You are on page 1of 20

Reviews

Clinical relevance of tumour-associated


macrophages
Mikael J. Pittet 1,2,3,4,5,6 ✉, Olivier Michielin3,6,7 and Denis Migliorini3,4,5,6

Abstract | In the past decade, substantial advances have been made in understanding the biology
of tumour-associated macrophages (TAMs), and their clinical relevance is emerging. A particular
aspect that is becoming increasingly clear is that the interaction of TAMs with cancer cells and
stromal cells in the tumour microenvironment enables and sustains most of the hallmarks of can-
cer. Therefore, manipulation of TAMs could enable improved disease control in a substantial frac-
tion of patients across a large number of cancer types. In this Review, we examine the diversity of
TAMs in various cancer indications and how this heterogeneity is being revisited with the advent
of single-cell technologies, and then explore the current knowledge on the functional roles of
different TAM states and the prognostic and predictive value of TAM-related signatures. We also
review agents targeting TAMs that are currently being or will soon be tested in clinical trials, and
how manipulations of TAMs can improve existing anticancer treatments. Finally, we discuss how
TAM-targeting approaches could be further integrated into routine clinical practice, considering
a precision oncology approach and viewing TAMs as a dynamic population that can evolve under
treatment pressure.

Immunotherapy has reshaped the therapeutic landscape effect of such manipulations on the outcomes in patients
of many tumour types and can produce exceptional clin- with certain cancers.
ical outcomes. Yet, only a minority of patients respond to As interest in macrophages grows, researchers can
1
Department of Pathology treatment and, therefore, more effective approaches rely on new technologies and tools to better character-
and Immunology, University to stimulate the immune system against cancer are ize these cells. For example, multiomic analyses5–7 and
of Geneva, Geneva,
urgently needed. Currently approved immunotherapies machine-learning approaches for digitized and spatial
Switzerland.
mainly rely on enhancing the functions of endogenous pathology8–12 are uncovering macrophage complexity
2
Ludwig Institute for Cancer
Research, Lausanne,
cytotoxic CD8+ T cells (for example, by administering at unprecedented resolution and in the geographical
Switzerland. immune-checkpoint inhibitors (ICIs)) or on infus- context of tumours. Combined with information on
3
Agora Cancer Research ing engineered T cells into the patient. In addition to clinical outcomes, these data provide a clearer picture of
Center, Lausanne, T cells, other immune components influence tumour macrophage states that might be functionally relevant
Switzerland. progression, either positively or negatively, and should in the tumour setting, and are leading to a better under-
4
Department of Oncology, be considered as additional therapeutic targets1. Of these standing of how to harness macrophages for therapeutic
Geneva University Hospitals, components, macrophages are often the most abundant purposes. In this Review, we focus on the clinical rel-
Geneva, Switzerland.
in tumours, and growing evidence indicates that at least evance of tumour-associated macrophages (TAMs). In
5
Center for Translational
some macrophage subpopulations potently influence particular, we discuss the current knowledge of distinct
Research in Onco-Hematology,
University of Geneva, Geneva,
tumour progression and interfere with virtually all phenotypic states within the TAM population, and their
Switzerland. types of cancer therapy2–4. Thus, increasing interest is potential value as prognostic and predictive markers,
6
Swiss Cancer Center Léman being focused on understanding the biology of macro­ and as therapeutic targets. We also describe the agents
(SCCL), Lausanne and phages, with the ultimate goal of overcoming the lim- targeting TAMs that have already entered clinical trials
Geneva, Geneva, Switzerland. itations of current therapies. Several aspects currently or might do so soon, and propose a strategy for effi-
7
Precision Oncology Center, being addressed include: (1) the complexity and diver- ciently integrating TAM-based therapeutic approaches
Department of Oncology, sity of macrophages, and whether distinct macrophage into the clinical arena.
Lausanne University Hospital,
Lausanne, Switzerland.
states can be identified; (2) whether and how different
✉e-mail: mikael.pittet@ macrophage states contribute to disease progression or TAM diversity
unige.ch suppression; (3) the possibility of rational manipulation TAMs can have multiple origins and functions, and are
https://doi.org/10.1038/ of macrophages to promote an antitumour immune distinct from other myeloid cell types also present in
s41571-022-00620-6 response while sparing non-malignant tissue; and (4) the tumours (Box 1). Our view of TAMs has evolved over the

402 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

Key points of these cells cannot easily be resolved with this binary
classification.
• Tumour-associated macrophages (TAMs) are phenotypically and functionally To overcome this limitation, some researchers have
diverse; therefore, some can promote tumour progression whereas others can exhibit attempted to classify macrophage cell populations into
antitumour activity. additional categories (such as M2a, M2b and M2c
• Some TAM states have prognostic value because their abundance is associated with instead of M2 (refs22,23)) or to use more permissive ter-
distinct clinical outcomes, as is the case for TAM states in patients with melanoma minologies (for example, M1-like and M2-like instead
and lung, colorectal, ovarian, pancreatic, stomach and kidney cancers.
of M1 and M2, respectively3). Supporting this approach,
• Some TAM states are associated with response to treatment, or lack thereof, as M2-like TAMs can be enriched in hypoxic tumour
demonstrated in seminal studies involving patients with breast cancer, renal cell
areas, where they have marked pro-angiogenic activity,
carcinoma and melanoma.
and their number increases with tumour progression.
• Not all tumour-promoting TAMs have M2-like phenotypes, thus highlighting the
Nevertheless, these adjustments in definitions might not
importance of defining TAM states beyond the M1/M2 dichotomy.
be sufficient to cover the full complexity of TAMs. For
• The diversity of TAMs suggests different possibilities for exploiting particular subsets
example, TAM subsets can co-express M1 and M2 gene
for therapeutic purposes; as a result, an arsenal of macrophage-targeted agents are
currently being tested in the clinic.
signatures24, and tumour-promoting macrophages can
express genes other than those classically associated with
• With the integration of single-cell RNA sequencing and other omics in therapeutic
activation towards M2 (refs24–28).
decision-making processes, the inclusion of TAM data in precision oncology molecular
tumour boards could become routine practice.
Single-cell omics-based classification
Single-cell RNA sequencing (scRNA-seq) and other
past decades, owing to an increasing understanding of single-cell analysis tools are increasingly being used to
their complexity. About half a century ago, TAMs were define TAM states. For example, spectral clustering of
identified as cells of the immune system that can kill scRNA-seq data uncovered various macrophage states.
and phagocytose cancer cells13 and, thus, can have anti- Initial studies of human tumours have focused on breast
tumour functions. Subsequent studies, however, showed cancer21,28,29, non-small-cell lung cancer (NSCLC)30,31,
that TAMs can also promote tumour progression. small-cell lung cancer32, hepatocellular carcinoma33,
Indeed, a detailed exploration of the biological complex- glioblastoma27,34, colorectal cancer (CRC)35 and renal
ity of tumour growth and metastasis revealed that the cell carcinoma (RCC)36, and a pan-cancer analysis is
tumour microenvironment (TME) can subvert TAMs, or available24. These scRNA-seq studies are important
at least some of them, into becoming cancer-promoting because they reveal the complexity of TAMs without
cells14. In this sense, an increased abundance of TAMs is defining their phenotypic attributes a priori. For exam-
associated with an overall reduction in patient survival ple, they have confirmed that macrophage activation
across many cancer indications15. within tumours might not follow a simple M1/M2 acti-
The plasticity and diversity of TAMs helps to explain vation model29,30. With this type of information, ration-
their divergent functions, as well as the existence of ally defining which TAM states need to be enhanced
TAMs with antitumour potential16. Hence, identifying or suppressed to treat cancer — and how to do so in a
markers that distinguish tumour-promoting from anti- therapeutic setting — should eventually become easier.
tumour TAMs has become of paramount importance. The different TAM states are not strongly demarcated
Traditionally used flow cytometry and histological by transcriptional signatures, but rather characterized by
approaches rely on a limited set of markers to define TAMs overlapping gene signatures and/or gradients of gene
and, therefore, might not capture the full diversity of these expression, and might be fairly conserved between
cells or distinguish them from some other cell popula- patients who are diagnosed with the same cancer
tions. In the past few years, single-cell omics approaches indication30. As a consequence, researchers could hypo-
have revealed TAM diversity with reduced bias. thetically obtain important insights into TAM biology
from small cohorts of patients, and use those insights
Dual classification to develop widely applicable therapies. The overall rep-
Macrophage diversity has long been recognized and ertoire of TAM states, however, is likely to differ across
thus, terms such as the so-called M1 and M2 pheno- cancer indications, at least in part24. Consequently, tar-
types were introduced to define the possible extremes geting TAMs might require therapeutic agents that are
of in vitro-polarized cells17; the same terminology has tailored to a given cancer type or tumour site. Another
been widely adopted to define macrophages in vivo. important approach would involve defining both the
Bacterial products (for example, lipopolysaccharide) and similarities and differences between TAMs in primary
pro-inflammatory cytokines (such as interferons and metastatic tumours in individual patients, and pos-
and TNF) can induce M1 macrophages, which produce sibly integrating this information when considering
angiostatic factors associated with antitumour immu- therapeutic options in patients with advanced-stage
nity (for example, IL-12 and CXCL10). By contrast, disease. Overall, further studies are needed to highlight
immunoregulatory cytokines (such as IL-4, IL-10 and the similarities and differences between TAMs that are
TGFβ) can induce M2 macrophages, which secrete present in tumours arising in different tissues and, in
tissue-remodelling and pro-angiogenic factors (such as doing so, to define what can be considered comparable
matrix metalloproteinases and VEGF) associated with or different in TAMs, beyond their M1-like and M2-like
tumour promotion. TAMs rarely display bona fide M1 phenotypes, which might only partially describe their
or M2 phenotypes14,17–21, and therefore the complexity activity in vivo.

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 403

0123456789();:
Reviews

Finally, additional TAM states might be discovered as Initial studies used pan-macrophage markers to
other cancer types are investigated, and/or some thera- reveal associations between TAM abundance and clin-
peutic interventions might lead to TAM states that have ical outcomes. These studies involved large cohorts
not yet been revealed. Defining macrophage-associated of patients across many cancer types, mostly with
genes or programmes that are functionally involved in advanced-stage cancers15. Typically, a high abundance
tumour progression or regression might help to uncover of TAMs was found to be associated with unfavourable
important TAM activities without defining macrophage clinical outcomes, with some exceptions. CD68 is often
diversity in its entirety. Arguably, what matters most is used for histological analyses as a pan-macrophage
cell function, not phenotype. marker, but its relevance might be limited because it
can be expressed by cells other than TAMs, and it might
Clinical relevance of TAM states not distinguish tumour-promoting TAMs from other
Once the diversity of TAMs is established, their func- macro­phage populations. CD163, another marker often
tionality needs to be addressed. Several approaches are used in these analyses, is associated with M2-like acti-
being considered to distinguish between ‘favourable’ vation and might help to identify tumour-promoting
and ‘unfavourable’ TAM states. Here, we discuss both TAMs. Gene signatures have also been used to character-
an indirect approach that consists of defining whether ize TAMs in bulk RNA-seq studies39; for example, a TAM
TAM-related signatures have prognostic value and signature including TNF, SIGLEC1, CCL8 and CSF1
a direct approach that involves assessing TAM func- was shown to be enriched in aggressive breast cancer
tion. Of note, in addition to TAMs, certain macro­ subtypes and associated with shorter disease-specific
phages not located in tumours can influence tumour survival26.
progression24,37,38 and should also be considered.
scRNA-seq-defined states and prognosis
Pan-macrophage markers and prognosis A re-evaluation of TAM signatures is being performed
Distinct TAM states can be divided into three groups: with single-cell transcriptomic data, and some have
those associated with unfavourable or with favoura- been associated with a better or worse prognosis (Fig. 1a).
ble clinical outcomes (thus possibly having tumour- Studies using samples from patients with NSCLC and
promoting or antitumour function, respectively), mouse models of lung cancer revealed that, among all
and those not associated with clinical outcome (thus TAMs, the MMP12-expressing state, which resembles
potentially constituting bystander cells in the TME). neither M1 nor M2 cells, is most strongly associated with a
Consequently, therapeutic interventions should involve poor clinical outcome30. A study of different cancer types
agonists or antagonists, or be designed to not affect described other TAM signatures associated with tumour
certain states. progression24. For example, the SPP1-expressing TAM
state, which upregulates pro-angiogenic M2-associated
genes, was identified in multiple tumour types, including
Box 1 | TAMs and other tumour-associated myeloid cells NSCLC, CRC, ovarian cancer and pancreatic adenocar-
Myeloid cells provide host defence against infection and injury, and include mononuclear cinoma (PDAC), in which a high abundance of this cell
cells (such as macrophages and dendritic cells) and polymorphonuclear cells (such as population was associated with a worse clinical outcome.
neutrophils, eosinophils and mast cells). All of these cells can infiltrate tumours and Additional TAM states with a pro-angiogenic signature
regulate their growth. were found in tumours lacking SPP1-expressing TAMs,
• The macrophages present in tumours (or tumour-associated macrophages (TAMs)) including VCAN-expressing, INHBA-expressing and
often originate from circulating monocytes, which are derived from bone-marrow FN1-expressing TAMs in melanoma, gastric cancer
haematopoietic stem cells. TAMs can also originate from tissue-resident cells, which
and RCC, respectively; high expression of VCAN or
are seeded during embryogenesis and persist locally throughout life. TAMs from both
FN1 is also associated with worse clinical outcomes24.
origins can affect tumour progression and have distinct functions61,67.
Therefore, TAM states that are phenotypically distinct
• Dendritic cells can present antigens and orchestrate antitumour T cell responses.
might nevertheless exhibit overlapping functions that
They are found in T cell areas of lymph nodes, where they activate CD4+ and CD8+
T cells, but also within tumours, where they control the fate of tumour-infiltrating contribute to tumour progression.
T cells74,199. An scRNA-seq study investigating the evolution of
• Neutrophils can promote tumour growth. Their presence in tumours is often associ- the TME during RCC progression40 showed an enrich-
ated with a poor clinical outcome. Similarly, a high neutrophil to lymphocyte ratio in ment of M2-like TAMs (expressing CD163, FOLR2 and
blood often predicts a poor prognosis. Nevertheless, neutrophils are heterogeneous MS4A4A) with advancing disease stage. Additionally,
cells, and some of them can have antitumour functions200,201. TAMs without M1/M2-demarcating features might be
• Eosinophils and mast cells can fight parasitic and other infections, but are also functionally important. For example, scRNA-seq analy­sis
involved in allergic reactions and can accumulate in tumours. The role of these of human breast cancers revealed the existence of several
cells in cancer remains largely unresolved, although some of them could have either such TAM states, including CXCL10-expressing TAMs
antitumour or tumour-promoting functions202,203. with M1-like attributes, EGR1-expressing TAMs and
• Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of imma- SIGLEC1-expressing TAMs with M2-like features,
ture cells that suppress T cell responses and promote cancer growth. This terminology and FABP5-expressing TAMs and APOE-expressing
has limitations: (1) the markers used to define MDSCs are also used to define bona TAMs that resemble lipid-associated macrophages.
fide neutrophils and monocytes, which may or may not have immunosuppressive Notably, FABP5-expressing TAMs were present at inva-
functions; (2) this cell population might have functions other than immunosuppres- sive cancer regions, produced various immunoregula-
sion, which should not be overlooked; and (3) mature myeloid cells, such as TAMs,
tory molecules (including PD-L1 and PD-L2) and their
might also be immunosuppressive2.
presence was correlated with unfavourable survival28.

