-
Various new spatially resolved genomic, transcriptomic and pro- tions, and those that were mutated were confined to single crypts57.
teomic technologies offer novel insights and answers to these unre- Similar observations have been made for glandular tissues such as the
solved questions (Fig. 1b). The technological aspects enabling the endometrium58: although most of the glands harboured driver muta-
nascent field of spatial cancer biology are summarized in Box 1 and have tions, they were limited to a single mutation or a few neighbouring
been extensively reviewed elsewhere47. Individual technologies differ mutations. Conversely, it is well established that the haematopoietic
in the multiplexity of their readouts and their spatial resolution as well system harbours mutant lineages whose frequencies can reach up
as in their sensitivity and available field of view. Combining genomic, to 100%59,60. Other epithelial tissues, including the skin, oesophagus

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 296


Review article

a Diagnosis

Subclonal frequencies
Cancer
founder
Zygote cell

Driver mutations

500 mutations
Evolutionary time

b Spatial biology Single cell


Genomics

Spatial subclonal Subclonal Single-cell genotypes,


variegation, early interfaces latest evolution
subclonal evolution

Anatomy

Cancer site Organ anatomy Micro-anatomy Cytomorphology

Microenvironment

Spatially segregated Interactions Cell typing


microenvironments between cells
1 cm 1–0.1 mm 10 μm
Fig. 1 | Spatial biology of cancer evolution. a, Somatic evolution and cancer the tumour clone. b, Spatial technologies add new layers of information. A tumour
development. Human cells accumulate mutations throughout their lifetime. is a mosaic of subclones with distinct micro-anatomy and tumour microenvironment.
A subset of these, so-called driver mutations, are associated with neoplastic Spatial analyses offer new avenues for charting and understanding the biology and
transformation. Ongoing mutation and selection cause subclonal divergence within evolution of the cancer ecosystem at scales ranging from a single cell to whole tissue.

or bladder, have an intermediate degree of tissue structure and are the haematopoietic system because the dynamics are confined to
patchworks of microscopic clones4,61–63 (Fig. 2a). In such tissues, the two dimensions, which reduces the interfaces at which a fitter variant
fixation probability is high but the time to fixation is greater than in may replace less-fit competitors. Of note, not all mutations causing

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 297


Review article

Box 1

Enabling technologies
A range of spatially resolved approaches for mapping DNA, RNA and tissue regions is coupled with later bulk sequencing of DNA or
proteins has been developed47,215. Higher spatial resolution typically RNA. Laser-capture microdissection (LCM)216 allows the profiling
implies smaller molecular quantities (figure, part a). Other important of even smaller regions of interest using laser ablation. LCM is very
parameters are the field of view, which ranges from millimetres versatile as it can be combined with high throughput sequencing,
squared to centimetres squared, and method sensitivity. including whole-genome sequencing, and single-cell RNA or DNA
sequencing21,217–219.
Macrodissection and microdissection
The foundations were laid by macrodissection followed by multi-omic Spatial molecular barcodes
profiling but new technologies offer a microscopic and single-cell This set of methods utilizes probes with unique barcode sequences
resolution (figure, part b). In multi-region sequencing27,30, the collection to capture molecules and then map them to specific spatial locations
of several biopsy samples from multiple geographically separated (figure, part c). So far, these technologies have mainly focused on

a MIBI, ImmunoSABER MIBI-TOF Stereo-seq b Macrodissection and microdissection


DNA, epigenetics
10–7
CODEX RNA
corrFISH Seq-Scope
epiMERFISH Protein
Base Scope DVP, DNA-MERFISH
RNAScope BaSISS STARmap, MERFISH DNA mutations via RNA
Resolution, m

10–6 FISSEQ
osmFISH ISS seqFISH+ HDST
IMC smFISH Subcellular
HybISS TSCS, resolution
pciSeq slide-DNA seq
10–5 Typical size of
a human cell
GeoMX Visium Visium HD,
Slide-seq, Supercellular
CosMX resolution
10–4 ST DBiT-seq Multi-regional sequencing LCM based
1 10 102 103 104 105 106 107 108 109
Plexity
Whole transcriptome Whole genome

c Molecular barcode-based methods d Image-based methods e Mass spectrometry

Spot based Beads based FISH based Multiplexed IHC

Type of assay Targeted Untargeted (whole) Type of molecule detected RNA DNA Protein

BaSISS, base-specific ISS; CODEX, co-detection by indexing; corrFISH, correlation FISH; DBiT-seq, deterministic barcoding in tissue for
spatial omics sequencing; epiMERFISH, epigenetic MERFISH; FISSEQ, fluorescent ISS; HDST, high-definition spatial transcriptomics; hybISS,
hybridization-based ISS; ImmunoSABER, immunostaining with signal amplification by exchange reaction; MIBI-TOF, MIBI by time of flight;
osmFISH, ouroboros smFISH, pciSeq, probabilistic cell typing by ISS; smFISH, single-molecule FISH; ST, spatial transcriptomics; STARmap,
spatially resolved transcript amplicon readout mapping; Stereo-seq, spatial enhanced resolution omics sequencing; TSCS, topographic
single-cell sequencing; Visium HD, Visium high definition.

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 298


Review article

(continued from previous page)


transcriptomic profiling with spatial transcriptomics125, 10x Visium protocols43. Immunohistochemistry (IHC) or immunofluorescence
and Slide-seq220,221 being the most popular protocols among many uses antibodies to localize proteins or other molecules. Highly
others222–224. Spatially resolved DNA sequence capture37 has enabled multiplexed spatial proteomics with dozens of targets can be
spatially resolved genome-wide copy number calling along with achieved using DNA barcodes245,246, multiplex immunofluorescence
several new methods for spatial epigenomic profiling225,226. Although (CyCIF)247 and other approaches248–250. Multi-omic approaches, such
these methods support unbiased discovery and result in data with as GeoMx or CosMx251,252, allow researchers to assess multiple layers
high molecular content, the resolution is limited by the diameter of of biological information within one cell simultaneously. Image-based
each spot and currently does not allow profiling at the single-cell technologies provide high-resolution tissue profiling but require
level or point mutation detection. Further improvements in resolution a pre-defined panel of targets.
are currently being developed222,224,227.
Mass spectrometry
Image-based technologies A further class of methods combines metal-conjugated antibodies
This group of methods involves in situ hybridization of fluorescently and mass spectrometry (figure, part e). Spatial resolution is provided
labelled probes (FISH) to nucleic acids of interest to identify their by spatial laser ablation (imaging mass cytometry (IMC))253 or ion
location with single-cell or subcellular resolution (figure, part d). beams (multiplexed ion beam imaging (MIBI))254. These approaches
A variety of multiplexed extensions228–238 exists, including multiplexed detect proteins and smaller molecules, such as lipids, metabolites,
error-robust FISH (MERFISH)239,240, sequential FISH (seqFISH)241, and drugs, in single cells253, as reviewed in ref.255. Deep Visual
RNAscope242 and in situ sequencing (ISS)243 for spatial transcriptomic Proteomics (DVP) combines artificial intelligence-driven LCM
profiling. The latter two protocols have been further adapted to with mass spectrometry to map protein abundances onto tissue
permit mutation detection using the Base Scope244 and BaSISS slides256.

a selective advantage are associated with malignant transformation. facilitated by the microenvironment (see the section ‘Mapping the
For example, in the skin and oesophagus, NOTCH1 mutant clones cancer ecosystem’).
expand and suppress malignant evolution while maintaining normal The consequence of this breakdown is likely the acceleration of
tissue function61,62,64–66. malignant evolution as the fixation probability and the rate of fixation
are higher in unstructured populations with a greater degree of mix-
Breakdown of evolutionarily confining tissue architectures in ing. Several theoretical investigations have demonstrated that spatial
cancer constraints can control the mode of tumour evolution73,74. Another
The fundamental observation that dedifferentiation and the loss of consequence is that different selective pressures apply, which may be
normal tissue architectures are correlated with cancer aggressiveness one cause of the characteristic sequences of cancer progression such
was made a century ago and still forms part of modern histopatho- as the classic APC-KRAS-TP53 model for colorectal carcinoma75,76 and
logical grading systems that direct cancer treatment67–70. In these sys- the four stages of TP53 inactivation in the progression of pancreatic
tems, higher grades correspond to more aggressive tumours, typically ductal adenocarcinoma from a pre-malignant to malignant state77.
composed of less differentiated cells. In this light, a pan-cancer analysis revealed that the portfolio of late
The breakdown of such evolutionarily confining tissue structures and subclonal driver gene mutations tends to be more diverse than
is a hallmark of cancer. It is especially noticeable among carcinomas, the set of early alterations76.
which derive from epithelial tissues (Fig. 2b). At the earliest stages
of progression, so-called carcinomas in situ are still restrained to the Metastatic dissemination
resident tissue structures, such as the duct of the breast. Similar to The ability of cancer cells to leave a primary tumour site, seed in distant
the considerations of normal tissues, the resident tissue structures may organs and generate metastatic deposits presents landmark stages
delay clonal sweeps. This tissue-based restraint was observed in a case that are strongly predictive of poor clinical outcomes78. This inherently
of breast cancer, where spatial genomic analysis revealed two distinct spatial process involves, upon tissue invasion, intravasation of single or
subclones occupying a checkerboard pattern of micro-anatomical small aggregates of cancer cells, circulation through the blood system,
niches within the ductal system43. arrest in the capillaries of the target organ, extravasation, and prolifera-
During invasion, tissue architectures are lost, probably via different tion79 (Fig. 2b). These steps thus involve a range of micro-anatomical
mechanisms in each tissue type, but a frequent property of carcinomas bottlenecks and possible differential selective pressures in different
is the epithelial-to-mesenchymal transition (EMT). EMT enables epi- stages and target organs.
thelial cells to lose their polarity and escape from the confinement of Several multi-region genomic sequencing studies have investi-
planar epithelial tissue organization (as reviewed in ref.71). In colorectal gated the phylogenetic relationships of primary tumours and metas-
adenocarcinoma, EMT can be induced via the WNT signalling pathway tases from the same patient27,28,30,32,80–85. These data show that most
by loss-of-function mutations in APC or somatic mutations of CTNNB1 metastases have diverged from the primary tumour clone typically
inhibiting its degradation72. Both alterations are believed to be a key shortly before or after its most recent common ancestor, which is
step in the development of adenomas, the neoplastic precursor to often dated 1–5 years before diagnosis83,84,86,87. The origin of meta­
invasive adenocarcinoma. A further step of invasion is the breakdown stasis appeared to be mostly clonal88–90 even though, in advanced
and remodelling of the extracellular matrix (ECM), thought to also be disease, intermixed subclones have been found in multiple metastases,

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 299


Review article
a Normal tissue organization
Population structure constraints

Shedding Colonic crypt

Differentiated Wild-type
cell mutates differentiated
cell Mutations in
Mutations in
differentiated stem cells are
cells are lost maintained and
as they get may transform the
replaced by crypt if all other
Differentiation wild-type cells stem cells are
also replaced
Stem cell
mutates
Wild-type stem cell

Physical architecture constraints


Colon Skin Blood

Subclones are confined within Clones and subclones are HSC


single crypts mosaically distributed

Crypts Planar epithelium Well mixed subclones

Slow evolution, small subclones Fast evolution, large subclones


More constrained Less constrained

b Carcinogenesis Tumour cells from different subclones


Without metastatic With metastatic
Normal tissue potential potential

Normal cells

Basement membrane
ECM Stroma
Intravasation of subclones with metastatic
potential into a lymphatic vessel Metastases in lymph nodes

Primary tumour
Polyclonal and
metastasis-to- Monoclonal
metastasis seeding seeding

Basement
membrane
breach Invasion into Intravasation of subclones with metastatic
surrounding Metastases in other organs
potential into a blood vessel
normal tissue
Vascularization
Possible recolonization of the primary site