404 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

a Prognostic value These data implicate FABP5-expressing TAMs as rele-


Brain vant therapeutic targets in patients with breast cancer,
– TREM2 although these cells would not have been identified as
tumour-promoting cells using the M2 terminology.
Lung
– • SPP1 FN1 VSIG4 CTSB TAM-related signatures and therapy
• TREM1
Monitoring of human tumours by scRNA-seq has also
Breast started to reveal changes in the TME following thera-
CXCL10 • FABP5 PDL1 PDL2 peutic intervention. Moreover, the presence of certain
• TNF SIGLEC10 CCL8 CSF1
TAM states has been associated with response to therapy
Liver or lack thereof (Fig. 1b). Such monitoring is likely to be
– TREM2 used more extensively in future studies, and will help
Kidney to formulate clearer hypotheses about the relevance of
• CD163 different states of TAMs and other cells in the context

• FABP5 of various therapeutic interventions. Yet, emerging
Pancreas studies are already highly informative.
– SPP1 An scRNA-seq study investigated the TME of
patients with breast cancer receiving anti-PD-1 anti-
Stomach bodies, and compared those with and those with-
– • INHBA out tumour-infiltrating T cell expansion following
• TREM2
treatment41. Pretreatment biopsy samples from patients
Ovary who subsequently had increased T cell infiltration
– SPP1 contained macrophages with distinct phenotypes. In
Colon and/or rectum comparison with patients without T cell expansion,
• SPP1 CD163 FOLR2 MS4A4A TAMs from these patients had higher expression

• TREM1 of genes encoding immune-checkpoint molecules
(such as CD274 and CD273, commonly referred to
Skin
– VCAN
as PDL1 and PDL2, respectively), pro-inflammatory
cytokines (CXCL9, CXCL10 and CXCL11) and pro-
teins from the interferon-stimulated gene (ISG) family
(RSAD2, ISG15 and IFI27). Conversely, a population
TAM state
of CX3CR1-expressing TAMs that also expressed C3,
Favourable TAMs
PLD4, TREM2 and CCL3, were largely absent from pre-
Unfavourable TAMs treatment biopsy samples from patients whose T cells
expanded, but present in patients whose T cells failed
to do so. The abundance of CX3CR1-expressing TAMs
remained much lower on treatment in patients whose
b Predictive value T cells expanded, suggesting that these cells might be
Melanoma24,209 mutually exclusive with tumour-reactive T cells in vivo.
Notably, other TAM states, including LYVE1+ TAMs
Anti-PD-1 VCAN
that co-expressed the M2 markers CD163, MRC1, IL10,
CD163+ PD-L1− FOLR2 and CD209, were detected at similar frequencies
in the two groups of patients.
Breast21,41 Additional studies have revealed associations
Anti-PD-1 PDL1 CXCL10 CX3CR1 TREM1
between the presence of certain TAM states and treat-
ISG C3 CCL3 ment outcomes in other disease contexts. Among
Anti-PD-1 + • CXCL9 MMP9 SPP1 TREM2
patients with triple-negative breast cancer (TNBC),
chemotherapy • CCL2 CXCL10 FN1 C3 those with a response to paclitaxel plus anti-PD-L1
• PLA2G2D antibodies initially had higher baseline levels of
TAMs that expressed MMP9, CXCL9, CXCL10 and
Kidney25,43 PLA2G2D, whereas non-responders had higher levels
Anti-PD-1 ISG of TAMs that expressed TREM2, SPP1, C3 and FN1
Sunitinib ISG (ref.21). In responders, MMP9-expressing TAMs were
co-enriched with CXCL13-expressing CD8+ T cells,
Fig. 1 | Prognostic and predictive value of TAM states. a | Reported tumour-associated whereas TREM2-expressing TAMs were inversely
macrophage (TAM) states with prognostic value in detectable tumours. Favourable and
correlated with CXCL13-expressing CD8 + T cells.
unfavourable TAM states are those for which an increased abundance within tumours
has been associated with shorter or longer overall survival, respectively. b | Reported
These data are consistent with those suggesting that
TAM states with possible predictive value. Favourable and unfavourable TAM states in an increased abundance of CXCL13-expressing CD8+
this context are those for which an increased abundance within tumours has been asso- T cells predicts a favourable response to treatment in
ciated with response to treatment or lack thereof, respectively. In both part a and part b, these patients42, and further indicate that TAMs can pro-
cancer types for which analyses of the TME have been performed are noted — some in foundly modulate tumour-reactive T cells in humans.
the setting of therapy21,24,25,41,43,209. ISG, interferon-stimulated gene. In patients with RCC, the presence of TAMs expressing

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 405

0123456789();:
Reviews

ubiquitin-like proteins of the ISG family was also pre- progression or treatment response, and were performed
dictive of improved progression-free survival (PFS) with in a limited number of patients. Yet, other multidimen-
sunitinib therapy43. Furthermore, in patients with mela- sional approaches, which are more feasible at larger
noma, a lower baseline abundance of VCAN-expressing scale, are also revealing the relevance of TAMs in ther-
TAMs (predicted from bulk RNA-seq datasets) was apy. For example, quantitative and spatially resolved
associated with better responses (defined by RECIST multispectral imaging of tumour tissue sections iden-
criteria) to immunotherapy24. An unresolved question tified CD163+ PD-L1− TAMs that were associated with
is the extent to which the TAM states identified in these lack of response to anti-PD-1 antibodies in pretreatment
studies resemble or differ from each other; addressing melanoma specimens44.
this question will require dedicated approaches that we
discuss below. Mechanistic studies of TAM functions
Other scRNA-seq studies have also indicated that The functions of the TAM states identified in scRNA-seq
favourable TAMs can concomitantly upregulate genes studies so far remain poorly understood; however, most
that might attenuate the antitumour immune response. often, the TME of growing tumours seems to foster the
In a study involving patients with RCC, in those with presence of TAM phenotypes that are associated with
a response to anti-PD-1 antibodies, TAMs broadly tumour promotion. Accordingly, TAMs can produce
shifted towards an M1-like phenotype, with increased factors that enable most of the hallmarks of cancer,
pro­teasome function and antigen presentation; however, including sustaining proliferative signalling, evading
the same TAMs upregulated several immunosuppres- growth suppression and immune destruction, activat-
sive genes25. Similarly, neoadjuvant anti-PD-1 antibody ing invasion and metastasis, inducing angiogenesis and
treatment in patients with recurrent glioblastoma trig- resisting cell death2,4,45. Several tools are available to
gered a CD8+ T cell intratumoural response as well as investigate the functions of TAMs and their relevance
changes in TAMs, suggesting an interferon response to treatment (Box 2).
that might facilitate T cell recruitment and activation. Given the plasticity of macrophages, those at dif-
However, myeloid cells continued to express high levels ferent locations within the same tumour might receive
of immunosuppressive factors43, a phenomenon that different signals from their immediate microenviron-
might prevent fully fledged antitumour responses after ment and develop distinct functions. For example,
anti-PD-1 antibody treatment and/or promote the devel- obesity can suppress the antitumour activity of TAMs46,
opment of resistance to treatment. In addition, some whereas cancer cell-derived succinate47, histamine and
drugs might inadvertently activate tumour-promoting allergic reactions48, and B cell-derived metabolites49
TAMs, as seems to be the case with paclitaxel in patients can all foster the generation of tumour-promoting
with TNBC, thus compromising the efficacy of ICIs21. TAMs. Furthermore, cancer cells can amplify
These studies only provide correlations between cell tumour-promoting TAMs by producing factors includ-
state abundance and clinical outcome, do not indicate ing CSF1 (ref.50), the E3 ubiquitin protein ligase COP1
a causal involvement of a given TAM state in tumour (ref.51), or the metabolite β-glucosylceramide that sim-
ulates an unconventional endoplasmic reticulum stress
response52. Some of these processes preferentially occur
Box 2 | Tools for investigating TAM function and clinical relevance
in the setting of antitumour CD8+ T cell infiltration, sug-
Phenotypic and functional characterization of TAMs will be crucial to guide therapeutic gesting that a TAM response can be viewed, at least in
decisions. Although the evidence on these variables is limited, new strategies are part, as an adaptive resistance mechanism. Accordingly,
becoming available to investigate them using mouse models and patient-derived intravital imaging studies have shown that physical
tumour samples. interactions between TAMs and CD8+ T cells can be
Mouse models long-lasting and promote T cell exhaustion53.
Most of our current knowledge of TAM function comes from experiments in syngeneic
mouse models, owing to the availability of a vast armamentarium of mouse strains and Association of TAMs with treatment outcomes
tools for mechanistic studies. In this regard, defining which TAM states in mice resemble TAM functions have also been dissected following ther-
those in patients is important. The value of cross-species comparisons remains limited, apeutic intervention. Indeed, TAMs can substantially
but current information indicates that phenotypic differences might exist between
affect the outcome of various treatment modalities,
human and mouse macrophage states30,204. These differences suggest potential chal-
lenges when using animal models as surrogates for human disease to study TAMs, and including ICIs, radiation, cytotoxic agents and small
indicate the need to define which TAM states are most relevant to assess mechanisti- molecules2, and therefore their influence in therapy
cally in these models. Humanized mice, such as MITRG and MISTRG, provide alternative needs to be harnessed. For example, TAMs can limit
models205,206. These models might be a step closer to the human context, and indeed the efficacy of ICIs by capturing anti-PD-1 antibodies
TAMs from these mice express M2 markers such as CD163 and CD206; however, the from the surface of PD-1+ T cells. This process depends
extent to which these mice recapitulate the different states of human TAMs remains on interaction of the antibody Fc-domain glycans with
unknown. FcγRs on TAMs, and can be alleviated by preventing
Ex vivo models this interaction54. The relevance of this process is being
Tumour explants190, organoids207 and cell co-cultures50 should also be considered as tested clinically with tislelizumab, a modified anti-PD-1
useful models. New organoid developments, such as submerged Matrigel culture208, antibody that does not bind to FcγRs.
are increasingly relevant to co-culture immune cells with other cell types, including In some settings, TAMs might limit the efficacy of
macrophages. Although these reductionist approaches do not fully recapitulate radiotherapy. Indeed, combining radiation with the
the complexity and dynamics of tumour microenvironments in vivo, they enable CSF1R inhibitor sotuletinib controlled glioblastoma
assessment of human cell functions and drug responses at a mechanistic level.
progression in mice more efficiently than each modality

406 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

alone55. By contrast, some TAMs might contribute to the need to integrate them with existing knowledge. Several
efficacy of radiotherapy, for example, when combined computational methods can be considered, such as those
with therapeutic nanoparticles. Indeed, TAMs adjacent projecting the new scRNA-seq data onto single-cell ref-
to the microvasculature can elicit ‘dynamic bursts’ of erence atlases73, or others directly comparing cell states
drug extravasation after radiation of tumour xenografts across different studies74. Meta-analyses have started
in mice, thereby enhancing drug uptake in neighbour- to reveal important information about mononuclear
ing cancer cells56. In other preclinical models, radiation phagocytes in various tissues75 and about TAMs in
reduced the number of monocyte-derived macrophages, diverse human tumours24,76. Such meta-analyses can
which promoted the survival of antitumour T cells in reveal important features of TAM states, including
liver metastases38, and delayed the progression of brain whether some of them are dependent on the tumour
metastases, albeit only transiently57. tissue in which they reside or, on the contrary, are con-
TAMs can have additional roles during anticancer served across tissues and cancer types. They can also
treatment. For example, PARP inhibitors enhanced both reveal gene expression programmes35 that are shared by
antitumour and tumour-promoting characteristics of multiple TAM states and are functionally important in
macrophages through reprogramming of glucose and each one24. In addition, meta-analyses help to identify
lipid metabolism in a mouse model of TNBC58. Thus, whether different laboratories have given distinct names
macrophage-mediated immunosuppression might be to TAM states that are phenotypically similar, and thus
a downside of PARP inhibitor therapy. Finally, TAMs can occasionally lead to simplifying terminology.
might contribute to the antitumour potency of some
monoclonal antibodies. For example, the antitumour Macrophage-targeting agents
activity of the anti-SLAMF7 antibody elotuzumab in The diversity of TAMs suggests that different approaches
a xenograft mouse model required macrophages and can be taken to exploit them for therapeutic purposes,
antibody-dependent cellular phagocytosis (ADCP) of including elimination, containment or suppression of
cancer cells59. tumour-promoting macrophages, amplification or acti-
vation of antitumour macrophages, subtype switching,
Towards a unified definition or any combination thereof (Fig. 2). As discussed here,
Although the diversity of TAMs remains poorly under- most of the agents currently used are thought to inhibit
stood, the available evidence clearly shows that it is broad. tumour-promoting macrophages, whereas others are
The use of additional analytical methods will surely pro- used owing to their agonistic properties against antitu-
vide further insights into the complexity of these cells. mour TAMs. A first caveat for therapeutic interventions
Indeed, cell states can be defined not only by transcrip- targeting TAMs is that, ideally, they should not affect
tomic data, but also by other measurable dimensions, macrophages that are not related to cancer, such as the
such as proteomic and epigenomic data, and bodily many tissue-resident macrophages that contribute to
localization. The diversity of TAMs further lies in their the homeostasis of non-malignant organs. A second
multiple origins; notably, TAMs include tissue-resident caveat is that these agents will probably be insufficient
cells that self-renew locally independently of adult to control cancer in a durable manner if used as single
haematopoiesis60–64, as well as short-lived cells that are agents. This limited efficacy might be explained in part
derived from circulating monocytes65–68. Integrating by the fact that long-term tumour control requires active
these variables to define TAMs will reveal even more engagement of the adaptive immune system, which
diversity57,61,69–71. TAM heterogeneity is daunting in itself, might not be fully triggered by TAM-targeting agents. In
but the results obtained by incorporating additional vari­ addition, mechanisms of resistance to treatment are likely
ables to define different cellular states could offer sub- to emerge. Therefore, the agents discussed here (Table 1;
stantial benefits. For example, spatial analyses are likely Supplementary Tables 1 and 2) should be considered as
to enable a better understanding of how different pop- entities to be used in combination or sequential therapies
ulations of TAMs interact with other cells and how they to achieve durable responses. Finally, the extent to which
operate within the TME72. Furthermore, the identifica- certain approaches targeting TAMs are better suited to
tion of cell-surface proteins expressed in distinct TAM specific tumour types or sites remains unclear. Given the
states has at least two advantages: (1) it should enable the diversity of both TAMs and the resistance mechanisms
study of newly identified cell states with less expensive that might be triggered by TAM-targeting agents, a bet-
and therefore more easily applicable techniques, such ter understanding of the mode of action of these agents,
as immunohistochemistry and flow cytometry; and (2) when used either alone or in combination with other
it should facilitate the development of new experimen- drugs, will be important. This knowledge should enable
tal drugs that target relevant TAM states while sparing a rational definition of treatment modalities that control
others, including macrophages in non-malignant tissue. disease progression in a greater number of patients and
Furthermore, to consolidate our understanding of are effective against a wider range of cancer types.
TAM diversity, combining and comparing scRNA-seq
data generated by different research laboratories is an CSF1–CSF1R inhibitors
urgent goal. Performing meta-analyses is technically CSF1R is a chemokine receptor expressed by macro­
complicated by the fact that scRNA-seq studies often phages, and across all TAM states, whereas CSF1,
use different protocols, platforms and bioinformatics its ligand, can be produced by cancer cells and other
approaches; however, this endeavour is urgent given components of the TME, such as tumour-associated
the rapidity at which new studies are published and the neutrophils. Activation of the CSF1–CSF1R (CSF1(R))

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 407

0123456789();:
Reviews

signalling pathway promotes the production and prolifer- suppressing tumour-promoting features55,79,80, leading to
ation of macrophage precursors and/or their recruitment reduced tumour progression in some models. CSF1(R)
and retention within inflamed sites, such as tumours14,77. targeting rarely leads to tumour elimination, but the effi-
In experimental models, targeting CSF1(R) can either cacy of treatment can be increased when combined with
reduce TAM numbers 78 or alter their phenotypes other therapeutic agents2.

1. Preventing TAM recruitment 2. Depleting TAMs 3. Reprogramming TAMs


TLR
CXCR4 Tumour TREM1 CD40
parenchyma PI3Kγ TREM1
CLEVER-1
CSF1R
TREM2
CCR2 HDACs
TREM2 BACE1
CCR5 CSF1R
Blood vessel MARCO
CSF1(R) inhibitors
CSF1(R) inhibitors CCR2–CCL2 inhibitors See 1
AMG 820, ARRY-382, Carlumab, MLN1202, • Tissue remodelling factors: Activators of T, NK
TREM1 inhibitor MMPs, cathepsins and B cell responses:
axatilimab, sotuletinib,
PF-04136309
PY159 • Proangiogenic factors: interferons,
cabiralizumab, CXCR4 inhibitors
emactuzumab, TREM2 inhibitor VEGF, IL-8, semaphorin-4D chemokines, IL-12,
Motixafortide,
LY3022855, pexidartinib PY314 • Immunosuppressive factors: ISGs
LY2510924, mavorixafor
IL-10, TGFβ, PD-L1, XBP1
ANG2-targeted agents CCR5–CCL5 inhibitors FRβ
Trebananib, BI836880 Leronlimab, maraviroc CD40 agonists PI3K inhibitors
Sotigalimab, ChiLob7/4, Eganelisib, umbralisib,
selicrelumab, dacetuzumab, tenalisib
Example of a CAR T-cell mitazalimab, SEA-CD40, 2141 V-11 TLR agonists
Engineered construct designed to CLEVER-1 inhibitors Resiquimod, vidutolimod
macrophage deplete TAMs Bexmarilimab TREM inhibitors
HDAC inhibitors See 2
5 Vorinostat, TMP195
IFNγ,
Cancer cell CD40L IL-18 GM-CSF IL-12
RN7SL1
TAM CAF
B cell 1
T cell
TAN Examples of CAR T-cell constructs designed
Monocyte for TAM reprogramming
4 2
DC
4. Exploiting antitumour functions of TAMs

Blood SIRP1α CD47


3 vessel
Tumour Cancer
APMAP TAM cell
microenviroment

SIGLEC10 CD24
5. Infusing engineered macrophages
HER2-targeted SIRP1α–CD47 inhibitors
CAR-I mac IL12-GEMys CAR-147 CAR M AO-176, BI 765063, CC-90002, evorpacept, magrolimab,
SRF231, TTI-621, IBI188, ZL-1201
SIGLEC10–CD24 inhibitors
CD24–Fc

Cytokines IL-12 MMPs Targets


Macrophages engineered to reprogramme the TME and drugs
and/or eliminate cancer cells

Fig. 2 | TAM-targeting agents and engineered macrophages for the reduction in tumour burden, but also in reprogramming of the tumour
treatment of solid tumours. Untreated detectable solid tumours are often microenvironment characterized by increased infiltration of cytotoxic T cells,
infiltrated by a variety of tumour-promoting immune cells, including decreased numbers of M2-like TAMs, increased numbers of M1-like
tumour-associated macrophages (TAMs), which locally suppress the TAMs, and reduced neoangiogenesis. APMAP, adipocyte plasma
infiltration and/or activity of CD8+ T cells and other antitumour immune cells. membrane-associated protein; CAF, cancer-associated fibroblast; CAR,
TAMs can also promote angiogenesis and foster cancer cell survival, invasion chimeric antigen receptor; CAR M, CAR macrophage; DC, dendritic cell; FRβ,
and metastasis. A range of agents targeting and/or exploiting macrophages folate receptor-β; HDAC, histone deacetylase; ISG, interferon-stimulated
have been developed and are listed according to their desired biological gene; NK, natural killer; TAN, tumour-associated neutrophil; TLR, Toll-like
effects. Successful therapeutic targeting of TAMs should result not only in a receptor; TREM, triggering receptor expressed on myeloid cells.

408 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

Table 1 | Clinical trials involving TAM-targeting agents in combination with other drugs
Agent Combination Trial Cancer type Patients SDR ORR Reported biological Clinical trials registry
partners phase (n) (%) (%) responses identifiera
ANG2
Trebananib Anti-PD-1 I Solid tumours 18 22 7 NR NCT03239145
Chemotherapy III Ovarian cancer 1,056 0 0 NR NCT01493505
CCR2–CCL2
Carlumab Chemotherapy I Solid tumours 19 38 5 ↓ Free CCL2 (transient) NCT01204996
PF-04136309 FOLFIRINOX I PDAC 39 14 49 ↓ TAMs NCT01413022
CCR5–CCL5
Leronlimab Chemotherapy I/II CCR5+ TNBC 48 NR NR ↓ TAMs NCT03838367
Maraviroc Anti-PD-1 I CRC 20 5 5 NR NCT03274804
Maraviroc Chemotherapy I CRC 8 37 37 ↑ M1-like TAMs NCT01736813
CD40
Sotigalimab Anti-PD-1, I PDAC 30 30 58 NR NCT03214250
chemotherapy
Anti-PD-1 I/II Melanoma, 19 31 21 ↑ CD8+ T cells, CXCL9 NCT03123783
NSCLC and CXCL10
Selicrelumab Chemotherapy I PDAC 22 53 40 ↑ Inflammatory NCT00711191
cytokines, ↓ B cells
Chemotherapy I Solid tumours 32 NR 20 ↑ Inflammatory NCT00607048
cytokines, ↓ B cells
Anti-CTLA4 I Melanoma 22 NR 27 ↑ T cells NCT01103635
Chemotherapy I Mesothelioma 15 53 40 ↓ CD19 B cells, ↑ CD27
+ +
ACTRN12609000294257
B cells
Anti-CSF1R I Solid tumours 38 NR 41 ↑ CD8+ T cells, ↓ B cells NCT02760797
and monocytes
Chemotherapy I PDAC 16 NR NR ↓ M2-like TAMs, ↑ DC NCT02588443
activation, CXCL10 and
CCL22
Anti-PD-L1 I Solid tumours 80 NR 9 NR NCT02304393
CD47–SIRP1α
AO176 Chemotherapy, I/II Solid tumours 27 26 3 NR NCT03834948
anti-PD-1
CC-90002 Rituximab I CD20+ NHL 28 12 13 NR NCT02367196
Evorpacept Anti-PD-1 I HNSCC, 52 NR 40 NR NCT03013218
gastroesophageal
cancer
Trastuzumab I HNSCC, 30 NR 20 NR NCT03013218
gastroesophageal
cancer
Magrolimab Anti-PD-L1 I Solid tumours 19 5 56 NR NCT03558139
Azacitidine I AML, MDS 24 NR 53 NR NCT03248479
Rituximab I NHL 22 NR 73 NR NCT02953509
Cetuximab I/II Solid tumours 78 23 2 ↑ TAMs NCT02953782
TTI-621 Rituximab I B cell NHL 164 NR 13 NR NCT02663518
Anti-PD-1, I Lymphoma 19 NR 5 NR NCT03530683
anti-PD-L1
CSF1R–CSF1
Axatilimab Anti-PD-L1 I Solid tumours 32 9 0 ↓ Monocytes and NCT03238027
M2-like TAMs
Sotuletinib Anti-PD-1 I/II Solid tumours 49 45 NR NR NCT02829723
Cabiralizumab Anti-PD-1+ I Solid tumours 26 31 4 NR NCT03502330
anti-CD40