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 300


Review article
Fig. 2 | Spatial cancer evolution. a, Normal tissue structure suppresses somatic basement membrane is followed by tumour invasion into the neighbouring
evolution. Top: in addition to physical constraints, the differentiation hierarchy stroma, lymphangiogenesis and angiogenesis. Through blood and lymphatic
in the colonic crypt limits the number of cells with fixation potential, which vessels, cancer cells with metastatic potential can transfer to lymph nodes
reduces the chances of malignant transformation. Bottom: highly structured and distant organs, giving rise to metastases at these new sites. Metastases
tissues, such as the colon, slow evolution by constraining the spread of clones. containing cancer cells of different lineages within the same lesion are
Physically unstructured tissues, such as the haematopoietic system, exhibit the called polyclonal, whereas metastases containing cancer cells of the same
fastest rate of evolution and clones, which replace the entire organ. b, Spatial lineage are called monoclonal. Metastatic subclones may later return to the
steps of cancer progression. Normal tissue structure breaks down during tumour primary site and recolonize it. ECM, extracellular matrix; HSC, haematopoietic
progression. This process is exemplified here by a carcinoma: breach of the stem cell.

suggesting spread from one metastasis to another84,86,91. Pan-cancer immune cells, such as macrophages, myeloid-derived suppressor
analyses of metastatic cancers have revealed that metastases exhibit cells and granulocytes, can actively promote tumour progression
fewer intra-metastatic subclones than primary tumours92–94, which may through two-way interactions with the tumour, other immune cells
in part be a consequence of sampling fewer cells (often core needle biop- and the stroma117–119. The stroma also plays an active role in shaping
sies). From the perspective of driver gene mutations, it has been argued the functional immune response to cancer. A variety of studies have
that treatment-naive metastasis has broadly the same driver gene muta- demonstrated that cancer-associated fibroblasts and myofibroblasts
tions as the matched primary95. Characteristics of metastatic cancer typically change shape, become more proliferative, undergo diverse
include high levels of genomic instability, TP53 mutations and whole- transcriptional and metabolic changes, engage in angiogenesis, and
genome duplications, shared by both the primary tumour and meta­ interact with leukocytes and tumour cells while the resulting ECM
stases92,94. However, adjuvant therapies can select for specific mutations, changes in chemical composition and mechanical properties120–124.
for example, prostate and breast cancers treated with endocrine depri-
vation therapies are enriched for resistance mutations in AR and ESR1, Cellular interactions and neighbourhoods
respectively93,94. Although scRNA-seq studies have generated comprehensive catalogues
Although it emerges that tissue architecture has a profound of detailed TME cell types and states, these data only allow circumstan-
influence on somatic evolution, it is important to realize that cellular tial insights into how cells interact. Spatial transcriptomic and prot-
interactions with normal cells are also likely to affect the fate of cancer eomic technologies are beginning to show that the cells constituting
clones. The composition of the TME and its role in cancer evolution will the cancer ecosystem self-organize into micro-anatomical neighbour-
be discussed in the next two sections. hoods, which could provide insights into the frequency and nature
of different cellular interactions (Fig. 3b–d and Box 2). The majority of
Mapping the cancer ecosystem spatial omic cancer studies to date have used spatial transcriptomics
The cellular composition of tumour masses is highly variable, with the as commercialized by 10x Visium (Box 1). These studies reveal spatially
neoplastic lineage contributing as few as 10% of cells in some cases96. structured TMEs in breast125–127, prostate128, pancreatic129, colorectal130,
The remaining cells derive from the TME, which is usually dominated skin131, brain132, liver133, bladder134 and other cancer types135,136. The ability
by immune and stromal cells in addition to vessels, adipocytes and peri- to use whole-transcriptome readouts also helped establish the presence
cytes. Together, these cells and other physical structures, such as ECM and broad co-localization of rare cell types (Fig. 3b). However, the cur-
and chemical gradients, form an environment termed the cancer eco- rent super-cellular resolution (55 µm, ~3–10 cells) requires informatic
system97. Over the past decade, single-cell RNA sequencing (scRNA-seq) deconvolution of the measured signals in each spot.
analyses have catalogued the cellular composition of the cancer eco- Highly multiplexed antibody detection approaches have been
system98–105. More recently, spatial transcriptomic and proteomic tech- used to generate single-cell resolution readouts of the spatial distribu-
nologies have started to uncover how these diverse cell types organize tion of the major cell types in breast cancer, colon cancer and mela-
into communication interfaces and niches106,107 (Boxes 1 and 2). noma, amongst others137–140. These approaches permit the detection
of microscopic neighbourhoods with distinct composition of tumour,
Cellular composition of the TME immune, stromal and vascular cells. In two studies that analysed hun-
The TME has been studied extensively and, therefore, we herein only dreds of breast cancers by applying imaging mass cytometry (Box 1)
present a high-level summary of major cell types. Inflammation is a to tissue microarrays, several recurrent neighbourhoods were identi-
hallmark of cancer108 and reflects the interplay of antitumorigenic and fied137,138 (Fig. 3c). The biological feasibility of the identified neighbour-
pro-tumorigenic immune responses (Fig. 3a). The antitumour response hoods was provided by their relationship with well-defined clinical
is primarily generated by cytotoxic CD8+ T lymphocytes that recognize subtypes and survival outcomes. For example, consistent with a failing
antigens presented on the tumour cell surface109,110. The success of this antitumorigenic response, ‘suppressed expansion structures’ con-
response is dependent on priming and prior education by professional tained many dysfunctional T and Treg cells and were enriched in breast
antigen-presenting cells (dendritic cells) and is facilitated by CD4+ cancers with poor clinical outcomes. By contrast, a ‘vascular stroma’
T helper cells111,112. B cells have diverse functions, including antigen signature was enriched in a very favourable outcome subgroup of hor-
presentation, antibody production and cytokine-mediated cytolytic mone receptor-positive breast cancers (Fig. 3c). The spatial patterns
effects113. In many cancers, B cells form dense aggregates with T cells formed by individual cell types were also examined. In breast cancers,
and dendritic cells to form tertiary lymphoid structures114 that are whereas T cells were diffusely scattered, B cells tended to form aggre-
generally associated with favourable cancer outcomes and responses gates, were in multiple neighbourhood types, and were associated with
to immune-checkpoint blockade115. Natural killer cells might have a disparate clinical subtypes and clinical outcomes, suggesting that B cell
particularly important role in anticancer immunity due to their ability function might be dependent on the precise TME context141 (Fig. 3c).
to recognize stressed cells irrespective of neoantigen presentation116. However, some tumour phenotypes, such as cell proliferation, can be
By contrast, CD4+ regulatory T (Treg) cells can suppress the immune shaped by further long-range interactions142.
response through cytokine release (for example, IL-10) or by modula- The proximity of cells does not necessarily equate to an interac-
tion of antigen presentation functions. In certain situations, some tion. To address this limitation, a combination of CyCIF (Box 1) and

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 301


Review article

Box 2

Spatial omic analysis


A typical tumour cross-section covers several centimetres of tissue be picked up by various topic and graph-based models. Mechanistic
containing up to 1,000,000 cells. Spatial omic methods characterize models attempt to explain expression plasticity mediated by cell–cell
the molecular features of these cells with a resolution as fine as interactions and signalling at the cell interface107.
~0.2 μm, depending on the method. The wealth and detail of data
bring a series of data analysis challenges257 (see the figure). Spatial lineage tracing
Spatial RNA or DNA sequencing technologies provide wide but low
Raw signal analysis genomic coverage, which limits lineage tracing to copy number alter­
In the case of molecular barcode-based technologies (Box 1), primary ations and a few point mutations37,279,280. Current protocols also have
steps of raw data processing include aligning and counting reads or a relatively low spatial resolution (Box 1). Microscopy methods target
molecular barcodes per genomic region of interest, with sequenc- specific point mutations and require prior knowledge of the clonal struc-
ing barcodes defining the spatial sector. Image-based methods, by ture. Yet, they achieve a higher spatial resolution to map a more fine-
contrast, utilize fluorescent signals, often measured across multiple grained clonal structure across wide fields of view43. The incremental
fluorescence channels and cycles. These signals must be detected, nature of branching evolution also provides opportunities to associate
quantified and decoded to their corresponding target molecule, the genetic changes on each branch with phenotypic changes.
which can be defined by a specific combination
of colours across cycles. For whole-slide imaging, Integration H&E slide Spatial analysis
Cells, morphology
fields of view need to be stitched together into micro-anatomy
one tissue image258. Background fluorescence,

Time
blur and signal crowding can negatively affect
the ability to decode signals243,259–262. Technolo-
gies with a super-cellular resolution require cell Space
decomposition algorithms, where known cell types
usually obtained from the single-cell data serve Genes with Genes with
Subclone-specific
as a reference43,129,263,264. Signals with a subcellular histology and significantly significantly
resolution are spatially aggregated either by clus- micro-anatomy higher expression higher expression
Genomics in red subclone in green subclone
tering265–268 or based on cell segmentation derived Mutations and
from nuclear DAPI signals, membrane markers or subclones
–log (P value)

haematoxylin and eosin (H&E) staining235,266,269,270.

Data integration
Single-cell resolved data can be mapped271 to
deconvolve larger spots or to annotate known cell 0
types. Single-cell whole transcriptomes or multi- Subclone-specific Log fold change
expression and
omics can be further mapped into space, leverag- tumour
Stromal cell type

ing a smaller common set of reference genes that microenvironment


Transcriptomics
are interrogated spatially270,272. Another option is
abundance

Expression
to integrate multiple spatially resolved modalities
from consecutive tissue sections. Alignment of
distorted sections and 3D reconstruction of a tissue
block can be done using image registration algo- Red Green
rithms273. Multimodal characterizations of micro- subclone subclone
scopically matched tissue areas, including spatial Cell neighbourhoods
transcriptomics, proteomics and histopathology, Subclone-specific Cellular neighbourhood
are also possible43,274. proteins
Proteomics
Spatial statistics Protein levels
Spatial differential expression models enable the
description of novel cell states showing distinct
spatial patterns that are difficult to recover from
single-cell data275,276. A group of cells may appear
enriched in a recurrent way forming distinct
neighbourhoods137,139,277,278, a pattern which could Tumour lesion

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 302


Review article

Glossary

Adjuvant therapies Immune evasion Mutation Subclones


Cancer treatments given after primary The process by which tumour cells Changes in the DNA sequence that Further clones emerging within a
therapy (particularly following surgical develop several mechanisms that are inherited across cell generations. tumour from one founder clone.
resection of a tumour) to reduce the risk help them to continue to grow and Sources of mutation are erroneous Subclonal mutations are limited to
of relapse. expand by escaping immune control. replication, biochemical alterations of a fraction of cancer cells and occur
Immune evasion is thought to be DNA and failed DNA repair. To date, all during tumour expansion.
Carcinomas in situ typically preceded by two other phases: cells within the human body are found
Neoplastic expansions of epithelial cells elimination, when the immune system to accumulate mutations over their Tissue architecture
that are confined to the normal tissue recognizes and eliminates tumour cells, lifetime. The micro-anatomical spatial
structure in which they arose, without and equilibrium, when the pressure organization of the tissue. Typical
invasion of the adjacent stroma. from the immune system stalls tumour Negative selection examples are layered epithelial tissues,
growth and expansion, and negative The removal of deleterious variants from glands and crypts. Tissue architecture
Clones selection takes place. the population. can be combined with other means
A clone is a population of cells that of tissue organization such as
derive from a common ancestor. Invasion Neoantigen differentiation hierarchies.
Cancers are found to be clones deriving The process of penetration and spread A tumour-specific antigen that is the
from a single cell that expanded during of cancer cells into the neighbouring result of a somatic coding mutation Tumour ecosystems
the lifetime of a host. Clonal alterations normal tissue. In the case of carcinomas, in the corresponding part of a gene. The collective set of heterogeneous
are mutations that are present in all cells invasion involves a breach of the Antigens are protein fragments cells in the vicinity of a tumour
of a cancer because their occurrence basement membrane. presented on the cell surface by comprising cancer cells and the TME.
preceded the expansion of the tumour. the human leukocyte antigen (HLA)
Metastasis complex and recognized by different Tumour microenvironment
Immune-checkpoint blockade A process of cancer spreading from the cells of the immune system. (TME). A combination of non-tumour
A type of anticancer immunotherapy primary disease site to lymph nodes or cells, such as stromal and immune
that blocks checkpoint proteins on other organs, resulting in the formation Positive selection cells, vessels, metabolites, signalling
T cells or their targets on cancer cells of metastatic deposits known as The spread of advantageous alleles molecules, and other extracellular
and promotes an immune response and metastases. within a population. components among which tumour
killing of the tumour. cells exist.
Most recent common Selection
ancestor In evolution, natural selection denotes
In cancer evolution, the most recent the process of survival and reproduction
founder cell from which all other of the fittest organism within a given
tumour cells have directly descended. environment.