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 409

0123456789();:
Reviews

Table 1 (cont.) | Clinical trials involving TAM-targeting agents in combination with other drugs
Agent Combination Trial Cancer type Patients SDR ORR Reported biological Clinical trials registry
partners phase (n) (%) (%) responses identifiera
CSF1R–CSF1 (cont.)
Emactuzumab Chemotherapy I Solid tumours 54 13 7 NR NCT01494688
Anti-CD40 I Solid tumours 37 40 41 ↓ Monocytes and B cells, NCT02760797
↑ CD8+ T cells
Pexidartinib MEK inhibitor I Genitourinary 2 100 0 NR NCT03158103
cancers
Chemotherapy I Advanced-stage 38 34 16 ↓ Monocytes NCT01525602
solid tumours
CXCL12–CXCR4
Motixafortide Anti-PD-1 I PDAC 15 10 5 ↑CD8+ T cells NCT02907099
Anti-PD-1 II PDAC 37 31 3 ↑ CD8+ T cells, ↓ Treg cells NCT02826486
Chemotherapy, II PDAC 22 45 32 NR NCT02826486
anti-PD-1
LY2510924 Chemotherapy, I Leukaemias 11 NR 36 Incomplete suppression NCT02652871
anti-PD-1 of CXCR4
Anti-PD-L1 I Solid tumours 9 0 0 NR NCT02737072
Mavorixafor Anti-PD-1 I Melanoma 13 NR NR ↑ CD8 T cells and IFNγ
+
NCT02823405
PI3K
Eganelisib Anti-PD-1 I Solid tumours 31 NR 6 ↑ CXCL9, CXCL10, NCT02637531
PD-L1 and T cell
activation
Umbralisib Brentuximab vedotin I Hodgkin 11 NR 64 NR NCT02164006
lymphoma
TLRs
Resiquimod NY-ESO-1 vaccine I/II Melanoma 6 NR NR ↑ NY-ESO-1-specific NCT00821652
CD4+ T cells
Representative list of clinical trials with agents targeting TAMs and for which clinical or biological responses have been reported. Targets are listed in alphabetical
order. Additional trials are included in Supplementary Tables 1 and 2. The results of each trial should be contextualized according to the type of tumour treated,
the drug combination used, and the parameters used to measure clinical outcomes. AML, acute myeloid leukaemia; CRC, colorectal cancer; DC, dendritic cell;
FOLFIRINOX, folinic acid, fluorouracil, irinotecan and oxaliplatin; HNSCC, head and neck squamous cell carcinoma; MDS, myelodysplastic syndrome; NHL,
non-Hodgkin lymphoma; NR, not reported; NSCLC, non-small-cell lung cancer; ORR, overall response rate; PDAC, pancreatic ductal adenocarcinoma; SDR, stable
disease rate; TAM, tumour-associated macrophage; TNBC, triple-negative breast cancer; Treg cell, regulatory T cell. aACTRN, Australia New Zealand Clinical Trials
Registry; NCT, ClinicalTrials.gov.

Various drugs targeting CSF1(R) have been or MSI-H CRC (NCT02777710)85. No overt toxicity was
are being tested in clinical trials. These drugs include reported with CSF1(R)-targeting agents as monother-
small-molecule inhibitors (such as sotuletinib, ARRY- apy or in combination with other immunotherapies or
382 and pexidartinib) and antagonistic monoclonal standard chemotherapies.
antibodies (such as emactuzumab, cabiralizumab, Experimental studies have started to evaluate poten-
LY3022855 and axatilimab). Emactuzumab has shown tial mechanisms leading to therapeutic resistance to
efficacy as a single agent in patients with tenosynovial CSF1(R)-targeted agents. For example, in a glioblastoma
sarcoma, a tumour type characterized by CSF1 overex- mouse model in which IGF1R–PI3K signalling was acti-
pression (NCT01494688)78,81. In addition, both alone vated, the emergence of this pathway can lead to resistance
or in combination with paclitaxel, emactuzumab can to sotuletinib, which therefore involves a crosstalk between
reduce the number of M2-like macrophages in patients TAMs and cancer cells. Combining IGF1R or PI3K block-
with solid tumours (NCT01494688) 82. However, ade with CSF1R inhibition delayed tumour progression
CSF1(R)-targeting agents have mostly had limited effi- in these mice86. Another resistance mechanism to CSF1R
cacy as single agents in clinical settings other than ten- targeting, discovered in a mouse model of breast can-
osynovial sarcoma; current trials are mostly evaluating cer brain metastasis, involves CSF2RB (also known as
them in combination with other agents, including ICIs CD131)–STAT5-mediated pro-inflammatory TAM
or chemotherapeutic agents4,83. Two phase I trials have activation71; combining STAT5 signalling inhibition with
reported responses: one found an objective response CSF1R blockade-induced tumour regression in this model.
with sotuletinib and the anti-PD-1 antibody spartal- These seminal studies highlight how pressure on macro­
izumab in one patient with head and neck squamous phages from drug exposure can accelerate the emergence
cell carcinoma (NCT02829723)84, and the other found of resistant cancers. Therefore, a better understanding of
prolonged stable disease with pexidartinib and the how to control or prevent such evolution of resistance to
anti-PD-L1 antibody durvalumab in two patients with treatment in a clinical setting is urgently needed.

410 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

CCL2–CCR2 inhibitors leronlimab, and the dual CCR2 and CCR5 antagonist
The chemokine receptor CCR2 is expressed by inflam- BMS-813160 have been tested clinically. A phase I trial
matory monocytes, which are circulating cells that can involving patients with metastatic CRC or liver metas-
differentiate into TAMs upon extravasation into the tasis from CRC showed that maraviroc treatment can
tumour stroma. CCR2 binds to the chemokine CCL2, favourably alter TAM phenotypes, reducing the expres-
which is produced by many cancer cell types and stromal sion of M2-like markers (NCT01736813)94. In addition,
cells14. CCL2 overexpression can be driven by genetic the FDA has granted Fast Track designation to leron-
mutations, such as in GNA13, and promote immune limab combined with carboplatin for the treatment of
evasion87, and is often associated with a poor prognosis patients with CCR5+ metastatic TNBC97.
in patients88. In mice, inhibition of CCL2–CCR2 signal-
ling can limit the accumulation of inflammatory mono- CXCL12–CXCR4 inhibitors
cytes in tumours, and can also delay cancer metastasis The chemokine receptor CXCR4, expressed by some
and delay tumour progression in mice68. Therefore, at TAMs and other cells, can promote macrophage recruit-
least some tumours might rely on this pathway to recruit ment to tumours by binding its ligand CXCL12, which
and retain TAMs with tumour-promoting functions. can be expressed by tumour-associated stromal cells.
However, interruption of exposure to CCL2-targeting CXCR4–CXCL12 signalling not only leads to TAM
agents can accelerate monocyte accumulation in mouse chemotaxis along CXCL12 gradients, but also activates
tumours and promote cancer growth 89, suggesting cellular programmes that promote cancer cell prolifer-
the importance of prolonged administration of drugs ation and survival98. In combination with cyclophos-
targeting the CCL2–CCR2 pathway. phamide, the CXCR4 inhibitor plerixafor reduces TAM
Several antagonistic antibodies and small-molecule accumulation in various tumour models and delays
inhibitors targeting CCR2 or CCL2 have been tested in tumour progression99. Further preclinical evidence
patients with cancer. For example, multiple early phase from organotypic cultures of human PDAC shows that
trials investigated the anti-CCL2 antibody carlumab CXCR4 blockade can mobilize CD8+ T cells to tumours
in patients with solid tumours refractory to previous and enhance the antitumour activity of anti-PD-1
standard-of-care (SOC) therapies. The findings indi- antibodies100.
cate an acceptable safety profile but a lack of objective Accordingly, in the phase IIa COMBAT trial, the
antitumour responses (NCT01204996) 90. Another peptide CXCR4 antagonist motixafortide increased
trial investigated the small-molecule CCR2 inhibitor the efficacy of ICIs in patients with metastatic PDAC
PF-04136309 in patients with non-metastatic pancreatic (NCT02826486)101. The data suggest that the com-
cancer91. The number of TAMs was lower in tumours bination treatment might decrease both TAMs and
from patients treated with PF-04136309 plus folinic acid, tumour-infiltrating regulatory T cells, and foster a CD8+
fluorouracil, irinotecan and oxaliplatin (FOLFIRINOX) T cell-inflamed TME. The ability of CXCR4-targeting
compared with those receiving FOLFIRINOX alone agents, such as plerixafor, to reduce the influx of TAMs
(NCT01413022). The combination treatment triggered after radiation, a concept called macrophage exclusion
additional changes in the TME, including a concomitant after radiotherapy, is also being tested102. Numerous other
decrease in regulatory T cells, and an increase in effector phase I clinical trials are underway, which combine, for
T cells. A single-arm trial combining the CCR2 inhibitor example, the CXCR4 antagonist peptide LY2510924
CCX872 with FOLFIRINOX indicated the possibility for with durvalumab in patients with advanced-stage solid
increased overall survival (OS) in patients with locally tumours, motixafortide with pembrolizumab (with an
advanced or metastatic, non-resectable pancreatic can- overall response in 10 of 38 patients103) or other immu-
cer, and with no safety concerns (NCT02345408)92. notherapeutics such as cemiplimab (NCT04543071) in
Intriguingly, favourable OS correlated with lower patients with PDAC or with the anti-PD-L1 antibody
peripheral blood monocyte counts at baseline, but no atezolizumab in patients with acute myeloid leukae-
information on TAM abundance was provided. Further mia (AML) (NCT01838395), and the small-molecule
clinical data are needed to assess the effect of this com- CXCR4 antagonist mavorixafor (given orally) with pem-
bination treatment on tumour progression and patient brolizumab in patients with advanced-stage melanoma
survival. (NCT02823405).

CCR5 inhibitors SIRPα–CD47 antagonists


The chemokine receptor CCR5, which can be expressed Tyrosine protein phosphatase non-receptor type sub-
by TAMs, other myeloid cells and T cells, is activated by strate 1 (commonly known as SIRPα) is an inhibitory
a variety of inflammatory cytokines, including CCL3, receptor expressed mostly by macrophages that interacts
CCL4 and CCL5, which might be expressed by can- with the ‘don’t-eat-me’ signal CD47 (ref.104). CD47 is a
cer cells or other cells in the TME. CCL5 and CCR5 cell-surface protein expressed ubiquitously by human
are overexpressed in various cancer types (breast93, cells and often overexpressed by cancer cells. In pre-
colorectal94 and melanoma95 to name a few), suggesting clinical models, blockade of SIRPα–CD47 signalling,
that CCL5–CCR5 signalling promotes tumour progres- for example with antagonistic anti-CD47 antibodies or
sion. Accordingly, various CCR5 inhibitors, including fusion proteins, can induce TAMs to phagocytose cancer
maraviroc, vicriviroc, TAK-779 and anibamine, have cells and stimulate antitumour T cell responses105–107.
demonstrated antitumour effects in mouse models of The clinical relevance of inhibiting SIRPα–CD47
cancer96. The CCR5 inhibitors maraviroc, vicriviroc and signalling has been confirmed in various settings.