high-resolution three-dimensional optical deconvolution was used to digital histopathology143,144. In some cases, histological patterns have
study the physical features that might be associated with cellular inter- been found to be indicative even of distinct subclonal alterations43,145,146.
actions139. In melanoma tissues, probable interactions were identified It is well established that tumours exhibit characteristic histological
amongst approximately 20% of immune cells at the tumour–stroma structures indicative of their natural history (Fig. 3e). Many cancers
interface (Fig. 3d). These interactions were often complex, involving exhibit adjacent areas of carcinoma in situ, which are considered to be
three or more cells (for example, a melanoma cell contacting two CD8+ precursor lesions. This phenomenon is well established in melanoma139
cytotoxic T lymphocytes and one CD4+ Treg cell) with associated cell sur- and many adenocarcinomas: in breast cancer up to two-thirds of cases
face molecular polarization and receptor–ligand interaction. In some have histologically distinct areas of ductal carcinoma in situ147, around
cases, cellular processes from macrophages extended more than 10 μm 15% of colorectal adenocarcinomas exhibit adjacent adenomas148, and
to make contact with other cells. Standard fine sections would miss ~45% of oesophageal adenocarcinomas show detectable areas of its
many of these interactions, whereas basic proximity measurements precursor lesion Barrett oesophagus149. Interestingly, the phylogenetic
would tend to overestimate the nature of interactions. As discussed and histological relationships between precursor lesions and invasive
in Box 2, although these analyses remain technically challenging and cancer are not always aligned with patterns indicative of branching150,151
have low throughput, they demonstrate the unique potential afforded or multiclonal invasion21. Therefore, spatial genomic and transcrip-
by spatial omics for the mapping of functional cellular interactions in tomic analyses using histologically informed dissection allow for the
tissue space. exploration of mechanisms and phenotypes of disease progression in
greater detail, revealing subclone and histology-specific changes43.
Tumour histology and macrostructure At the scale of millimetres, cancers are often described in terms of a
The histological appearance of tumour cells and their micro-anatomical tumour core that is separated from the surrounding stroma by an inva-
structures also reflect the underlying genomic alterations as revealed sive margin (Fig. 3e). Transcriptional diversity has been reported across
by two recent pan-cancer analyses using artificial intelligence-based these landscapes128, with hypoxia and EMT-like signatures localizing

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 303


Review article
a Cytolysis of tumour Antitumour immunity ECM Single-cell data provide high molecular
cells by NK or CTL Oxygen inhibition by Treg cells detail of TME components but lack
spatial context
ECM remodelling by
CAFs facilitates tumour
cell migration

Tumour-promoting M2
macrophages are recruited
to hypoxic areas and release
Immune activation immuno-suppressive cytokines
by M1 macrophages
via cytokine release Capillary
Single-cell RNA sequencing:
High-resolution TME composition

Tumour cells CAFs


Immune activation of Endothelial cells T cells
CD4+ cell by B cell Dendritic cells B cells
Dendritic cell CD4+ T cell Macrophages
Antigen presentation to neighbouring T cells

Tumour inhibiting Tumour promoting

b Co-localization c Cellular neighbourhoods and structures


Cell type A Cell type B
Suppressed
expansion structure

Putative
interaction Blood
vessel
% % Vascular
0.3 0.4 stroma
Tumour lesion

Visium spots 0 0
B cell
aggregates

Tertiary lymphoid
Diffuse structure
d Interfaces T cells

Ligand X
Receptor Y Denritic
cell
DNA
Immunofluorescence reveals receptor–ligand
interactions such as PD1–PDL1 B cell T cell

e Histology Pathological annotation Clonal map

Carcinoma Invasive
in situ front with
EMT
Invasive Immune
cancer infiltration Immune-
with large excluded Genetically
cells area distinct
Tumour
lesion subclones
Hypoxic
Invasive area with
cancer necrosis
with small
cells Stroma

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 304


Review article
Fig. 3 | Mapping the cancer ecosystem. a, Cellular composition of the tumour proteomic studies detect clinically relevant neighbourhoods of molecularly
microenvironment (TME). The TME is composed of different cell types, involved related cell types137. d, Cellular interfaces. Subcellular resolution imaging reveals
in tumour-suppressing (on the left) and tumour-promoting (on the right) receptor–ligand localization at cellular interfaces139. e, Tumour macrostructure.
interactions with cancer cells. While TME composition can be captured by single- Different tumour regions can be identified in some tumours based on histological
cell omic approaches (such as single-cell RNA sequencing), its spatial context is staining. Such histopathological findings can be supplemented and underpinned
lost as a consequence of tissue dissociation. b, Cellular interactions. Spatial by detailed molecular maps. CAFs, cancer-associated fibroblasts; CTL, cytotoxic
co-localization and concomitant expression of receptors and ligands are T lymphocyte; ECM, extracellular matrix; EMT, epithelial-to-mesenchymal
indications of cellular interactions. c, Cellular neighbourhoods. Spatially resolved transition; NK, natural killer; Treg, regulatory T cell.

to the core and invasive margin, respectively101,152. The core–stroma Furthermore, selective pressures of the immune system determine
distinction is fundamental to common classifications of immune infil- prognosis and immune evasion is found in relapses. It has been reported
trates that correlate with cancer subtype, clinical outcome and therapy that strongly immunogenic neoantigens are prognostically favour-
response153–155. At one end of the immune infiltrate continuum are able in pancreatic cancer166 but are preferentially lost at recurrence167.
inflamed tumours, where plentiful immune cells infiltrate the tumour In melanoma, mutations of JAK1 or JAK2, which inhibit cytolysis, and in
core, at the other end are immune deserts that are devoid of infiltrates, B2M, which reduce antigen presentation, are often found in samples
and between these are immune-excluded tumours where immune cells that acquired resistance to immune-checkpoint inhibitors168,169.
are largely restricted to the neighbouring stroma156. However, recent
spatial studies in pancreatic and breast cancer reveal that these immune Maps of cancer–TME interactions
characteristics can be highly localized and coexist in different parts Establishing associations between genomic alterations and the micro-
of the same tumour137,157,158. A challenge will therefore be to decipher environment can be challenging due to the large genomic heterogene-
how localized variation in immune response shapes the evolutionary ity between primary cancers. Conceptually, spatial genomics offers new
outcome of the tumour. ways of analysing such interactions at high spatial resolution and also
within the same tumour. For intratumour diversity, the observations
Evolution of the cancer ecosystem occur in an isogenic background and the set of genomic differences
The cancer ecosystem detailed in the previous section changes the between subclones is small compared to the large intertumour diver-
dynamics of cancer evolution as tumour cells face selection pressures sity (Fig. 4b). A proof of concept has been established in breast cancer,
that maximize growth-promoting and minimize growth-inhibiting where various degrees of immune infiltration were found at the level
interactions. Foremost are cell–cell and other local interactions with of cancer subclones43. Interestingly, such observations could also be
the immune system but also with fibroblasts, vasculature and even linked to the stage of invasion, with some TME regions defined by the
neurons, which can lead to the selection of specific genomic alterations subclonal lineage and others mostly by histological progression from
(Fig. 4a). The fact that the TME is itself spatially heterogeneous suggests carcinoma in situ to invasive cancer. Direct experimental evidence was
that tumour cells experience localized variations in selective pressure. provided by Perturb-map, a mouse in vivo spatial CRISPR screen, which
revealed that specific gene knockouts lead to characteristic changes in
Evolution of immune evasion the immune microenvironment. For example, it was shown that T cell
Various parts of the immune system constrain tumour growth. The infiltration is increased by Socs1 knockout in the tumour but decreased
observed range of associations between specific genetic alterations by Tgfbr2 knockout170 (Fig. 4b).
and the immune system cells of the TME159,160 illustrates the presence of Further cell types beyond immune cells have been associ-
selective pressures on cancer evolution. Neoantigens, which derive from ated with cancer evolution. Fibroblasts are an extremely versatile
coding mutations in cancer genomes, have been reported to elicit T cell group of cells that perform various roles that are exploited during
reactivity in lung cancer161. In tumours with greater cytolytic activity, tumour growth. For example, cancer-associated fibroblasts can be
loss of neoantigen presentation capabilities was more frequent, which is educated by cancer cells with gain-of-function mutations in TP53 to cre-
indicative of greater selective pressures159,162. Other signs of selection for ate a pro-tumorigenic microenvironment171. Furthermore, fibroblasts
immune evasion include mutations in CASP8, which serve as a potential can amplify tumour-intrinsic oncogenic signalling in KRASG12D-mutant
way to resist apoptosis induced by FASL159. Furthermore, gains of immu- pancreatic adenocarcinoma172. Spatial genomics adds to these obser-
nosuppressors, such as CD274 (also known as PDL1), are widespread vations. In breast cancer, imaging mass cytometry (Box 1) revealed
among Hodgkin lymphomas and are detected in ~0.7% of solid tumours163. enrichment of different fibroblast and myofibroblast populations
The signs of immune evasion increase during later stages in cancer in TP53-mutant and genomically unstable cancers173; however, the
evolution. No evidence of negative selection was reported in diploid nature of those associations is unclear. The aforementioned Perturb-
genomes of normal tissues and among clonal alterations in primary map screening revealed that Tgfbr2 knockout created a fibro-muci-
cancers, indicating that there is no depletion of coding mutations nous stroma characterized by TGFβ-activated fibroblasts and T cell
during normal evolution8,164. Yet, the signs of selection for immune exclusion170.
evasion become stronger after clonal expansion at the subclonal level. Another cell-extrinsic and environmental factor influencing can-
For example, immune evasion via deletions of human leukocyte antigen cer evolution is hypoxia, the often localized depletion of oxygen in
(HLA) class I alleles, predominantly subclonally and in metastases, has tumours; it is well established as a marker of poor outcomes174 and
been described in lung cancer165 and multiple other cancer types33 has been shown to select for certain mutations such as TP53 (ref.175).
(Fig. 4a). This mode of evolution is plausible as it may require a certain A recent study combining scRNA-seq and immunofluorescence found
tumour size for the immune system to recognize its neoantigens; at this that hypoxic microniches contain quiescent cancer cells resistant to
stage, however, a mutation enabling escape from these new selective T cell attack176.
pressures arises in a single cell of the tumour and seeds the growth Lastly, spatial genomics may also shed light on how different can-
of a tumour subclone. While it is possible for it to replace all other cer subclones interact with each other. Reports of clonal cooperation,
tumour cells, the timing at which this occurs depends on the strength observed in heterologous xenotransplants, date back to at least 1989
of immune control and its escape rate. (ref.177). Elements of clonal cooperation have since been described as

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 305


Review article

Recruits Cancer cell


Neoantigen

Fibroblast
Presents
antigen
HLA1 Lost HLA1
Cytolysis
Microenvironment 1 follows the expansion of the subclone

Cytotoxic
T cell

Microenvironment 2
creates selective pressure
and favours the subclone
that developed immune
evasion via loss of HLA1

b
Fibro-mucinous stroma
Tgfbr2 knockout
tumour

Fibroblasts

T cells

Socs1 knockout
tumour
Tumour cells Fibroblasts

Primary tumour

Seeding in
different organs

Metastastases

Favourable Unfavourable Unfavourable Favourable


microenvironment microenvironment microenvironment microenvironment

Lung cancer subclone survives in brain and Colon cancer subclone survives in liver and
gives rise to metastasis but does not gives rise to metastasis but does not
metastasize in liver metastasize in brain