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 411

0123456789();:
Reviews

For example, the anti-CD47 antibody magrolimab has with unfavourable clinical outcomes in patients with
shown clinical success in patients with non-Hodgkin certain solid tumour types 119–121. TREM1-targeted
lymphoma, in whom it was associated with an over- agents, including the agonistic anti-TREM1 antibod-
all response rate (ORR) of 73% when combined with ies PY319 and PY159, are being investigated in phase I
the anti-CD20 antibody rituximab (NCT02953509)108. trials involving patients with refractory solid tumours
Additionally, in a phase Ib trial involving patients with (NCT04682431).
AML who were not eligible for intensive chemotherapy,
the ORR was 63% (NCT02678338)109. In 2020, the FDA SIGLEC10–CD24 inhibitors
assigned Breakthrough and Fast Track designations Sialic acid-binding immunoglobulin-like lectins
to magrolimab in combination with azacitidine for (SIGLECs) belong to the immunoglobulin superfamily
the treatment of adult patients with newly diagnosed and have important roles in the regulation of immune
myelodysplastic syndromes, diffuse large B cell lym- cell functions. Among them, SIGLEC10 is an inhibitory
phoma or follicular lymphomas. Magrolimab is also I-type lectin that can be expressed by TAMs and binds the
being explored in combination with the anti-EGFR don’t-eat-me signal CD24, a small, heavily glycosylated,
antibody cetuximab in patients with solid tumours glycosylphosphatidylinositol-anchored, cell-surface pro-
(NCT02953782). This combination might provide ben- tein that can be overexpressed in several solid tumour
efit to patients with CRC (with an overall response in types. In preclinical models, xenografts in Cd24-deficient
2 of 30 patients)110. In preclinical models, magrolimab mice or in wild-type mice exposed to an anti-CD24 anti-
facilitates macrophage-mediated phagocytosis when body had a macrophage-dependent reduction in tumour
combined with anti-HER2 antibodies111, and thus this growth122. As such, the SIGLEC10–CD24 axis acts as an
approach could be tested in patients with breast can- innate immune checkpoint.
cer. Despite CD47 expression by non-malignant cells, The expression of CD24 and CD47 is inversely cor-
no overt toxicity was observed using CD47 targeting related in patients with diffuse large B cell lymphoma,
approaches in patients. As an exception, treatment suggesting the existence of distinct, and perhaps mutu-
with anti-CD47 antibodies can trigger on-target anae- ally exclusive anti-phagocytic signals that can pro-
mia, which is manageable with prime and maintenance mote cancer immune evasion. Consequently, some
dosing of these agents108. Also, it is possible that some tumours might respond differently to treatment with
of these agents might trigger toxicities when used in anti-CD24 versus anti-CD47 antibodies. The ability of
combination with other drugs112. CD24–Fc, a recombinant agonistic fusion protein, to
limit immunotherapy-related adverse events is being
TREM inhibitors tested in patients with advanced-stage solid tumours or
Of the six isoforms of the triggering receptor expressed specifically with metastatic melanoma, and this agent
on myeloid cells (TREM) family, TREM2 has received might also have antitumour activity. Chimeric antigen
the most attention in cancer research owing to its receptor (CAR) natural killer (NK) cells123 and CAR
expression on a subset of TAMs across various tumour T cells124 targeting CD24, as well as antibody–drug con-
types113. TREM2-mediated signalling can control car- jugates, have also been developed and have antitumour
dinal features in macrophages, including cytokine pro- activity in vitro or in xenograft models.
duction, migration and survival114. For example, TREM2
binding to phospholipids can simultaneously abrogate PI3Kγ inhibitors
the production of pro-inflammatory cytokines and TAMs expressing the enzyme PI3Kγ can promote
reactive oxygen species, and increase the production immuno­s uppression and tumour angiogenesis 125.
of immunosuppressive factors. As a result, TREM2+ Accordingly, T cell activation and antitumour activity
TAMs are often associated with a tumour-promoting are more effective in mice deficient in PI3Kγ or exposed
M2-like phenotype, and increased TREM2 expression to the PI3Kγ inhibitors IPI549 or TG100-115 than in
levels in tumour tissues might correlate with poorer their wild-type counterparts not exposed to these agents,
survival115–117. Preclinical studies have further indi- particularly when PI3Kγ inhibitors are combined with
cated a role for TREM2 in promoting cancer progres- anti-PD-1 antibodies126. Several PI3K inhibitors are
sion. For instance, the growth of cancer xenografts is approved for different cancer indications, including
delayed in Trem2-deficient mice and in wild-type mice idelalisib, which is used in combination with rituximab
receiving anti-TREM2 antibodies, and both responded in patients with chronic lymphocytic leukaemia (CLL),
more effectively to anti-PD-1 antibodies than wild-type non-Hodgkin lymphoma and other haematological
mice not exposed to antibodies115. PY314 is a neutral- malignancies127. Of note, idelalisib seems to have more
izing anti-TREM2 humanized monoclonal antibody potent activity against PI3Kδ, and perhaps its effect in
currently being tested in a trial involving patients with TAMs is indirect, at least in part.
advanced-stage solid tumours refractory to all SOC
therapies, including anti-PD-1 antibodies. PY314 spe- CD40 agonists
cifically binds TREM2 and depletes TREM2-expressing CD40, a costimulatory molecule of the TNFR super-
TAMs through antibody-dependent cellular cytotoxicity family, is expressed by some macrophages, dendri­
and/or ADCP. tic cells (DCs), B cells, endothelial cells and some stromal
TREM1, another TREM isoform that is expressed cells, such as bone marrow mesenchymal cells. CD40
by some myeloid cells, also controls inflammatory signalling in macrophages and DCs can stimulate IL-12
processes118. High expression of TREM1 is associated production and promote T helper 1 (TH1) cell-dependent

412 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

immunity, including the production of IFNγ by lympho- was well tolerated and associated with pharmacody-
cytes and the upregulation of MHC class I molecules, namic responses in patients with advanced-stage solid
which are features typically associated with antitumour tumours refractory to SOC therapies (NCT02379741)137.
activity128. Agonistic anti-CD40 antibodies might be par- The serum concentrations of mitazalimab and the
ticularly useful to treat tumours that are poorly infiltrated levels of pro-inflammatory cytokine release were both
by T cells, because they could activate antigen-presenting higher in patients receiving injections into deep metas-
cells (such as macrophages or DCs) that would foster tases than in those receiving injections into superficial
T cell activation and expansion within tumours. metastases. These differences need to be confirmed and
Agonistic anti-CD40 antibodies had substantial anti- understood, although they suggest that deep intral-
tumour activity as single agents in several preclinical esional administration of CD40 agonists might be
models129,130. Also, the combination of these antibodies preferred to activate CD40 signalling in patients.
with other agents, such as anti-CSF1R50 or anti-PD-1 Several approaches are being tested to improve the
antibodies131, can effectively control tumour progres- activation of antitumour immune responses by CD40
sion in animal models, including in those with resist- agonists while sparing non-malignant tissues. For exam-
ance to other agents. In mice, both macrophages and ple, Fc mutations to increase the affinity of this class
DCs probably contribute to tumour control in response of antibodies for FcγR2b might result in more potent
to these antibodies. However, macrophages that reside agonistic activity138, whereas the use of bispecific anti-
in non-malignant tissues can also be activated by ago- bodies could be particularly effective in limiting CD40
nistic anti-CD40 antibodies, which might result in agonist activity to tumours and thus preventing systemic
immunotherapy-related adverse events132. Therefore, a toxicities139.
better understanding of how to maximize the antitumour
activity of these agents while preventing concomitant TLR agonists
activation of unwanted toxicities is important. The human genome encodes several Toll-like recep-
Seven agonistic anti-CD40 antibodies are currently tors (TLRs), which are expressed by macrophages and
being tested in early phase clinical trials. Five of them other myeloid cells, and recognize pathogen-associated
(SEA-CD40, sotigalimab, mitazalimab, ChiLob7/4 and and/or danger-associated molecular patterns. TLR
2141-V11) are of IgG1 isotype and therefore require activation typically triggers potent inflammatory reac-
selective FcγR engagement, while the other two (seli- tions, including the generation of macrophages with
crelumab and CDX-1140) are IgG2 antibodies. This M1-like phenotypes, which might not only protect the
distinction could be important because the Fc region of host against infections but also promote antitumour
antibodies can strongly affect their overall functional- immunity. Consequently, TLR agonists are considered
ity, either positively or negatively, on the basis of their immune enhancers (or adjuvants) in cancer therapy. For
relative affinity for Fc receptors133. Among these seven example, the TLR7 agonist motolimod, the TLR8 ago-
antibodies, selicrelumab has been tested most exten- nist vesatolimod, and the dual TLR7/8 agonist resiqui-
sively to date, alone or in combination with radiation, mod can drive TAM repolarization towards an M1-like
chemotherapy or ICIs128. This antibody has a favour­ phenotype. When loaded in β-cyclodextrin nanoparti-
able safety profile, but is associated with modest ORRs cles, resiquimod could be efficiently delivered to TAMs
when used as a single agent. By contrast, selicrelumab in mice, inducing IL-12 production by these cells and
was associated with an ORR of 41% in multiple solid promoting tumour control140.
tumour types when combined with the anti-CSF1R Drugs that activate various TLRs, including TLR2,
antibody emactuzumab134. Evaluations of other combi- TLR3, TLR4, TLR5, TLR7, TLR8 and TLR9, have
nations, such as one with atezolizumab (NCT02304393), shown efficacy in preclinical studies and have been or
and with either the anti-ANG2 and VEGF bispecific are being tested in clinical trials for indications other
antibody vanucizumab or the anti-VEGF antibody than cancer. Studies comparing different agonists in
bevacizumab (NCT02665416), are underway. patients with cancer are currently lacking. Nevertheless,
Other phase I trials are testing the combination of activating TLRs might provide therapeutic benefit in
agonistic anti-CD40 antibodies with various ICIs and some settings. For example, the TLR7 agonist imiqui-
cytotoxic agents, such as SEA-CD40 plus pembroli- mod is approved by the FDA for topical treatment of
zumab and gemcitabine (NCT02376699)135, and soti- superficial basal cell carcinoma141, the TLR3 agonist
galimab plus gemcitabine, nab-paclitaxel and nivolumab poly ICLC might be associated with meaningful clinical
(NCT03214250)136, both in patients with metastatic and immune responses notably in patients with glioma
PDAC. The sotigalimab combination was associated (NCT00766753 and NCT01920191)142–144; and the TLR9
with an ORR of 58%, although this study included a agonist vidotolimod delivered intralesionally might be
high percentage of patients (27%) with KRAS wild-type useful in overcoming resistance to anti-PD-1 antibodies
disease, who tend to have a favourable prognosis. in patients with metastatic melanoma145.
Randomized controlled trials will help to determine Studies in mice and patients with breast cancer have
whether treatment with a CD40 agonist could replace further revealed that monophosphoryl lipid A, a TLR4
chemotherapy alone as the SOC in this setting. agonist, can be combined with IFNγ to stimulate a tumo-
To avoid the systemic toxicities that can be seen after ricidal phenotype in TAMs and produce the cytokines
intravenous delivery of CD40 agonists, some trials have IL-12 and TNF, which promote a cytotoxic CD8+ T cell
tested intratumoural administration of these drugs. For response146. This agent is already approved by the FDA
example, intratumoural administration of mitazalimab as a vaccine adjuvant and therefore could soon be tested