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 306


Review article

Fig. 4 | Evolution of the tumour ecosystem. a, Co-evolution of tumour cells clones and TMEs. The yellow Tgfbr2 knockout tumour clone exhibits a fibroblast-
and microenvironments. Some tumour subclones attract characteristic tumour enriched stroma with no presence of T cells, whereas the green Socs1 knockout
microenvironments (TMEs), which subsequently spatially overlap with the clone has a stroma infiltrated with T cells170. c, Organotropism and the seed and
clonal territories. Alternatively, microenvironments may select for specific soil hypothesis of metastasis. Cancer cells from the primary tumour migrate
tumour characteristics such as immune evasion; here, immune evasion is shown throughout the body via blood vessels, with some of them able to seed metastatic
as impaired antigen presentation due to loss of expression of human leukocyte lesions in a distant organ. The target TME determines the fitness of clones at the
antigen 1 (HLA1). b, Examples of experimentally observed associations between site of metastasis favouring some tumour lineages over others.

involving direct and microenvironmentally mediated interactions178–182. Furthermore, DTCs can enter dormant niches that facilitate long-
The ability to comprehensively map genomic evolution, subclone- term survival and even evasion of therapy. In prostate cancer, DTCs
specific gene expression and TMEs is thus likely to further elucidate can compete with haematopoietic stem cells for the occupation of
the full extent and mechanisms of this phenomenon. established endosteal niches191, whereas perivascular niches have been
observed in bone, lung and brain192,193. In melanoma, age-dependent
Disseminated tumour cells, metastatic niches and changes of fibroblasts have been reported to induce emergence from
organotropism dormancy in lung deposits194. Similarly, in a mouse model of melanoma,
Metastases are seeded by disseminated tumour cells (DTCs). The metastases in lymph nodes were found to promote immune evasion
mechanisms that determine whether any given DTC survives, enters a and facilitate further metastasis in distant organs195. As all of these
dormant state or forms a metastasis are poorly defined, but it is evident phenomena involve cellular and spatial interactions, spatial genomic,
that these cells can exploit a variety of existing cellular interactions and transcriptomic and proteomic analyses are likely to provide further
spatial microenvironments. For example, dissemination was shown insights into these processes.
to depend on environmental triggers tied to the circadian rhythm183.
In breast cancer, DTCs physically associated with neutrophils were Translational opportunities
shown to exhibit a more aggressive phenotype184. Additionally, aggre- As discussed in the preceding sections, spatial genomic methods gener-
gates or even fusions of DTCs and macrophages have been described, ate rich insights into the cellular composition, organization and interac-
further illustrating that metastatic dissemination may also rely on tions that shape the evolving cancer ecosystem. For any given cancer,
a range of cellular interactions185. the genome bears the scars of its unique, past evolutionary journey;
The existence of organotropism, whereby certain cancer types however, clinical interventions are designed to shape the future disease
and specific molecular alterations exhibit distinct patterns of meta- course (Fig. 5). A major hope for cancer care is therefore that evolution-
static organ involvement, is well established and is often considered ary features might be leveraged to better predict the clinical trajectory.
in terms of the classical seed and soil hypothesis suggested by Paget, Because tumour evolution is a spatial process, spatially resolved fea-
which posits that certain tissues provide host environments suscep- tures may provide additional means to predict outcomes. Furthermore,
tible to specific tumour types79,186 (Fig. 4c). Organotropism seems to a better understanding of the cellular interactions within the ecosys-
be mostly determined by the cell of origin of the primary tumour, the tem might identify potential therapeutic vulnerabilities that could be
ability of DTCs to interact with the metastatic host environment and exploited to control the evolutionary process127,129,131,196,197. Initial studies
the anatomical proximity of certain target organs such as liver meta­ hint towards the opportunities afforded by these approaches for novel
stases in colorectal cancer. In breast cancer, brain metastases were biomarker discovery137,154,197, in understanding the clinical relevance of
shown to be facilitated by the interactions with glutamatergic neurons187, genetic subclonal structure37,43,198 and in identifying therapeutic vulner-
and a similar pattern was also observed in primary brain cancers where abilities through a deepened understanding of cellular interactions
neoplastic cells form networks with normal astrocytes via synapses188. in both human samples129,131,154 and preclinical animal models170.
The notion that both intrinsic and microenvironmental features
of cellular populations in the primary tumour influence metastatic Spatial biomarker discovery
rates is further supported by lineage-tracing studies in mouse mod- Spatial biomarkers predict clinical outcomes by leveraging information
els189. However, a genomic element also emerges. A recent pan-cancer about cellular organization or intercellular relationships. In its most
analysis of 25,000 metastases of 10 cancer types revealed 57 genetic simplistic form, this approach might measure the distribution of a
associations of organotropism within the same primary tumour type94. certain cell type in relation to another. This is the basic concept of com-
Although the presence of many genomic alterations was associated monly used, histomorphological scoring of tertiary lymphoid struc-
with increased rates of metastasis in multiple organs, there were also tures or tumour infiltrating lymphocytes (TILs) within the tumour or
examples of genomic alterations associated with reduced burden, for stromal compartment (Fig. 3c). Stromal TILs are associated with a supe-
example, RBM10 mutations being less prevalent in brain metastases of rior prognosis and response to chemotherapy or immune-checkpoint
lung cancers. However, it is worth noting that these genetic patterns inhibitors in triple-negative breast cancer, non-small-cell lung cancer
appeared to be mostly specific to certain primary tumour sites rather and melanomas199–203. However, in isolation, the reliability of TILs as
than to target sites. a biomarker is hampered by heterogeneity and irreproducibility203.
A series of studies reported that tumours prime target tissues to Recent studies that use highly multiplexed, in situ, proteomic-based
create pre-metastatic niches facilitating colonization and metastatic assays have started to dissect this complexity by characterizing TIL
outgrowth190. This occurs prior to or on the arrival of DTCs through subtypes, expression, cellular co-localization and neighbourhood pat-
soluble factors and extracellular vesicles released by the primary terns, identifying prognostically meaningful TIL-related patterns197,154.
tumour that can alter the systemic immune response and induce local- In general, head-to-head clinical trials are needed to confirm whether
ized regions of vascular and stromal remodelling in target organs. multiplex biomarkers carry additional prognostic and predictive value

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 307


Review article

Map of subclones Microenvironment Histological context

Normal
Blood vessel
TILs Invasive cancer
Cancer
subclones Necrotic core Carcinoma
TLS in situ
Basement
membrane

Anatomic location Evolutionary history Present Future

Genetics, histology
and TME suggest
high risk of
metastasis if left
untreated

Genetic subclones
Metastasis
ne

y
og
TM

Driver mutation
lo

ol
bc

st
Su

Hi

Fig. 5 | Translational opportunities. Subclone maps, tumour microenvironment component alone. For example, the metastatic potential of two invasive
(TME) composition and histological context each provide clinically and subclonal lineages may differ based not only on the portfolio of their driver
prognostically relevant features. Integrating these levels of information may mutations but also on the TMEs that they inhabit. TILs, tumour infiltrating
help identify ill-fated clonal lineages that may not be found using either lymphocytes; TLS, tertiary lymphoid structure.

compared to single biomarkers such as PDL1 immunohistochemis- data will be critical factors for successful implementation within
try204. Notably, the Immunoscore205 is a digital pathology-based assay large-scale clinical studies.
that measures CD3+ and CD8+ lymphocyte density in the tumour core
and invasive margin and has been proven to surpass clinical staging Clinically relevant subclones
systems for disease-free survival prediction, demonstrating that An unanswered question for personalized medicine is how to deal
higher-dimension datasets could be clinically transformative206. with subclonal driver mutations that can be targeted by antitumour
Beyond a role in refining histology-based biomarkers, spatial therapy. By integrating additional layers of spatial data, it is possi-
omic approaches can also facilitate de novo biomarker discovery. This ble to establish the spatial context and thus the clinical relevance of
was demonstrated by two breast cancer studies (discussed in the sec- subclonal alterations43,208 (Fig. 5). For example, systemic therapies
tion ‘Mapping the cancer ecosystem’) that extracted recurrent cellular administered after primary breast cancer surgery are directed towards
communities from multiplex proteomic and morphological data using covert metastatic disease, with the intention of eliminating these cells
machine learning algorithms137,138. Another use of multiplexed tissue before metastatic deposits have a chance to form. As we cannot directly
imaging was an ovarian cancer study, which found that interactions assay disseminated tumour cells, treatment paradigms are based upon
of exhausted CD8+ T cells, PDL1+ macrophages and PDL1+ tumour cells the properties of the primary tumour. It is reasonable to assume that a
were linked to treatment response207. In these studies, the power to subclonal mutation within the invasive cancer might also be carried by
detect clinically meaningful features was derived from the fact that disseminated tumour cells and could drive metastasis formation.
hundreds of samples could be analysed at high throughput by using By contrast, if the mutation is entirely restricted to precursor lesions,
tissue microarrays. However, the trade-off was that only a tiny part and hence a clone that has not yet even exhibited invasive capacity,
of the main tumour mass was sampled (core needle biopsies with a we can be relatively confident that it will not be a driver of metastasis.
diameter of 0.6–0.8 mm), whereas the invasive margin and stroma Integrating spatial transcriptomic or proteomic data provide
were largely unsampled. An important next step for spatial mole­ further insights into the TME and the gene expression properties of
cular analyses is to derive catalogues of typical cancer ecosystems subclones. This additional characterization will be helpful for deriving
at a tissue-wide scale and at single-cell resolution. These reference predictions of the functional relevance of a particular subclonal driver
sets could serve as a basis for designing more focused studies that mutation, such as by considering a range of intrinsic and extrinsic
might take advantage of lower-plex, higher-throughput or rational phenotypes, including vascular invasion or a high-risk TME niche
subsampling approaches. The scalability and reproducibility of omic (Fig. 5). Although further research is needed, it is possible that spatial

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 308


Review article

analyses could be used in early detection settings or to complement Developing computational approaches such as integrating mathemati-
histopathology-determined completeness of excision, with the latter cal modelling210, mathematical oncology and artificial intelligence-
application yielding opportunities for biomarker-driven surgical and based algorithms may help to overcome these challenges and generate
adjuvant radiotherapy clinical trials. further quantitative insights.
These challenges notwithstanding, the opportunities for spatial
Preclinical studies cancer genomics are formidable. Charting the tumour ecosystem
Although spatial genomic analysis enables reconstruction of the evo- with clonal resolution as well as cellular and molecular details of the
lutionary past and prediction of the clinical future, spatial genomic TME will have great value. Doing so across whole tumour sections and
experiments in animal models can add direct mechanistic evidence cancer types will create insightful atlases that not only provide snap-
of the roles of particular cancer-associated mutations and/or the shots of cancer composition but also insights into past tumour evolu-
effects of spatial aspects of the cancer TME. Such experiments are tion. In addition to a more detailed understanding of tumour growth,
expected to provide clinically important insights into the extracellular these atlases have the potential to reveal the mechanism by which the
consequences of perturbations in cell-intrinsic processes, with many microenvironment is co-opted by tumour cells and how the immune
opportunities for biomarker discovery196,209. A particularly exciting system suppresses tumour proliferation. These processes are already
approach is the combination of spatial profiling with in vivo CRISPR exploited by immune-checkpoint inhibitors, and enhanced molecular
screens131,170. Using this approach, both cell-intrinsic phenotypic and and spatial detail may yield further therapeutic targets or facilitate
local TME associations of tens of genetically distinct clones can be more effective use of existing therapies. Furthermore, the derivation
studied simultaneously in a single animal, leading to insights into of spatial biomarkers and the detection of diagnostic applications of
the precise mechanisms that lead to diverse clinical fates of geneti- spatial genomics are likely to inform treatment; this is especially
cally divergent clones that nonetheless share many genetic and host the case now that spatial genomic, transcriptomic, proteomic and
characteristics170. These models could lead the way in developing pan- metabolomic methods are starting to be used in clinical trials. Such a
ecosystem therapeutic strategies that simultaneously target cell- multimodal approach is required to elucidate the inherent complexity
intrinsic vulnerabilities and harness the properties of the TME in an of the TME — the result will be a revolution of histopathology that is
attempt to terminate cancer evolution. genomically and molecularly informed.
The availability of various single-cell and spatially resolved assays
Conclusions and future perspectives and the emerging insights that these approaches offer thus warrant
While it is widely accepted that the population genetics of carcinogen- new concerted efforts. Previous initiatives, such as The Cancer Genome
esis can be well described in terms of somatic evolution, the spatiotem- Atlas211 and the International Cancer Genome Consortium212,213, have
poral details of the process are not fully understood. Open questions created valuable community resources detailing the molecular and
remain over the extent to which cellular interactions with the TME genomic properties across cancers. It thus seems natural for emerging
and micro-anatomical constraints influence cancer evolution. This projects, such as The Human Tumour Atlas Network214, to follow in this
situation begins to change with the availability of spatial genomic, spirit in order to create the next generation of molecularly resolved
transcriptomic and proteomic technologies that enable charting of cancer atlases.
the growth patterns of distinct subclones and characterization of the
composition and structure of their microenvironments. Connecting Published online: 9 December 2022
these different levels of intratumour heterogeneity is therefore key to
understanding how tumours grow as it reveals how tumour subclones References
1. Cairns, J. Mutation selection and the natural history of cancer. Nature 255, 197–200 (1975).
interact with their microenvironment and how this may lead to the A seminal paper discussing the role of somatic evolution as well as tissue anatomies
selection of aggressive traits. protective against evolution.
2. Nowell, P. C. The clonal evolution of tumor cell populations. Science 194, 23–28 (1976).
Although there is a broad range of technologies with different 3. Martincorena, I. & Campbell, P. J. Somatic mutation in cancer and normal cells. Science
advantages and disadvantages, it emerges that cellular resolution 349, 1483–1489 (2015).
is essential for understanding the micro-anatomy of the cancer eco- 4. Moore, L. et al. The mutational landscape of human somatic and germline cells. Nature
597, 381–386 (2021).
system and localizing molecular signals to specific cells. Mapping A pan-body atlas of mutations and clonal expansion in normal tissues.
how cells are spatially organized into subclones and neighbourhoods 5. Li, R. et al. A body map of somatic mutagenesis in morphologically normal human
and how different cells interface is a key step for understanding the tissues. Nature 597, 398–403 (2021).
6. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature
transcriptional diversity revealed by catalogues of scRNA-seq studies. 578, 94–101 (2020).
Another consideration is the field of view. A typical primary tumour is 7. Greenman, C., Wooster, R., Futreal, P. A., Stratton, M. R. & Easton, D. F. Statistical analysis
more than 1 cm in diameter at diagnosis; hence, technologies enabling of pathogenicity of somatic mutations in cancer. Genetics 173, 2187–2198 (2006).
8. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell
the mapping of entire tumour sections are desirable. Lastly, enabling 171, 1029–1041.e21 (2017).
spatially resolved genomics with the resolution of single nucleotides 9. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new
is important for mapping the subclonal landscape. Whereas spatial cancer-associated genes. Nature 499, 214–218 (2013).
10. Gonzalez-Perez, A. et al. IntOGen-mutations identifies cancer drivers across tumor types.
transcriptomic and proteomic approaches are reaching maturity and Nat. Methods 10, 1081–1082 (2013).
are available as commercial platforms, spatial genomics lags behind. 11. Sondka, Z. et al. The COSMIC Cancer Gene Census: describing genetic dysfunction
A reason for this is the limited amount of DNA per cell, compared to across all human cancers. Nat. Rev. Cancer 18, 696–705 (2018).
12. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000).
the much greater number of copies of RNA and protein, which may be 13. Hanahan, D. Hallmarks of cancer: new dimensions. Cancer Discov. 12, 31–46 (2022).
compounded by tissue sections only containing fractions of a nucleus. 14. Shah, S. P. et al. Mutational evolution in a lobular breast tumour profiled at single
Even though it is possible to calculate copy number alterations using nucleotide resolution. Nature 461, 809–813 (2009).
15. Nik-Zainal, S. et al. The life history of 21 breast cancers. Cell 149, 994–1007 (2012).
low-coverage DNA sequencing or even RNA sequencing, it remains A seminal paper demonstrating that subclonal diversity is common in breast cancer
challenging to estimate phylogenetic relationships and distances. and reconstructing the evolutionary history of copy number alterations.

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 309


Review article

16. McGranahan, N. et al. Clonal status of actionable driver events and the timing 52. Lee-Six, H. et al. Population dynamics of normal human blood inferred from somatic
of mutational processes in cancer evolution. Sci. Transl. Med. 7, 283ra54 (2015). mutations. Nature 561, 473–478 (2018).
17. Andor, N. et al. Pan-cancer analysis of the extent and consequences of intratumor 53. Sender, R., Fuchs, S. & Milo, R. Revised estimates for the number of human and bacteria
heterogeneity. Nat. Med. 22, 105–113 (2015). cells in the body. PLoS Biol. 14, e1002533 (2016).
18. Dentro, S. C. et al. Characterizing genetic intra-tumor heterogeneity across 2658 human 54. Humphries, A. & Wright, N. A. Colonic crypt organization and tumorigenesis. Nat. Rev.
cancer genomes. Cell 184, 2239–2254.e39 (2021). Cancer 8, 415–424 (2008).
A demonstration of universal subclonal divergence across cancer types. 55. Nowak, M. A., Michor, F. & Iwasa, Y. The linear process of somatic evolution. Proc. Natl
19. Navin, N. et al. Tumour evolution inferred by single-cell sequencing. Nature 472, 90–94 Acad. Sci. USA 100, 14966–14969 (2003).
(2011). 56. Sender, R. & Milo, R. The distribution of cellular turnover in the human body. Nat. Med.
A study pioneering the use of single-cell sequencing for understanding cancer 27, 45–48 (2021).
evolution. 57. Lee-Six, H. et al. The landscape of somatic mutation in normal colorectal epithelial cells.
20. Wang, Y. et al. Clonal evolution in breast cancer revealed by single nucleus genome Nature 574, 532–537 (2019).
sequencing. Nature 512, 155–160 (2014). 58. Moore, L. et al. The mutational landscape of normal human endometrial epithelium.
21. Casasent, A. K. et al. Multiclonal invasion in breast tumors identified by topographic Nature 580, 640–646 (2020).
single. Cell Sequencing. Cell 172, 205–217.e12 (2018). 59. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood
22. McPherson, A. et al. Divergent modes of clonal spread and intraperitoneal mixing DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).
in high-grade serous ovarian cancer. Nat. Genet. 48, 758–767 (2016). 60. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes.
23. Laks, E. et al. Clonal decomposition and DNA replication states defined by scaled N. Engl. J. Med. 371, 2488–2498 (2014).
single-cell genome sequencing. Cell 179, 1207–1221.e22 (2019). Genovese et al.59 and Jaiswal et al.60 revealed the frequent presence of macroscopic
24. Salehi, S. et al. Clonal fitness inferred from time-series modelling of single-cell cancer clones with leukaemogenic mutations in the blood of healthy individuals.
genomes. Nature 595, 585–590 (2021). 61. Martincorena, I. et al. Tumor evolution. High burden and pervasive positive selection
25. Williams, M. J., Werner, B., Barnes, C. P., Graham, T. A. & Sottoriva, A. Identification of somatic mutations in normal human skin. Science 348, 880–886 (2015).
of neutral tumor evolution across cancer types. Nat. Genet. 48, 238–244 (2016). A pioneering study revealing that normal skin is a patchwork of microscopic mutant
26. Navin, N. et al. Inferring tumor progression from genomic heterogeneity. Genome Res. clones.
20, 68–80 (2010). 62. Martincorena, I. et al. Somatic mutant clones colonize the human esophagus with age.
27. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed Science 362, 911–917 (2018).
by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012). 63. Lawson, A. R. J. et al. Extensive heterogeneity in somatic mutation and selection in the
One of the first studies using whole-genome sequencing to study patterns human bladder. Science 370, 75–82 (2020).
of branching and subclonal variegation in kidney cancer. 64. Colom, B. et al. Mutant clones in normal epithelium outcompete and eliminate emerging
28. de Bruin, E. C. et al. Spatial and temporal diversity in genomic instability processes tumours. Nature 598, 510–514 (2021).
defines lung cancer evolution. Science 346, 251–256 (2014). 65. Fowler, J. C. et al. Selection of oncogenic mutant clones in normal human skin varies
29. Gerlinger, M. et al. Genomic architecture and evolution of clear cell renal cell with body site. Cancer Discov. 11, 340–361 (2021).
carcinomas defined by multiregion sequencing. Nat. Genet. 46, 225–233 (2014). 66. Abby, E. et al. Notch1 mutation drives clonal expansion in normal esophageal epithelium
30. Yates, L. R. et al. Subclonal diversification of primary breast cancer revealed by but impairs tumor growth. Preprint at bioRxiv https://doi.org/10.1101/2021.06.18.448956
multiregion sequencing. Nat. Med. 21, 751–759 (2015). (2021).
31. Morrissy, A. S. et al. Spatial heterogeneity in medulloblastoma. Nat. Genet. 49, 780–788 67. Louis, D. N. et al. The 2007 WHO classification of tumours of the central nervous system.
(2017). Acta Neuropathol. 114, 97–109 (2007).
32. Jamal-Hanjani, M. et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. 68. Elston, C. W. & Ellis, I. O. Pathological prognostic factors in breast cancer. I. The value
J. Med. 376, 2109–2121 (2017). of histological grade in breast cancer: experience from a large study with long-term
33. Watkins, T. B. K. et al. Pervasive chromosomal instability and karyotype order in tumour follow-up. Histopathology 19, 403–410 (1991).
evolution. Nature 587, 126–132 (2020). 69. Epstein, J. I. et al. The 2014 international society of urological pathology (ISUP) consensus
34. Heide, T. et al. The co-evolution of the genome and epigenome in colorectal cancer. conference on gleason grading of prostatic carcinoma: definition of grading patterns
Nature 611, 733–743 (2022). and proposal for a new grading system. Am. J. Surg. Pathol. 40, 244–252 (2016).
35. Woodcock, D. J. et al. Prostate cancer evolution from multilineage primary to single 70. Greenough, R. B. Varying degrees of malignancy in cancer of the breast. J. Cancer Res. 9,
lineage metastases with implications for liquid biopsy. Nat. Commun. 11, 5070 (2020). 453–463 (1925).
36. Grossmann, S. et al. Development, maturation, and maintenance of human prostate 71. Polyak, K. & Weinberg, R. A. Transitions between epithelial and mesenchymal states:
inferred from somatic mutations. Cell Stem Cell 28, 1262–1274.e5 (2021). acquisition of malignant and stem cell traits. Nat. Rev. Cancer 9, 265–273 (2009).
37. Zhao, T. et al. Spatial genomics enables multi-modal study of clonal heterogeneity 72. Morin, P. J. et al. Activation of β-Catenin-Tcf signaling in colon cancer by mutations
in tissues. Nature 601, 85–91 (2022). in β-catenin or APC. Science 275, 1787–1790 (1997).
38. Heide, T. et al. Multiregion human bladder cancer sequencing reveals tumour evolution, 73. West, J., Schenck, R. O., Gatenbee, C., Robertson-Tessi, M. & Anderson, A. R. A. Normal
bladder cancer phenotypes and implications for targeted therapy. J. Pathol. 248, tissue architecture determines the evolutionary course of cancer. Nat. Commun. 12,
230–242 (2019). 2060 (2021).
39. Su, F. et al. Spatial intratumor genomic heterogeneity within localized prostate cancer 74. Noble, R. et al. Spatial structure governs the mode of tumour evolution. Nat. Ecol. Evol. 6,
revealed by single-nucleus sequencing. Eur. Urol. 74, 551–559 (2018). 207–217 (2022).
40. Bao, L. et al. Coexisting genomic aberrations associated with lymph node metastasis 75. Fearon, E. R. & Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 61,
in breast cancer. J. Clin. Invest. 128, 2310–2324 (2018). 759–767 (1990).
41. Househam, J. et al. Phenotypic plasticity and genetic control in colorectal cancer 76. Gerstung, M. et al. The evolutionary history of 2658 cancers. Nature 578, 122–128 (2020).
evolution. Nature 611, 744–753 (2022). 77. Baslan, T. et al. Ordered and deterministic cancer genome evolution after p53 loss.
42. Biswas, D. et al. A clonal expression biomarker associates with lung cancer mortality. Nature 608, 795–802 (2022).
Nat. Med. 25, 1540–1548 (2019). 78. Lambert, A. W., Pattabiraman, D. R. & Weinberg, R. A. Emerging biological principles
43. Lomakin, A., Svedlund, J., Strell, C., Gataric, M. & Shmatko, A. Spatial genomics maps of metastasis. Cell 168, 670–691 (2017).
the structure, character and evolution of cancer clones. Nature 611, 594–602 (2022). 79. Fidler, I. J. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis
44. Sottoriva, A. et al. A Big Bang model of human colorectal tumor growth. Nat. Genet. 47, revisited. Nat. Rev. Cancer 3, 453–458 (2003).
209–216 (2015). 80. Jones, S. et al. Comparative lesion sequencing provides insights into tumor evolution.
45. Waclaw, B. et al. A spatial model predicts that dispersal and cell turnover limit intratumour Proc. Natl Acad. Sci. USA 105, 4283–4288 (2008).
heterogeneity. Nature 525, 261–264 (2015). 81. Mamlouk, S. et al. DNA copy number changes define spatial patterns of heterogeneity
46. Gallaher, J. A., Brown, J. S. & Anderson, A. R. A. The impact of proliferation-migration in colorectal cancer. Nat. Commun. 8, 14093 (2017).
tradeoffs on phenotypic evolution in cancer. Sci. Rep. 9, 2425 (2019). 82. Zhou, H. et al. Multi-region exome sequencing reveals the intratumoral heterogeneity
47. Lewis, S. M. et al. Spatial omics and multiplexed imaging to explore cancer biology. of surgically resected small cell lung cancer. Nat. Commun. 12, 5431 (2021).
Nat. Methods 18, 997–1012 (2021). 83. Yates, L. R. et al. Genomic evolution of breast cancer metastasis and relapse. Cancer Cell
48. Lieberman, E., Hauert, C. & Nowak, M. A. Evolutionary dynamics on graphs. Nature 433, 32, 169–184.e7 (2017).
312–316 (2005). 84. Noorani, A. et al. Genomic evidence supports a clonal diaspora model for metastases
49. Frean, M., Rainey, P. B. & Traulsen, A. The effect of population structure on the rate of esophageal adenocarcinoma. Nat. Genet. 52, 74–83 (2020).
of evolution. Proc. Biol. Sci. 280, 20130211 (2013). 85. Karlsson, J. et al. Four evolutionary trajectories underlie genetic intratumoral variation
50. Tkadlec, J., Pavlogiannis, A., Chatterjee, K. & Nowak, M. A. Population structure in childhood cancer. Nat. Genet. 50, 944–950 (2018).
determines the tradeoff between fixation probability and fixation time. Commun. Biol. 2, 86. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature
138 (2019). 520, 353–357 (2015).
51. Lopes, J. V., Pacheco, J. M. & Dingli, D. Acquired hematopoietic stem-cell disorders 87. Hu, Z., Li, Z., Ma, Z. & Curtis, C. Multi-cancer analysis of clonality and the timing of systemic
and mammalian size. Blood 110, 4120–4122 (2007). spread in paired primary tumors and metastases. Nat. Genet. 52, 701–708 (2020).