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 413

0123456789();:
Reviews

in combination with the NY-ESO-1 vaccine in clinical (genetically engineered myeloid cells), which produce
trials (NCT01584115). IL-12, reverse immunosuppression and promote anti-
tumour immunity in mice, thereby reducing metas-
TAM-targeted CAR T cells tasis and delaying tumour growth157. CAR-I Mac are
CAR T cells can be used to eliminate TAMs. For CD19-targeted CAR macrophages induced from pluri-
example, folate receptor-β-targeted CAR T cells can potent stem cells (iPSCs) that acquire M1-like attributes
kill human M2-like macrophages in vitro and mouse upon target recognition and mediate specific killing of
M2-like TAMs in vivo147. This treatment can also pro- CD19+ leukaemia cells158. Additionally, nanoparticles
mote amplification of tumour-infiltrating M1-like cells have been designed to contain both a mannose moiety
and tumour-specific T cells, thereby delaying tumour and DNA encoding for a molecule of interest, such as
progression in mice147, and increase the efficacy of sub- a CAR and/or an inflammatory cytokine. The man-
sequent treatments, suggesting the possibility of using nose moiety confers specificity for mannose receptors
these cells for preconditioning of the TME147. Also, CAR expressed on macrophages and, together with the CAR
T cells expressing RN7SL1, an activator of RIG-I–MDA5 and/or cytokine, enable macrophage-mediated phagocy-
signalling, can eliminate immunosuppressive TAMs and tosis of cancer cells and subsequent activation of adaptive
concurrently activate DCs148. immunity through antigen presentation to T cells159.
CAR T cells can also be used to promote antitumour At present, CAR macrophages pose some manufac-
functions of TAMs. For example, preclinical studies have turing challenges. For example, unlike CAR T cells, they
shown that T cells engineered to release GM-CSF and do not proliferate either ex vivo or in vivo160. Also, solid
IFNγ can activate TAM precursors that produce IL-12 tumours often have antigen heterogeneity, which might
and promote tumour rejection149. Similarly, T cells engi- facilitate tumour escape through antigen loss when using
neered to release inducible IL-12 upon CAR engagement a single CAR approach — the same principle is true for
with cancer cells can eradicate antigen-negative cancer CAR T cells. Finally, a high multiplicity of infection is
cells that under regular conditions would have escaped. required to obtain sufficient levels of CAR expression
In this elimination process, IL-12 activates endogenous in macrophages, which are more resistant to viral trans-
macrophages150. CAR T cells engineered to produce IL-18 duction than T cells154. Nevertheless, CAR macrophages
can also increase the number of M1-like macrophages and have desirable features: they can infiltrate solid tumours
concomitantly decrease that of M2-like macrophages in more efficiently than CAR T cells; they could be derived
mice151. Finally, CAR T cells engineered to constitutively from iPSCs, which opens the possibility of allogeneic
express CD40L can elicit antitumour activity in mice; this treatment without triggering graft-versus-host disease
response is dependent on the activation of endogenous or host rejection; and they can not only naturally engulf
CD40+ cells, including some DCs and TAMs152. cancer cells, but also (cross)present antigens to T cells,
Thus, targeting TAMs with CAR T cell-derived which is important for generating an adaptive antitu-
cytokines and other factors might be used to modify mour immune response160. Ongoing advances in gene
the TME and induce antitumour immune responses. editing and cell engineering techniques could facilitate
Despite the complexity of CAR T cell engineering, one clinical research on this technology.
can envisage the development of even more components
that target either cancer cells or different components Oncolytic viruses
of the TME, including TAMs. In all these approaches, A variety of oncolytic viruses have been used clinically
stable expression of CAR T cell targets over time would to infect cancer cells and produce factors that alter the
be an asset. TME and thereby inhibit tumour growth. The first
oncolytic virus approved by several regulatory agencies
Engineered macrophages including the FDA and EMA, talimogene laherparepvec
Early work showed that TIE-2-expressing monocytes (T-VEC), is a herpesvirus genetically engineered to pro-
modified to produce IFNα can be used to inhibit tumour duce GM-CSF. In patients with advanced-stage mela-
angiogenesis and promote an immune response against noma, T-VEC is associated with higher ORRs and OS
cancer cells in mice153. Subsequently, primary human than GM-CSF alone161. Importantly, intratumoural
macrophages were modified using an adenoviral vector replication of T-VEC might not be restricted to cancer
with a CAR targeting HER2 to recognize and eliminate cells, but also occur in immune cells and particularly
cancer cells. A single infusion of these so-called CAR in TAMs162. Oncolytic viruses have also been used to
macrophages, which can be genetically engineered with produce cytokines typically secreted by antitumour
an M1-like phenotype, can decrease tumour burden and TAMs. For example, a phase I trial tested the ability
delay progression of solid tumour xenografts in mice154. of the oncolytic virus Ad-RTS-hIL-12 to deliver IL-12
This approach received Breakthrough designation by the to resection sites of recurrent high-grade gliomas
FDA (NCT04660929)155. (NCT02026271). Re-resection in five of five patients
Several other CAR constructs have been reported. suspected to have recurrence after treatment revealed
CAR-147 macrophages become activated after CAR primarily pseudoprogression with increased numbers of
binding to HER2 on cancer cells, which activates intra- IFNγ-producing lymphocytes, suggesting the presence
cellular CD147 signalling and leads to increased expres- of treatment-induced antitumour immune responses163.
sion of matrix metalloproteinases; infusion of CAR-147 The non-pathogenic recombinant polio–rhinovirus
macrophages into mice reduced collagen deposition and chimera (PVS–RIPO) recognizes the poliovirus recep-
promoted T cell infiltration into tumours156. IL12-GEMys tor CD155, which is expressed in multiple solid tumour

414 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

types. PVS–RIPO immunotherapy is expected to induce BACE1. Glioblastoma-promoting TAMs express


innate and adaptive antitumour immune responses and, β-secretase 1 (also known as BACE1). Accordingly, the
indeed, in a trial involving patients with grade IV gli- small-molecule BACE1 inhibitor verubecestat can repro-
oma resulted in extended OS compared with a histor- gramme TAMs and stimulate them to phagocytose can-
ical control cohort (NCT01491893)164. Some oncolytic cer cells in xenograft tumour models of glioblastoma175.
viruses have also been used for intravenous infusion. For Given that low-dose radiation can improve TAM
example, viruses from the Orthoreoviridae family (also infiltration176, it might be combined with verubecestat to
referred to as reoviruses) are safe and also preferentially further control tumour growth. Verubecestat and other
infect cancer cells and increase tumour leukocyte infil- BACE1 inhibitors have already been developed for the
tration in patients with malignant glioma or brain metas- treatment of patients with Alzheimer disease and might
tases (Eudract 2011-005635-10)165. Data from a phase I therefore be tested for the treatment of cancer in the
trial also suggest that such viruses lead to increased near future.
cytotoxic T cell tumour infiltration and upregulate
IFN-regulated gene expression as well as PD-L1 expres- CLEVER-1. The scavenger receptor stabilin 1 (also
sion in tumours. Reoviruses have also been tested in known as CLEVER-1) can be expressed by TAMs with
combination with paclitaxel and carboplatin in a phase II tumour-promoting phenotypes. The anti-CLEVER-1
trial involving women with ovarian, tubal or peritoneal antibody bexmarilimab has been tested in patients
cancer (NCT01166542)166, although no significant dif- with advanced-stage solid tumours, with an ORR of 3%
ference in PFS was observed in patients receiving the (NCT03733990). The available data, however, suggested
combination therapy. A phase I trial (NCT02444546) is activation of TH1 cells, increases in the numbers of NK
testing the same oncolytic virus, in combination with and B cells, and higher levels of IFNγ and CXCL10
GM-CSF, in children with relapsed brain tumours. (ref.177), indicating that CLEVER-1 inhibition should
The possibility of incorporating long genetic perhaps be tested in combination with ICIs.
sequences into oncolytic virus expression cassettes is
also being explored. For example, in preclinical stud- HDACs. Histone deacetylase (HDAC) inhibitors can
ies, oncolytic viruses genetically engineered to express open chromatin and thus be used to activate transcrip-
full-length human anti-CD47 antibodies were function- tion of genes that foster tumour control178. For example,
ally active and blocked CD47, thus promoting ADCP by the class IIa HDAC inhibitor TMP195 can induce the dif-
macrophages167. This approach opens the possibility of ferentiation of phagocytic and stimulatory macrophages
making the oncolytic virus platform even more versatile. within mammary tumours towards tumour-promoting
phenotypes179. TMP195 can also trigger the phagocytic
Other therapeutic targets activity of macrophages in the context of CLL180.
Other factors are being explored as therapeutic targets
to modulate TAMs. Here we discuss notable candidates MARCO. TAMs expressing macrophage receptor
whose relevance is mainly supported by preclinical stud- with collagenous structure (also known as MARCO)
ies at present. Some of these factors are being targeted can promote tumour outgrowth, in part through their
and tested in phase I clinical trials, and others are likely immuno­suppressive properties181. Anti-MARCO anti-
to be tested soon. bodies can alter TAM polarization towards antitumour
phenotypes in mice and induce activation of antitu-
ANG2. The growth factor angiopoietin 2 (ANG2) mour NK cells181,182. Combined targeting of MARCO
enhances the pro-angiogenic activities of TIE2- and IL-37 was used to facilitate polarization of human
expressing monocytes168. Ang2 knockdown in mice can TAMs in vitro, resulting in amplification of antitumour
reduce angiogenesis by inhibiting macrophage infiltra- NK cells and T cells and decreased activity of regulatory
tion, thereby controlling tumour progression169. In a T (Treg) cells183.
phase III trial involving patients with advanced-stage
ovarian cancer, the ANG2 agonist trebananib did not Microbiota. Signals from microbiota can shape the func-
improve PFS when combined with standard chemother- tions of TAMs and, in doing so, influence the efficacy
apy (NCT01493505)170. Nevertheless, the same agent is of various cancer treatments. For example, a favourable
being tested in combination with anti-PD-1 antibody microbiota can programme TAMs through the produc-
ezabenlimab in a phase Ib trial involving patients tion of STING agonists to secrete type I interferons and
with advanced-stage solid tumours (NCT03697304). promote antitumour immunity in mice184. The micro-
The bispecific ANG2–VEGF antibody BI836880 has biota could be directly exploited to modulate TAMs;
been tested preclinically for its ability to reprogramme indeed, a high-fibre diet or faecal transplantation has a
TAMs171 and to promote antitumour and anti-angiogenic modulatory effect on the microbiota, which can promote
functions172,173. type I interferon responses by TAMs and improve the
efficacy of immunotherapy185,186.
APMAP. Genome-wide CRISPR knockout and over-
expression screens in cancer cells and macrophages TAM states and precision oncology
have identified regulators of susceptibility to ADCP. Knowledge of the functional relevance of TAM states
One such regulator is the enzyme adipocyte plasma should help to define how best to harness them for
membrane-associated protein (APMAP), which is therapy; however, uncovering the biological activi-
another don’t-eat-me signal of interest174. ties of TAM states is challenging. Many studies have

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 415

0123456789();:
Reviews

characterized TAMs based on the expression of a selec- Nevertheless, the use of additional omics techniques
tion of M1 and M2 markers. This reductionist approach such as scRNA-seq as part of the routine sample process-
has the advantage of greatly simplifying our view of ing of next-generation MTBs187 creates an opportunity to
TAM complexity and thus is easily exploitable; however, integrate TAM data within therapeutic decision-making
M1 and M2 markers might not always provide informa- strategies (Fig. 3). Although no prospective studies have
tion on cell functions and cannot entirely capture the validated the use of TAM data for precision oncology,
diversity of TAM states, as discussed. Consequently, as most MTBs are considering patients with disease pro-
our knowledge of TAM diversity evolves, the readouts gression after receiving SOC treatment, or patients for
used to assess TAM functions are likely to be refined. whom several SOC options are available without vali-
Furthermore, the usefulness of animal models to study dated predictive biomarkers. In both situations, includ-
TAM functions in vivo might be limited by the fact ing TAM derived omics data to guide decisions, while
that the diversity of TAMs in mice does not always collecting and monitoring outcomes, is an interesting
reflect that of TAMs in humans, highlighting the need alternative to empirical treatment assignment.
to develop additional tools to study macrophages,
which could include ex vivo-based or in vitro-based Exploiting fresh tissue samples. Transitioning to
experimental approaches. scRNA-seq has important logistic consequences as it
requires moving away from using commonly accessi-
Integrating TAMs in precision oncology ble formalin-fixed paraffin-embedded archival mate-
To date, TAMs have not been a direct focus of stand- rial to dedicated fresh biopsy samples, requiring active
ard molecular tumour boards (MTBs), which have patient involvement and, in most cases, immediate
mostly relied on bulk DNA and RNA sequencing data. on-site processing. MTBs are currently building the

Fresh tumour material

DC
TAM
TAN

Vessels
B cell
T cell CAF
Immune cells Fibroblasts
Fresh-tumour processing
• scRNA-seq
• FAST-FNA
• Organoids
• Xenografts
Cancer cells • Mechanistic studies

Fine needle aspirates,


liquid biopsies

Molecular
tumour board

Fixed tumour material


Fixed-tumour processing
• IHC, IF
Patient • Spatial transcriptomics
with disease • Digital pathology
progression

CT, MRI, PET molecular imaging TAM-based


innovative
Image processing therapeutic
• Radiomics options

Fig. 3 | Proposed integration of TAM-related data in precision oncology molecular tumour boards. Schematic over-
view of the various sample and image sources that can be used for this purpose. Fresh tumour material represents the rich-
est source of information because it can be used, in addition to standard omics techniques, in ex vivo strategies such as
developing organoids or xenograft models. Fixed samples remain of paramount importance as they facilitate an optimal
use of several omics approaches. This source of material is generally available prospectively as well as in large retrospec-
tive cohorts. Finally, clinical imaging is also expected to provide increasingly relevant information through new radiomics
approaches, which have the advantage of enabling simultaneous evaluation of all tumour sites. As these different data
streams are collected in various precision oncology centres, the inclusion of tumour-associated macrophage (TAM)-
derived data in the therapeutic decision-making process could become feasible. CAF, cancer-associated fibroblast;
FAST–FNA, fast analytical screening technique of fine-needle aspirates; IF, immunofluorescence; IHC, immunohistochemistry;
scRNA-seq, single-cell RNA sequencing; TAN, tumour-associated neutrophil.

416 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

infrastructure to process fresh samples within clini- tumours but are expensive and logistically complex. As
cally relevant timeframes187 and integrated pipelines are important TAM signatures emerge and are validated,
being developed for comprehensive analyses of surgical assessment of those specifically is likely to become eas-
specimens188. Additional emerging techniques can use ier. Multiplexed analyses will probably remain relevant
fresh tissue samples, including fast analytical screen- because they enable simultaneous analysis of multiple
ing techniques of fine-needle aspirates (FAST–FNA)189, components of the TME. Also, many cell states, includ-
ex vivo tumour fragment platforms190, organoids191 ing TAM states, might each require several markers for
and mouse co-clinical trials based on xenografts192. detection. Although tissue-based methods can be use-
Liquid biopsy enabled by contemporary cell capture ful to assess the composition of the TME, on-treatment
techniques193 can bring additional insights through monitoring will require more accessible samples and
less invasive approaches, although blood-derived data sources given that direct access to the tumour com-
data represent only surrogates of the tumour biology. partment with serial biopsies can become clinically and
These analyses of rich fresh tumour material are likely ethically challenging. FNAs can provide on-treatment
to bring highly valuable insights on the use of TAM fresh material containing TAMs. Such procedures can be
data to guide certain precision oncology programmes. performed frequently and, importantly, serially within
However, owing to the high complexity in establishing, the same metastatic lesion, providing informative lon-
normalizing and validating biomarkers based on such gitudinal data using FAST–FNA198. This approach might
data, a crucial aspect for the successful development and be used to assess the evolving TME in patients and pro-
subsequent deployment of TAM-based predictive bio- vide early insights into treatment response189. While this
markers is the ability to bring back some of the features minimally invasive technique can be used to assess a few
derived from fresh samples to commonly accessible, lesions simultaneously, it cannot provide a comprehen-
fixed tissue-based data sources. sive overview of all metastases. Liquid biopsies could
have an important role in providing such patient-level
Exploiting fixed-tissue material. Fixed-tissue material evaluations, although most of the data will be correlative.
constitutes a very rich source of data for TAM-based PET-based molecular imaging paired with advanced
biomarkers in precision oncology. Baseline predic­ radiomics analyses could provide essential data for the
tive biomarkers that use existing fixed material can be patient-level longitudinal monitoring of all metastatic
based on multiplexed immunohistochemistry194, newer lesions. Comparing radiomics features with tissue-based
spatial transcriptomics technologies194–196 or even standard data will help to ascertain what are the key radiomics
haematoxylin and eosin staining analysed with advanced features that most truly reflect the ongoing TAM biology
deep learning methodologies197. Owing to their widespread at the tumour site.
use in clinical routine and the availability of large retro-
spective repositories for validation, fixed-tissue samples Prediction of treatment resistance
could continue to have a key role in the coming years. Advanced on-treatment monitoring of changes in the
TME can yield fundamental insights into mechanisms
Translational end points. A precision oncology approach of response to immunotherapies and other anticancer
targeting the TAM compartment could assess antitu- treatments. Another expected benefit of such close
mour programmes that are orthogonal to those trig- follow-up examinations is the identification, or even
gered by other immunotherapies. Given that a number anticipation, of mechanisms of resistance to TAM-based
of preclinical studies and clinical data suggest that target- therapies. Limited evidence is currently available in this
ing TAMs might enhance antitumour T cell responses, area. Interesting data suggest that anticipating resistance
the extent to which or the circumstances under which to CSF1R-targeted agents is feasible in mice with brain
TAM-targeting approaches mostly unleash adaptive tumours71,86,189, but these results remain to be confirmed
immune responses against cancer remain to be defined. in other tumour types. In addition to determining the
In this context, such approaches might not present an biological mechanism of resistance at play in each clin-
independent mechanistic route to treat tumours as com- ical setting, next-generation omics-based MTBs should
pared with existing T cell-focused immunotherapies; aim to characterize the dynamic patterns of treatment
instead, they would be used to enhance the activity of the resistance at different tumour sites, whether the resis­
adaptive immune response and strengthen the efficacy tance mechanisms are exclusive for a tumour site or
of ICIs. Additionally, TAM-targeting agents might con- systemic and, finally, how to counter them with optimal
trol tumour progression at least in part independently treatment combinations or sequences.
of T cells, for example, by fostering the production of
macrophages with cytotoxic functions. Whether this Conclusions
phenomenon is quantitatively and qualitatively relevant TAMs are innate immune cells that are frequently
in human tumours remains to be defined and will be observed in large numbers in many types of cancer. Their
assessed with the next-generation, omics-based MTB. presence is increasingly recognized as a key parameter
of the tumour-associated immune response. TAMs are
Monitoring treatment outcomes especially important in this response because they can
As discussed, researchers have started to use scRNA-seq either support or inhibit tumour growth depending on
to evaluate cells of the TME, including TAMs, before and their phenotypes and functions, which are diverse, and
during therapeutic intervention. These studies are cru- can also affect the efficacy of virtually any type of ther-
cial to our understanding of treatment effects in human apy. Therefore, understanding the complexity of TAMs

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 417

0123456789();:
Reviews

and exploiting it with treatments targeting defined of TAMs to better understand a particular tumour and
cellular states or their associated functions is crucial. select a treatment.
Moreover, as the phenotype of TAMs can change pro- In this Review, we describe some of the functional states
foundly in response to perturbations, assessing the evo- of TAMs that seem most relevant in clinical and therapeu-
lution of the repertoire and function of TAMs during a tic settings. For some of these states, innovative therapeutic
given treatment is equally important. interventions acting on target pathways are currently being
Our understanding of TAM heterogeneity has tested (Table 1; Supplementary Tables 1 and 2), with prom-
evolved with the advent of single-cell resolution omics ising signs of activity already reported. Understanding
technologies. The resulting analyses indicate that the tra- TAM states and their functions forms the basis for
ditional M1/M2 classification of TAMs does not fully ongoing efforts to include these cells in next-generation,
capture the complexity of all their states. By contrast, the omics-based MTBs. We expect that the integration of
fact that some TAM states have prognostic value because TAM-related parameters into precision immuno-oncology
their abundance is associated with the clinical outcomes will improve the quality of clinical decision-making.
in patients across different types of cancer is becoming Considering TAMs as a dynamic population should
increasingly clear. Some TAM states are also associated be essential for tailoring drug regimens as cancer cells
with a response to immunotherapy, whereas others are, and their TME evolve under treatment pressure.
instead, associated with resistance to treatment, which
further supports the utility of integrating information Published online 30 March 2022

1. Binnewies, M. et al. Understanding the tumor 21. Zhang, Y. et al. Single-cell analyses reveal key immune 38. Yu, J. et al. Liver metastasis restrains immunotherapy
immune microenvironment (TIME) for effective cell subsets associated with response to PD-L1 efficacy via macrophage-mediated T cell elimination.
therapy. Nat. Med. 24, 541–550 (2018). blockade in triple-negative breast cancer. Cancer Cell Nat. Med. 27, 152–164 (2021).
2. Engblom, C., Pfirschke, C. & Pittet, M. J. The role of 39, 1578–1593.e3 (2021). 39. Gentles, A. J. et al. The prognostic landscape of genes
myeloid cells in cancer therapies. Nat. Rev. Cancer 16, 22. Mantovani, A. et al. The chemokine system and infiltrating immune cells across human cancers.
447–462 (2016). in diverse forms of macrophage activation and Nat. Med. 21, 938–945 (2015).
3. Mantovani, A. et al. Tumour-associated macrophages polarisation. Trends Immunol. 25, 677–686 40. Braun, D. A. et al. Progressive immune dysfunction
as treatment targets in oncology. Nat. Rev. Clin. (2004). with advancing disease stage in renal cell carcinoma.
Oncol. 14, 399–416 (2017). 23. Murray, P. J. et al. Macrophage activation and Cancer Cell 39, 632–648.e8 (2021).
4. Bejarano, L., Jordāo, M. J. C. & Joyce, J. A. polarisation: nomenclature and experimental 41. Bassez, A. et al. A single-cell map of intratumoral
Therapeutic targeting of the tumor microenvironment. guidelines. Immunity 41, 14–20 (2014). changes during anti-PD1 treatment of patients with
Cancer Discov. 11, 933–959 (2021). 24. Cheng, S. et al. A pan-cancer single-cell transcriptional breast cancer. Nat. Med. 27, 820–832 (2021).
5. Yofe, I., Dahan, R. & Amit, I. Single-cell genomic atlas of tumor infiltrating myeloid cells. Cell 184, 42. Yang, M. et al. CXCL13 shapes immunoactive tumor
approaches for developing the next generation of 792–809.e23 (2021). microenvironment and enhances the efficacy of PD-1
immunotherapies. Nat. Med. 26, 171–177 (2020). 25. Bi, K. et al. Tumor and immune reprogramming during checkpoint blockade in high-grade serous ovarian
6. Cohen, M. et al. Lung single-cell signaling interaction immunotherapy in advanced renal cell carcinoma. cancer. J. Immunother. Cancer 9, e001136 (2021).
map reveals basophil role in macrophage imprinting. Cancer Cell 39, 649–661.e5 (2021). 43. Krishna, C. et al. Single-cell sequencing links
Cell 175, 1031–1044.e18 (2018). 26. Cassetta, L. et al. Human tumor-associated multiregional immune landscapes and tissue-resident
7. Hara, T. et al. Interactions between cancer cells and macrophage and monocyte transcriptional landscapes T cells in ccRCC to tumor topology and therapy
immune cells drive transitions to mesenchymal-like reveal cancer-specific reprogramming, biomarkers, efficacy. Cancer Cell 39, 662–677.e6 (2021).
states in glioblastoma. Cancer Cell 39, 779–792.e11 and therapeutic targets. Cancer Cell 35, 588–602. 44. Berry, S. et al. Analysis of multispectral imaging with
(2021). e10 (2019). the AstroPath platform informs efficacy of PD-1
8. Alonso-Curbelo, D. et al. A gene-environment-induced 27. Pombo Antunes, A. R. et al. Single-cell profiling blockade. Science 372, eaba2609 (2021).
epigenetic program initiates tumorigenesis. Nature of myeloid cells in glioblastoma across species and 45. Hanahan, D. & Coussens, L. M. Accessories to
590, 642–648 (2021). disease stage reveals macrophage competition the crime: functions of cells recruited to the tumor
9. La Manno, G. et al. Molecular architecture of the and specialization. Nat. Neurosci. 24, 595–610 microenvironment. Cancer Cell 21, 309–322 (2012).
developing mouse brain. Nature 596, 92–96 (2021). (2021). 46. Ringel, A. E. et al. Obesity shapes metabolism in the
10. Satpathy, A. T. et al. Massively parallel single-cell 28. Wu, S. Z. et al. A single-cell and spatially resolved tumor microenvironment to suppress anti-tumor
chromatin landscapes of human immune cell atlas of human breast cancers. Nat. Genet. 53, immunity. Cell 183, 1848–1866.e26 (2020).
development and intratumoral T cell exhaustion. 1334–1347 (2021). 47. Wu, J. Y. et al. Cancer-derived succinate promotes
Nat. Biotechnol. 37, 925–936 (2019). 29. Azizi, E. et al. Single-cell map of diverse immune macrophage polarisation and cancer metastasis via
11. Schede, H. H. et al. Spatial tissue profiling by phenotypes in the breast tumor microenvironment. succinate receptor. Mol. Cell 77, 213–227.e5 (2020).
imaging-free molecular tomography. Nat. Biotechnol. Cell 174, 1293–1308.e36 (2018). 48. Li, H. et al. The allergy mediator histamine confers
39, 968–977 (2021). 30. Zilionis, R. et al. Single-cell transcriptomics of human resistanceto immunotherapy in cancer patients via
12. Wagner, D. E. & Klein, A. M. Lineage tracing meets and mouse lung cancers reveals conserved myeloid activation of the macrophage histamine receptor H1.
single-cell omics: opportunities and challenges. populations across individuals and species. Immunity Cancer Cell 40, 36–52.e9 (2021).
Nat. Rev. Genet. 21, 410–427 (2020). 50, 1317–1334.e10 (2019). 49. Zhang, B. et al. B cell-derived GABA elicits IL-10+
13. Evans, R. & Alexander, P. Cooperation of immune 31. Leader, A. M. et al. Single-cell analysis of human macrophages to limit anti-tumour immunity. Nature
lymphoid cells with macrophages in tumour immunity. non-small cell lung cancer lesions refines tumor 599, 471–476 (2021).
Nature 228, 620–622 (1970). classification and patient stratification. Cancer Cell 50. Neubert, N. J. et al. T cell-induced CSF1 promotes
14. Qian, B. Z. & Pollard, J. W. Macrophage diversity 39, 1594–1609.e12 (2021). melanoma resistance to PD1 blockade. Sci. Transl.
enhances tumor progression and metastasis. Cell 141, 32. Chan, J. M. et al. Signatures of plasticity, metastasis, Med. 10, eaan3311 (2018).
39–51 (2010). and immunosuppression in an atlas of human small 51. Wang, X. et al. In vivo CRISPR screens identify the
15. Zhang, Q. W. et al. Prognostic significance of cell lung cancer. Cancer Cell 39, 1479–1496.e18 E3 ligase Cop1 as a modulator of macrophage
tumor-associated macrophages in solid tumor: a (2021). infiltration and cancer immunotherapy target. Cell
meta-analysis of the literature. PLoS ONE 7, e50946 33. Zhang, Q. et al. Landscape and dynamics of single 184, 5357–5374.e22 (2021).
(2012). immune cells in hepatocellular carcinoma. Cell 179, 52. Di Conza, G. et al. Tumor-induced reshuffling of lipid
16. Vesely, M. D. et al. Natural innate and adaptive 829–845.e20 (2019). composition on the endoplasmic reticulum membrane
immunity to cancer. Annu. Rev. Immunol. 29, 34. Chen, A. X. et al. Single-cell characterization of sustains macrophage survival and pro-tumorigenic
235–271 (2011). macrophages in glioblastoma reveals MARCO as a activity. Nat. Immunol. 22, 1403–1415 (2021).
17. Locati, M., Curtale, G. & Mantovani, A. Diversity, mesenchymal pro-tumor marker. Genome Med. 13, 53. Kersten, K. et al. Spatiotemporal co-dependency
mechanisms, and significance of macrophage 88 (2021). between macrophages and exhausted CD8+ T cells
plasticity. Annu. Rev. Pathol. 15, 123–147 (2020). 35. Pelka, K. et al. Spatially organized multicellular in cancer. Preprint at bioRXiv https://doi.org/10.1101/
18. Gabrusiewicz, K. et al. Glioblastoma-infiltrated innate immune hubs in human colorectal cancer. Cell 184, 2021.09.27.461866 (2021).
immune cells resemble M0 macrophage phenotype. 4734–4752.e20 (2021). 54. Arlauckas, S. P. et al. In vivo imaging reveals a
JCI Insight 1, 85841 (2016). 36. Mujal, A. M. et al. Holistic characterization of tumor tumor-associated macrophage-mediated resistance
19. Szulzewsky, F. et al. Glioma-associated microglia/ monocyte-to-macrophage differentiation integrates pathway in anti-PD-1 therapy. Sci. Transl. Med. 9,
macrophages display an expression profile different distinct immune phenotypes in kidney cancer. eaal3604 (2017).
from M1 and M2 polarisation and highly express Preprint at bioRxiv https://doi.org/10.1101/ 55. Akkari, L. et al. Dynamic changes in glioma
Gpnmb and Spp1. PLoS ONE 10, e0116644 (2015). 2021.07.07.451502 (2021). macrophage populations after radiotherapy reveal
20. Yang, M. et al. Diverse functions of macrophages in 37. Pucci, F. et al. SCS macrophages suppress melanoma CSF-1R inhibition as a strategy to overcome
different tumor microenvironments. Cancer Res. 78, by restricting tumor-derived vesicle-B cell interactions. resistance. Sci. Transl. Med. 12, eaaw7843
5492–5503 (2018). Science 352, 242–246 (2016). (2020).