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 310


Review article

88. Brastianos, P. K. et al. Genomic characterization of brain metastases reveals branched 124. Davidson, S. et al. Single-cell RNA sequencing reveals a dynamic stromal niche that
evolution and potential therapeutic targets. Cancer Discov. 5, 1164–1177 (2015). supports tumor growth. Cell Rep. 31, 107628 (2020).
89. Brown, D. et al. Phylogenetic analysis of metastatic progression in breast cancer using 125. Ståhl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial
somatic mutations and copy number aberrations. Nat. Commun. 8, 14944 (2017). transcriptomics. Science 353, 78–82 (2016).
90. De Mattos-Arruda, L. et al. The genomic and immune landscapes of lethal metastatic 126. Wu, S. Z. et al. A single-cell and spatially resolved atlas of human breast cancers.
breast cancer. Cell Rep. 27, 2690–2708.e10 (2019). Nat. Genet. 53, 1334–1347 (2021).
91. Aceto, N. et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer 127. Andersson, A. et al. Spatial deconvolution of HER2-positive breast cancer delineates
metastasis. Cell 158, 1110–1122 (2014). tumor-associated cell type interactions. Nat. Commun. 12, 6012 (2021).
92. Priestley, P. et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature 128. Berglund, E. et al. Spatial maps of prostate cancer transcriptomes reveal an unexplored
575, 210–216 (2019). landscape of heterogeneity. Nat. Commun. 9, 2419 (2018).
93. Martínez-Jiménez, F. et al. Pan-cancer whole genome comparison of primary and 129. Moncada, R. et al. Integrating microarray-based spatial transcriptomics and single-cell
metastatic solid tumors. Preprint at bioRxiv https://doi.org/10.1101/2022.06.17.496528 RNA-seq reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat. Biotechnol.
(2022). 38, 333–342 (2020).
94. Nguyen, B. et al. Genomic characterization of metastatic patterns from prospective 130. Qi, J. et al. Single-cell and spatial analysis reveal interaction of FAP+fibroblasts and
clinical sequencing of 25,000 patients. Cell 185, 563–575.e11 (2022). SPP1+macrophages in colorectal cancer. Nat. Commun. 13, 1742 (2022).
One of the largest genomic analyses of metastasis and organotropism to date. 131. Ji, A. L. et al. Multimodal analysis of composition and spatial architecture in human
95. Reiter, J. G. et al. An analysis of genetic heterogeneity in untreated cancers. Nat. Rev. squamous cell carcinoma. Cell 182, 497–514.e22 (2020).
Cancer 19, 639–650 (2019). 132. Ravi, V. M. et al. T-cell dysfunction in the glioblastoma microenvironment is mediated
96. Aran, D., Sirota, M. & Butte, A. J. Systematic pan-cancer analysis of tumour purity. by myeloid cells releasing interleukin-10. Nat. Commun. 13, 925 (2022).
Nat. Commun. 6, 8971 (2015). 133. Wu, R. et al. Comprehensive analysis of spatial architecture in primary liver cancer.
97. Somarelli, J. A. The hallmarks of cancer as ecologically driven phenotypes. Front. Ecol. Sci. Adv. 7, eabg3750 (2021).
Evol. 9, 661583 (2021). 134. Gouin, K. H. III et al. An N-Cadherin 2 expressing epithelial cell subpopulation predicts
98. Darmanis, S. et al. Single-cell RNA-Seq analysis of infiltrating neoplastic cells at the response to surgery, chemotherapy and immunotherapy in bladder cancer. Nat. Commun.
migrating front of human glioblastoma. Cell Rep. 21, 1399–1410 (2017). 12, 4906 (2021).
99. Venteicher, A. S. et al. Decoupling genetics, lineages, and microenvironment 135. Erickson, A. et al. Spatially resolved clonal copy number alterations in benign and
in IDH-mutant gliomas by single-cell RNA-seq. Science 355, 1391 (2017). malignant tissue. Nature 608, 360–367 (2022).
100. Pombo Antunes, A. R. et al. Single-cell profiling of myeloid cells in glioblastoma across 136. Barkley, D. et al. Cancer cell states recur across tumor types and form specific
species and disease stage reveals macrophage competition and specialization. interactions with the tumor microenvironment. Nat. Genet. 54, 1192–1201 (2022).
Nat. Neurosci. 24, 595–610 (2021). 137. Danenberg, E. et al. Breast tumor microenvironment structures are associated with
101. Puram, S. V. et al. Single-cell transcriptomic analysis of primary and metastatic tumor genomic features and clinical outcome. Nat. Genet. 54, 660–669 (2022).
ecosystems in head and neck cancer. Cell 171, 1611–1624.e24 (2017). A detailed spatial proteomic analysis of more than 500 breast cancers revealing
102. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single- recurrent cellular neighbourhoods associated with clinical outcomes.
cell RNA-seq. Science 352, 189–196 (2016). 138. Jackson, H. W. et al. The single-cell pathology landscape of breast cancer. Nature 578,
103. Izar, B. et al. A single-cell landscape of high-grade serous ovarian cancer. Nat. Med. 26, 615–620 (2020).
1271–1279 (2020). 139. Nirmal, A. J. et al. The spatial landscape of progression and immunoediting in primary
104. Lambrechts, D. et al. Phenotype molding of stromal cells in the lung tumor melanoma at single-cell resolution. Cancer Discov. 12, 1518–1541 (2022).
microenvironment. Nat. Med. 24, 1277–1289 (2018). A detailed multiplexed spatial proteomics analysis of melanoma revealing spatially
105. Chung, W. et al. Single-cell RNA-seq enables comprehensive tumour and immune cell variegated TMEs and cellular interactions.
profiling in primary breast cancer. Nat. Commun. 8, 15081 (2017). 140. Schürch, C. M. et al. Coordinated cellular neighborhoods orchestrate antitumoral
106. Schapiro, D. et al. histoCAT: analysis of cell phenotypes and interactions in multiplex immunity at the colorectal cancer invasive front. Cell 182, 1341–1359.e19 (2020).
image cytometry data. Nat. Methods 14, 873–876 (2017). 141. Wieland, A. et al. Defining HPV-specific B cell responses in patients with head and
107. Arnol, D., Schapiro, D., Bodenmiller, B., Saez-Rodriguez, J. & Stegle, O. Modeling cell-cell neck cancer. Nature 597, 274–278 (2021).
interactions from spatial molecular data with spatial variance component analysis. 142. Gaglia, G. et al. Temporal and spatial topography of cell proliferation in cancer. Nat. Cell
Cell Rep. 29, 202–211.e6 (2019). Biol. 24, 316–326 (2022).
108. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 143. Kather, J. N. et al. Pan-cancer image-based detection of clinically actionable genetic
646–674 (2011). alterations. Nat. Cancer 1, 789–799 (2020).
109. Raskov, H., Orhan, A., Christensen, J. P. & Gögenur, I. Cytotoxic CD8+ T cells in cancer 144. Fu, Y. et al. Pan-cancer computational histopathology reveals mutations, tumor
and cancer immunotherapy. Br. J. Cancer 124, 359–367 (2021). composition and prognosis. Nat. Cancer 1, 800–810 (2020).
110. Philip, M. & Schietinger, A. CD8+ T cell differentiation and dysfunction in cancer. Nat. Rev. 145. Coudray, N. et al. Classification and mutation prediction from non-small cell lung cancer
Immunol. 22, 209–223 (2022). histopathology images using deep learning. Nat. Med. 24, 1559–1567 (2018).
111. Borst, J., Ahrends, T., Bąbała, N., Melief, C. J. M. & Kastenmüller, W. CD4+ T cell help 146. Loeffler, C. M. L. et al. Artificial intelligence–based detection of FGFR3 mutational status
in cancer immunology and immunotherapy. Nat. Rev. Immunol. 18, 635–647 (2018). directly from routine histology in bladder cancer: a possible preselection for molecular
112. Waldman, A. D., Fritz, J. M. & Lenardo, M. J. A guide to cancer immunotherapy: from T cell testing? Eur. Urol. Focus. 8, 472–479 (2022).
basic science to clinical practice. Nat. Rev. Immunol. 20, 651–668 (2020). 147. Kole, A. J. et al. Overall survival is improved when DCIS accompanies invasive breast
113. Sharonov, G. V., Serebrovskaya, E. O., Yuzhakova, D. V., Britanova, O. V. & Chudakov, D. M. cancer. Sci. Rep. 9, 9934 (2019).
B cells, plasma cells and antibody repertoires in the tumour microenvironment. Nat. Rev. 148. Ponz de Leon, M. et al. Evidence for the existence of different types of large bowel tumor:
Immunol. 20, 294–307 (2020). suggestions from the clinical data of a population-based registry. J. Surg. Oncol. 44,
114. Schumacher, T. N. & Thommen, D. S. Tertiary lymphoid structures in cancer. Science 375, 35–43 (1990).
eabf9419 (2022). 149. Sawas, T. et al. Identification of prognostic phenotypes of esophageal adenocarcinoma
115. Sautès-Fridman, C., Petitprez, F., Calderaro, J. & Fridman, W. H. Tertiary lymphoid in 2 independent cohorts. Gastroenterology 155, 1720–1728.e4 (2018).
structures in the era of cancer immunotherapy. Nat. Rev. Cancer 19, 307–325 (2019). 150. Ross-Innes, C. S. et al. Whole-genome sequencing provides new insights into the clonal
116. Wolf, N. K., Kissiov, D. U. & Raulet, D. H. Roles of natural killer cells in immunity to architecture of Barrett’s esophagus and esophageal adenocarcinoma. Nat. Genet. 47,
cancer, and applications to immunotherapy. Nat. Rev. Immunol. https://doi.org/10.1038/ 1038–1046 (2015).
s41577-022-00732-1 (2022). 151. Stachler, M. D. et al. Paired exome analysis of Barrett’s esophagus and adenocarcinoma.
117. Goswami, S., Anandhan, S., Raychaudhuri, D. & Sharma, P. Myeloid cell-targeted Nat. Genet. 47, 1047–1055 (2015).
therapies for solid tumours. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-022- 152. Thomlinson, R. H. & Gray, L. H. The histological structure of some human lung cancers
00737-w (2022). and the possible implications for radiotherapy. Br. J. Cancer 9, 539–549 (1955).
118. Murdoch, C., Muthana, M., Coffelt, S. B. & Lewis, C. E. The role of myeloid cells in the 153. Hammerl, D. et al. Spatial immunophenotypes predict response to anti-PD1 treatment
promotion of tumour angiogenesis. Nat. Rev. Cancer 8, 618–631 (2008). and capture distinct paths of T cell evasion in triple negative breast cancer. Nat. Commun.
119. DeNardo, D. G. & Ruffell, B. Macrophages as regulators of tumour immunity and 12, 5668 (2021).
immunotherapy. Nat. Rev. Immunol. 19, 369–382 (2019). 154. Keren, L. et al. A structured tumor-immune microenvironment in triple negative
120. Valkenburg, K. C., de Groot, A. E. & Pienta, K. J. Targeting the tumour stroma to improve breast cancer revealed by multiplexed ion beam imaging. Cell 174, 1373–1387.e19
cancer therapy. Nat. Rev. Clin. Oncol. 15, 366–381 (2018). (2018).
121. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 155. Galon, J. et al. Towards the introduction of the ‘Immunoscore’ in the classification
(2016). of malignant tumours. J. Pathol. 232, 199–209 (2014).
122. Lendahl, U., Muhl, L. & Betsholtz, C. Identification, discrimination and heterogeneity 156. Hegde, P. S. & Chen, D. S. Top 10 challenges in cancer immunotherapy. Immunity 52,
of fibroblasts. Nat. Commun. 13, 3409 (2022). 17–35 (2020).
123. Sahai, E. et al. A framework for advancing our understanding of cancer-associated 157. Grünwald, B. T. et al. Spatially confined sub-tumor microenvironments in pancreatic
fibroblasts. Nat. Rev. Cancer 20, 174–186 (2020). cancer. Cell 184, 5577–5592.e18 (2021).

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 311


Review article

158. Tavernari, D. et al. Nongenetic evolution drives lung adenocarcinoma spatial 194. Fane, M. E. et al. Stromal changes in the aged lung induce an emergence from
heterogeneity and progression. Cancer Discov. 11, 1490–1507 (2021). melanoma dormancy. Nature 606, 396–405 (2022).
159. Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic 195. Reticker-Flynn, N. E. et al. Lymph node colonization induces tumor-immune tolerance
properties of tumors associated with local immune cytolytic activity. Cell 160, 48–61 to promote distant metastasis. Cell 185, 1924–1942.e23 (2022).
(2015). 196. van Maldegem, F. et al. Characterisation of tumour microenvironment remodelling
160. Thorsson, V. et al. The immune landscape of cancer. Immunity 51, 411–412 (2019). following oncogene inhibition in preclinical studies with imaging mass cytometry.
161. McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity Nat. Commun. 12, 5906 (2021).
to immune checkpoint blockade. Science 351, 1463–1469 (2016). 197. Moldoveanu, D. et al. Spatially mapping the immune landscape of melanoma using
162. Shukla, S. A. et al. Comprehensive analysis of cancer-associated somatic mutations imaging mass cytometry. Sci. Immunol. 7, eabi5072 (2022).
in class I HLA genes. Nat. Biotechnol. 33, 1152–1158 (2015). 198. Erickson, A. et al. Spatially resolved clonal copy number alterations in benign and
163. Goodman, A. M. et al. Prevalence of PDL1 amplification and preliminary response malignant tissue. Nature 608, 360–367 (2022).
to immune checkpoint blockade in solid tumors. JAMA Oncol. 4, 1237–1244 (2018). 199. Azimi, F. et al. Tumor-infiltrating lymphocyte grade is an independent predictor
164. Van den Eynden, J., Jiménez-Sánchez, A., Miller, M. L. & Larsson, E. Lack of detectable of sentinel lymph node status and survival in patients with cutaneous melanoma.
neoantigen depletion signals in the untreated cancer genome. Nat. Genet. 51, 1741–1748 J. Clin. Oncol. 30, 2678–2683 (2012).
(2019). 200. Lee, H. J. et al. Tertiary lymphoid structures: prognostic significance and relationship
165. McGranahan, N. et al. Allele-specific HLA loss and immune escape in lung cancer with tumour-infiltrating lymphocytes in triple-negative breast cancer. J. Clin. Pathol. 69,
evolution. Cell 171, 1259–1271.e11 (2017). 422–430 (2016).
A seminal study that revealed ample evidence of subclonal immune evasion. 201. Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy
166. Balachandran, V. P. et al. Identification of unique neoantigen qualities in long-term response. Nature 577, 549–555 (2020).
survivors of pancreatic cancer. Nature 551, 512–516 (2017). 202. Chen, B. et al. Prognostic value of the common tumour-infiltrating lymphocyte subtypes
167. Łuksza, M. et al. Neoantigen quality predicts immunoediting in survivors of pancreatic for patients with non-small cell lung cancer: a meta-analysis. PLoS One 15, e0242173
cancer. Nature 606, 389–395 (2022). (2020).
168. Sade-Feldman, M. et al. Resistance to checkpoint blockade therapy through inactivation 203. Salgado, R. et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer:
of antigen presentation. Nat. Commun. 8, 1136 (2017). recommendations by an International TILs Working Group 2014. Ann. Oncol. 26, 259–271
169. Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade (2015).
in melanoma. N. Engl. J. Med. 375, 819–829 (2016). 204. Lu, S. et al. Comparison of biomarker modalities for predicting response to PD-1/PD-L1
170. Dhainaut, M. et al. Spatial CRISPR genomics identifies regulators of the tumor checkpoint blockade: a systematic review and meta-analysis. JAMA Oncol. 5, 1195–1204
microenvironment. Cell 185, 1223–1239.e20 (2022). (2019).
A mouse experimental CRISPR model demonstrating direct effects of gene knockouts 205. Galon, J. et al. Type, density, and location of immune cells within human colorectal
and TMEs. tumors predict clinical outcome. Science 313, 1960–1964 (2006).
171. Vennin, C. et al. CAF hierarchy driven by pancreatic cancer cell p53-status creates 206. Pagès, F. et al. International validation of the consensus Immunoscore for the
a pro-metastatic and chemoresistant environment via perlecan. Nat. Commun. 10, 3637 classification of colon cancer: a prognostic and accuracy study. Lancet 391, 2128–2139
(2019). (2018).
172. Tape, C. J. et al. Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. 207. Färkkilä, A. et al. Immunogenomic profiling determines responses to combined PARP
Cell 165, 1818 (2016). and PD-1 inhibition in ovarian cancer. Nat. Commun. 11, 1459 (2020).
173. Ali, H. R. et al. Imaging mass cytometry and multiplatform genomics define the 208. González-Silva, L., Quevedo, L. & Varela, I. Tumor functional heterogeneity unraveled
phenogenomic landscape of breast cancer. Nat. Cancer 1, 163–175 (2020). by scRNA-seq technologies. Trends Cancer Res. 6, 13–19 (2020).
174. Harris, A. L. Hypoxia — a key regulatory factor in tumour growth. Nat. Rev. Cancer 2, 209. Janiszewska, M. et al. Subclonal cooperation drives metastasis by modulating local and
38–47 (2002). systemic immune microenvironments. Nat. Cell Biol. 21, 879–888 (2019).
175. Graeber, T. G. et al. Hypoxia-mediated selection of cells with diminished apoptotic 210. Gatenbee, C. D. et al. Immunosuppressive niche engineering at the onset of human
potential in solid tumours. Nature 379, 88–91 (1996). colorectal cancer. Nat. Commun. 13, 1798 (2022).
176. Baldominos, P. et al. Quiescent cancer cells resist T cell attack by forming an 211. Weinstein, J. N. et al. The cancer genome atlas pan-cancer analysis project. Nat. Genet.
immunosuppressive niche. Cell 185, 1694–1708.e19 (2022). 45, 1113–1120 (2013).
177. Miller, B. E., Miller, F. R. & Heppner, G. H. Therapeutic perturbation of the tumor 212. International Cancer Genome Consortium. International network of cancer genome
ecosystem in reconstructed heterogeneous mouse mammary tumors. Cancer Res. 49, projects. Nature 464, 993–998 (2010).
3747–3753 (1989). 213. ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium. Pan-cancer analysis
178. Alonso-Curbelo, D. et al. A gene-environment-induced epigenetic program initiates of whole genomes. Nature 578, 82–93 (2020).
tumorigenesis. Nature 590, 642–648 (2021). 214. Rozenblatt-Rosen, O. et al. The human tumor atlas network: charting tumor transitions
179. Cleary, A. S., Leonard, T. L., Gestl, S. A. & Gunther, E. J. Tumour cell heterogeneity across space and time at single-cell resolution. Cell 181, 236–249 (2020).
maintained by cooperating subclones in Wnt-driven mammary cancers. Nature 508, 215. Rao, A., Barkley, D., França, G. S. & Yanai, I. Exploring tissue architecture using spatial
113–117 (2014). transcriptomics. Nature 596, 211–220 (2021).
180. Zhou, H., Neelakantan, D. & Ford, H. L. Clonal cooperativity in heterogenous cancers. 216. Emmert-Buck, M. R. et al. Laser capture microdissection. Science 274, 998–1001 (1996).
Semin. Cell Dev. Biol. 64, 79–89 (2017). 217. Ellis, P. et al. Reliable detection of somatic mutations in solid tissues by laser-capture
181. Tammela, T. et al. A Wnt-producing niche drives proliferative potential and progression microdissection and low-input DNA sequencing. Nat. Protoc. 16, 841–871 (2021).
in lung adenocarcinoma. Nature 545, 355–359 (2017). 218. Nichterwitz, S. et al. Laser capture microscopy coupled with Smart-seq2 for precise
182. Williams, J. B. et al. Tumor heterogeneity and clonal cooperation influence the immune spatial transcriptomic profiling. Nat. Commun. 7, 12139 (2016).
selection of IFN-γ-signaling mutant cancer cells. Nat. Commun. 11, 602 (2020). 219. Cui Zhou, D. et al. Spatially restricted drivers and transitional cell populations cooperate
183. Diamantopoulou, Z. et al. The metastatic spread of breast cancer accelerates during with the microenvironment in untreated and chemo-resistant pancreatic cancer.
sleep. Nature 607, 156–162 (2022). Nat. Genet. 54, 1390–1405 (2022).
184. Szczerba, B. M. et al. Neutrophils escort circulating tumour cells to enable cell cycle 220. Rodriques, S. G. et al. Slide-seq: a scalable technology for measuring genome-wide
progression. Nature 566, 553–557 (2019). expression at high spatial resolution. Science 363, 1463–1467 (2019).
185. Adams, D. L. et al. Circulating giant macrophages as a potential biomarker of solid 221. Stickels, R. R. et al. Highly sensitive spatial transcriptomics at near-cellular resolution
tumors. Proc. Natl Acad. Sci. USA 111, 3514–3519 (2014). with Slide-seqV2. Nat. Biotechnol. 39, 313–319 (2021).
186. Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 133, 222. Vickovic, S. et al. High-definition spatial transcriptomics for in situ tissue profiling.
571–573 (1889). Nat. Methods 16, 987–990 (2019).
187. Zeng, Q. et al. Synaptic proximity enables NMDAR signalling to promote brain 223. Liu, Y. et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding
metastasis. Nature 573, 526–531 (2019). in tissue. Cell 183, 1665–1681.e18 (2020).
188. Venkataramani, V. et al. Glioblastoma hijacks neuronal mechanisms for brain invasion. 224. Cho, C.-S. et al. Microscopic examination of spatial transcriptome using Seq-Scope. Cell
Cell 185, 2899–2917.e31 (2022). 184, 3559–3572.e22 (2021).
189. Quinn, J. J. et al. Single-cell lineages reveal the rates, routes, and drivers of metastasis 225. Deng, Y. et al. Spatial-CUT&Tag: spatially resolved chromatin modification profiling at the
in cancer xenografts.Science 371, eabc1944 (2021). cellular level. Science 375, 681–686 (2022).
190. Peinado, H. et al. Pre-metastatic niches: organ-specific homes for metastases. Nat. Rev. 226. Deng, Y. et al. Spatial profiling of chromatin accessibility in mouse and human tissues.
Cancer 17, 302–317 (2017). Nature 609, 375–383 (2022).
191. Shiozawa, Y. et al. Human prostate cancer metastases target the hematopoietic stem cell 227. Chen, A. et al. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA
niche to establish footholds in mouse bone marrow. J. Clin. Invest. 121, 1298–1312 (2011). nanoball-patterned arrays. Cell 185, 1777–1792.e21 (2022).
192. Kienast, Y. et al. Real-time imaging reveals the single steps of brain metastasis formation. 228. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. RNA imaging. Spatially
Nat. Med. 16, 116–122 (2010). resolved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).
193. Ghajar, C. M. et al. The perivascular niche regulates breast tumour dormancy. Nat. Cell 229. Lyubimova, A. et al. Single-molecule mRNA detection and counting in mammalian
Biol. 15, 807–817 (2013). tissue. Nat. Protoc. 8, 1743–1758 (2013).

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 312


Review article

230. Chen, F. et al. Nanoscale imaging of RNA with expansion microscopy. Nat. Methods 13, 264. Nieto, P. et al. A single-cell tumor immune atlas for precision oncology. Genome Res.
679–684 (2016). 31, 1913–1926 (2021).
231. Coskun, A. F. & Cai, L. Dense transcript profiling in single cells by image correlation 265. Park, J. et al. Cell segmentation-free inference of cell types from in situ transcriptomics
decoding. Nat. Methods 13, 657–660 (2016). data. Nat. Commun. 12, 3545 (2021).
232. Codeluppi, S. et al. Spatial organization of the somatosensory cortex revealed 266. Petukhov, V. et al. Cell segmentation in imaging-based spatial transcriptomics.
by osmFISH. Nat. Methods 15, 932–935 (2018). Nat. Biotechnol. 40, 345–354 (2022).
233. Wang, X. et al. Three-dimensional intact-tissue sequencing of single-cell transcriptional 267. Soldatov, R. et al. Spatiotemporal structure of cell fate decisions in murine neural crest.
states. Science 361, eaat5691 (2018). Science 364, eaas9536 (2019).
234. Lee, J. H. et al. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression 268. Prabhakaran, S. Sparcle: assigning transcripts to cells in multiplexed images.
profiling in intact cells and tissues. Nat. Protoc. 10, 442–458 (2015). Bioinforma. Adv. 2, vbac048 (2022).
235. Qian, X. et al. Probabilistic cell typing enables fine mapping of closely related cell types 269. Schapiro, D. et al. MCMICRO: a scalable, modular image-processing pipeline
in situ. Nat. Methods 17, 101–106 (2020). for multiplexed tissue imaging. Nat. Methods 19, 311–315 (2022).
236. Chen, X., Sun, Y.-C., Church, G. M., Lee, J. H. & Zador, A. M. Efficient in situ barcode 270. Lohoff, T. et al. Integration of spatial and single-cell transcriptomic data elucidates
sequencing using padlock probe-based BaristaSeq. Nucleic Acids Res. 46, e22 (2018). mouse organogenesis. Nat. Biotechnol. 40, 74–85 (2022).
237. Alon, S. et al. Expansion sequencing: Spatially precise in situ transcriptomics in intact 271. Zhang, Y. et al. Reference-based cell type matching of spatial transcriptomics data.
biological systems. Science 371, eaax2656 (2021). Preprint at bioRxiv https://doi.org/10.1101/2022.03.28.486139 (2022).
238. Gyllborg, D. et al. Hybridization-based in situ sequencing (HybISS) for spatially resolved 272. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21
transcriptomics in human and mouse brain tissue. Nucleic Acids Res. 48, e112 (2020). (2019).
239. Su, J.-H., Zheng, P., Kinrot, S. S., Bintu, B. & Zhuang, X. Genome-scale imaging of the 273. Kiemen, A. et al. In situ characterization of the 3D microanatomy of the pancreas
3D organization and transcriptional activity of chromatin. Cell 182, 1641–1659.e26 (2020). and pancreatic cancer at single cell resolution. Preprint at bioRxiv https://doi.org/
240. Lu, T., Ang, C. E. & Zhuang, X. Spatially resolved epigenomic profiling of single cells in 10.1101/2020.12.08.416909 (2020).
complex tissues. Cell 185, 4448-4464.e17 (2022). 274. Velten, B. et al. Identifying temporal and spatial patterns of variation from multimodal
241. Eng, C.-H. L. et al. Transcriptome-scale super-resolved imaging in tissues by RNA data using MEFISTO. Nat. Methods 19, 179–186 (2022).
seqFISH. Nature 568, 235–239 (2019). 275. Svensson, V., Teichmann, S. A. & Stegle, O. SpatialDE: identification of spatially variable
242. Wang, F. et al. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, genes. Nat. Methods 15, 343–346 (2018).
paraffin-embedded tissues. J. Mol. Diagn. 14, 22–29 (2012). 276. Lopez, R. et al. DestVI identifies continuums of cell types in spatial transcriptomics data.
243. Ke, R. et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods Nat. Biotechnol. 40, 1360–1369 (2022).
10, 857–860 (2013). 277. McCaffrey, E. F. et al. The immunoregulatory landscape of human tuberculosis granulomas.
244. Baker, A.-M. et al. Robust RNA-based in situ mutation detection delineates colorectal Nat. Immunol. 23, 318–329 (2022).
cancer subclonal evolution. Nat. Commun. 8, 1998 (2017). 278. Dong, K. & Zhang, S. Deciphering spatial domains from spatially resolved transcriptomics
245. Goltsev, Y. et al. Deep profiling of mouse splenic architecture with CODEX multiplexed with an adaptive graph attention auto-encoder. Nat. Commun. 13, 1739 (2022).
imaging. Cell 174, 968–981.e15 (2018). 279. Nagasawa, S. et al. Genomic profiling reveals heterogeneous populations of ductal
246. Saka, S. K. et al. Immuno-SABER enables highly multiplexed and amplified protein carcinoma in situ of the breast. Commun. Biol. 4, 438 (2021).
imaging in tissues. Nat. Biotechnol. 37, 1080–1090 (2019). 280. Elyanow, R., Zeira, R., Land, M. & Raphael, B. J. STARCH: copy number and clone
247. Lin, J.-R. et al. Highly multiplexed immunofluorescence imaging of human tissues and inference from spatial transcriptomics data. Phys. Biol. 18, 035001 (2021).
tumors using t-CyCIF and conventional optical microscopes. eLife 7, e31657 (2018).
248. Lin, J.-R., Fallahi-Sichani, M. & Sorger, P. K. Highly multiplexed imaging of single cells using Acknowledgements
a high-throughput cyclic immunofluorescence method. Nat. Commun. 6, 8390 (2015). We would like to thank S. Dentro, I. Martincorena and T. Grünewald for critical feedback on
249. Gerdes, M. J. et al. Highly multiplexed single-cell analysis of formalin-fixed, the manuscript. L.R.Y. is funded by a Wellcome Trust Clinical Research Career Development
paraffin-embedded cancer tissue. Proc. Natl Acad. Sci. USA 110, 11982–11987 (2013). Fellowship ref: 214584/Z/18/Z.
250. Keren, L. et al. MIBI-TOF: a multiplexed imaging platform relates cellular phenotypes
and tissue structure. Sci. Adv. 5, eaax5851 (2019).
251. He, S. et al. High-plex multiomic analysis in FFPE at subcellular level by spatial molecular
Author contributions
Z.S. and A.L. conducted the literature research. Z.S. drew the initial figures based on
imaging. Preprint at bioRxiv https://doi.org/10.1101/2021.11.03.467020 (2022).
discussions with all authors. M.G. conceived and supervised the study with L.R.Y. All authors
252. Bergholtz, H. et al. Best practices for spatial profiling for breast cancer research with
wrote the manuscript.
the GeoMx® digital spatial profiler. Cancers 13, 4456 (2021).
253. Giesen, C. et al. Highly multiplexed imaging of tumor tissues with subcellular resolution
by mass cytometry. Nat. Methods 11, 417–422 (2014). Competing interests
254. Angelo, M. et al. Multiplexed ion beam imaging of human breast tumors. Nat. Med. 20, The authors declare no competing interests.
436–442 (2014).
255. Mund, A., Brunner, A.-D. & Mann, M. Unbiased spatial proteomics with single-cell Additional information
resolution in tissues. Mol. Cell 82, 2335–2349 (2022). Correspondence should be addressed to Moritz Gerstung.
256. Mund, A. et al. Deep Visual Proteomics defines single-cell identity and heterogeneity.
Nat Biotechnol 40, 1231–1240 (2022). Peer review information Nature Reviews Genetics thanks Alexander Anderson, Alexander
257. Palla, G., Fischer, D. S., Regev, A. & Theis, F. J. Spatial components of molecular tissue Swarbrick, who co-reviewed with Sonny Ramkomuth, and the other, anonymous, reviewer(s)
biology. Nat. Biotechnol. 40, 308–318 (2022). for their contribution to the peer review of this work.
258. Lin, J.-R. et al. Multiplexed 3D atlas of state transitions and immune interactions in
colorectal cancer. Preprint at bioRxiv https://doi.org/10.1101/2021.03.31.437984 (2021). Reprints and permissions information is available at www.nature.com/reprints.
259. Gataric, M. et al. PoSTcode: Probabilistic image-based spatial transcriptomics decoder.
Preprint at bioRxiv https://doi.org/10.1101/2021.10.12.464086 (2021). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
260. Dries, R. et al. Advances in spatial transcriptomic data analysis. Genome Res. 31, published maps and institutional affiliations.
1706–1718 (2021).
261. Perkel, J. M. Starfish enterprise: finding RNA patterns in single cells. Nature 572, 549–551 Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this
(2019). article under a publishing agreement with the author(s) or other rightsholder(s); author self-
262. Hickey, J. W. et al. Spatial mapping of protein composition and tissue organization: archiving of the accepted manuscript version of this article is solely governed by the terms
a primer for multiplexed antibody-based imaging. Nat. Methods 19, 284–295 (2022). of such publishing agreement and applicable law.
263. Kleshchevnikov, V. et al. Cell2location maps fine-grained cell types in spatial
transcriptomics. Nat. Biotechnol. 40, 661–671 (2022). © Springer Nature Limited 2022

Nature Reviews Genetics | Volume 24 | May 2023 | 295–313 313

You might also like