418 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

56. Miller, M. A. et al. Radiation therapy primes tumors 82. Gomez-Roca, C. A. et al. Phase I study of emactuzumab pancreatic adenocarcinoma (mPDAC) [abstract].
for nanotherapeutic delivery via macrophage- single agent or in combination with paclitaxel in Cancer Res. 81 (Suppl. 13), CT177 (2021).
mediated vascular bursts. Sci. Transl. Med. 9, patients with advanced/metastatic solid tumors reveals 104. Feng, M. et al. Phagocytosis checkpoints as new
eaal0225 (2017). depletion of immunosuppressive M2-like macrophages. targets for cancer immunotherapy. Nat. Rev. Cancer
57. Niesel, K. et al. The immune suppressive Ann. Oncol. 30, 1381–1392 (2019). 19, 568–586 (2019).
microenvironment affects efficacy of radio- 83. DeNardo, D. G. & Ruffell, B. Macrophages as 105. Chao, M. P. et al. Anti-CD47 antibody synergizes with
immunotherapy in brain metastasis. EMBO Mol. regulators of tumour immunity and immunotherapy. rituximab to promote phagocytosis and eradicate
Med. 13, e13412 (2021). Nat. Rev. Immunol. 19, 369–382 (2019). non-Hodgkin lymphoma. Cell 142, 699–713 (2010).
58. Mehta, A. K. et al. Targeting immunosuppressive 84. Lin, C.-C. et al. Phase I study of BLZ945 alone and 106. Gholamin, S. et al. Disrupting the CD47-SIRPα
macrophages overcomes PARP inhibitor resistance with spartalizumab (PDR001) in patients (pts) with anti-phagocytic axis by a humanized anti-CD47
in BRCA1-associated triple-negative breast cancer. advanced solid tumors [abstract]. Cancer Res. 80 antibody is an efficacious treatment for malignant
Nat. Cancer 2, 66–82 (2021). (Suppl. 16), CT171 (2020). pediatric brain tumors. Sci. Transl. Med. 9, eaaf2968
59. Kurdi, A. T. et al. Antibody-dependent cellular 85. Cassier, P. A. et al. MEDIPLEX: a phase 1 study (2017).
phagocytosis by macrophages is a novel mechanism of durvalumab (D) combined with pexidartinib (P) 107. Weiskopf, K. et al. Engineered SIRPα variants
of action of elotuzumab. Mol. Cancer Ther. 17, in patients (pts) with advanced pancreatic ductal as immunotherapeutic adjuvants to anticancer
1454–1463 (2018). adenocarcinoma (PDAC) and colorectal cancer antibodies. Science 341, 88–91 (2013).
60. Bowman, R. L. et al. Macrophage ontogeny underlies (CRC) [abstract]. J. Clin. Oncol. 37 (Suppl. 15), 2579 108. Advani, R. et al. CD47 blockade by Hu5F9-G4 and
differences in tumor-specific education in brain (2019). rituximab in non-Hodgkin’s lymphoma. N. Engl. J.
malignancies. Cell Rep. 17, 2445–2459 (2016). 86. Quail, D. F. et al. The tumor microenvironment Med. 379, 1711–1721 (2018).
61. Casanova-Acebes, M. et al. Tissue-resident underlies acquired resistance to CSF-1R inhibition 109. Sallman, D. A. et al. The first-in-class anti-CD47
macrophages provide a pro-tumorigenic niche to in gliomas. Science 352, aad3018 (2016). antibody magrolimab (5F9) in combination with
early NSCLC cells. Nature 595, 578–584 (2021). 87. Martin, T. D. et al. The adaptive immune system azacitidine is effective in MDS and AML patients:
62. Loyher, P. L. et al. Macrophages of distinct origins is a major driver of selection for tumor suppressor ongoing phase 1b results [abstract]. Blood 134
contribute to tumor development in the lung. gene inactivation. Science 373, 1327–1335 (2021). (Suppl. 1), 569 (2019).
J. Exp. Med. 215, 2536–2553 (2018). 88. Cassetta, L. & Pollard, J. W. Targeting macrophages: 110. Fisher, G. A. et al. A phase Ib/II study of the anti-CD47
63. Zhang, N. et al. LYVE1+ macrophages of murine therapeutic approaches in cancer. Nat. Rev. Drug antibody magrolimab with cetuximab in solid
peritoneal mesothelium promote omentum-independent Discov. 17, 887–904 (2018). tumor and colorectal cancer patients [abstract].
ovarian tumor growth. J. Exp. Med. 218, e20210924 89. Bonapace, L. et al. Cessation of CCL2 inhibition J. Clin. Oncol. 38 (Suppl. 4), 114 (2020).
(2021). accelerates breast cancer metastasis by promoting 111. Upton, R. et al. Combining CD47 blockade with
64. Zhu, Y. et al. Tissue-resident macrophages in angiogenesis. Nature 515, 130–133 (2014). trastuzumab eliminates HER2-positive breast cancer
pancreatic ductal adenocarcinoma originate from 90. Sandhu, S. K. et al. A first-in-human, first-in-class, cells and overcomes trastuzumab tolerance. Proc. Natl
embryonic hematopoiesis and promote tumor phase I study of carlumab (CNTO 888), a human Acad. Sci. USA 118, e2026849118 (2021).
progression. Immunity 47, 323–338.e6 (2017). monoclonal antibody against CC-chemokine ligand 2 112. Gilead. Gilead announces partial clinical hold for
65. Cortez-Retamozo, V. et al. Origins of tumor-associated in patients with solid tumors. Cancer Chemother. studies evaluating magrolimab in combination with
macrophages and neutrophils. Proc. Natl Acad. Sci. Pharmacol. 71, 1041–1050 (2013). azacitidine. GILEAD https://www.gilead.com/news-
USA 109, 2491–2496 (2012). 91. Nywening, T. M. et al. Targeting tumour-associated and-press/press-room/press-releases/2022/1/gilead-
66. Cortez-Retamozo, V. et al. Angiotensin II drives macrophages with CCR2 inhibition in combination with announces-partial-clinical-hold-for-studies-evaluating-
the production of tumor-promoting macrophages. FOLFIRINOX in patients with borderline resectable and magrolimab-in-combination-with-azacitidine
Immunity 38, 296–308 (2013). locally advanced pancreatic cancer: a single-centre, (2022).
67. Movahedi, K. et al. Different tumor microenvironments open-label, dose-finding, non-randomised, phase 1b 113. Deczkowska, A., Weiner, A. & Amit, I. The physiology,
contain functionally distinct subsets of macrophages trial. Lancet Oncol. 17, 651–662 (2016). pathology, and potential therapeutic applications of
derived from Ly6C(high) monocytes. Cancer Res. 70, 92. Linehan, D. et al. Overall survival in a trial of orally the TREM2 signaling pathway. Cell 181, 1207–1217
5728–5739 (2010). administered CCR2 inhibitor CCX872 in locally (2020).
68. Qian, B. Z. et al. CCL2 recruits inflammatory advanced/metastatic pancreatic cancer: correlation 114. Shirotani, K. et al. Aminophospholipids are
monocytes to facilitate breast-tumour metastasis. with blood monocyte counts [abstract]. J. Clin. Oncol. signal-transducing TREM2 ligands on apoptotic cells.
Nature 475, 222–225 (2011). 36 (Suppl. 5), 92 (2018). Sci. Rep. 9, 7508 (2019).
69. Friebel, E. et al. Single-cell mapping of human 93. Jiao, X. et al. CCR5 governs DNA damage repair and 115. Molgora, M. et al. TREM2 modulation remodels
brain cancer reveals tumor-specific instruction of breast cancer stem cell expansion. Cancer Res. 78, the tumor myeloid landscape enhancing anti-PD-1
tissue-invading leukocytes. Cell 181, 1626–1642.e20 1657–1671 (2018). immunotherapy. Cell 182, 886–900.e17 (2020).
(2020). 94. Halama, N. et al. Tumoral immune cell exploitation 116. Katzenelenbogen, Y. et al. Coupled scRNA-seq
70. Guldner, I. H. et al. CNS-native myeloid cells drive in colorectal cancer metastases can be targeted and intracellular protein activity reveal an
immune suppression in the brain metastatic niche effectively by anti-CCR5 therapy in cancer patients. immunosuppressive role of TREM2 in cancer. Cell
through Cxcl10. Cell 183, 1234–1248.e25 (2020). Cancer Cell 29, 587–601 (2016). 182, 872–885.e19 (2020).
71. Klemm, F. et al. Compensatory CSF2-driven 95. Liu, J. et al. High expression of CCR5 in melanoma 117. Binnewies, M. et al. Targeting TREM2 on
macrophage activation promotes adaptive resistance enhances epithelial-mesenchymal transition and tumor-associated macrophages enhances
to CSF1R inhibition in breast-to-brain metastasis. metastasis via TGFβ1. J. Pathol. 247, 481–493 immunotherapy. Cell Rep. 37, 109844 (2021).
Nat. Cancer 2, 1086–1101 (2021). (2019). 118. Klesney-Tait, J., Turnbull, I. R. & Colonna, M.
72. Bruni, D., Angell, H. K. & Galon, J. The immune 96. Aldinucci, D., Borghese, C. & Casagrande, N. The The TREM receptor family and signal integration.
contexture and immunoscore in cancer prognosis and CCL5/CCR5 axis in cancer progression. Cancers 12, Nat. Immunol. 7, 1266–1273 (2006).
therapeutic efficacy. Nat. Rev. Cancer 20, 662–680 1765 (2020). 119. Fontana, R. et al. Nuclear receptor ligands induce
(2020). 97. CytoDyn. CytoDyn announces cancer update: 12-month TREM-1 expression on dendritic cells: analysis of
73. Andreatta, M. et al. Interpretation of T cell states analysis of 28 mTNBC patients receiving leronlimab their role in tumors. Oncoimmunology 8, 1554967
from single-cell transcriptomics data using reference suggests an increase of 3600% in 12-month OS in (2019).
atlases. Nat. Commun. 12, 2965 (2021). 75% of patients with a lower level of circulating cells 120. Kuemmel, A. et al. Soluble triggering receptor
74. Gerhard, G. M. et al. Tumor-infiltrating dendritic after leronlimab induction or at baseline; 12-month expressed on myeloid cells 1 in lung cancer. Sci. Rep.
cell states are conserved across solid human cancers. PFS continues at near 600% increase. CytoDyn 8, 10766 (2018).
J. Exp. Med. 218, e20200264 (2021). https://www.cytodyn.com/newsroom/press-releases/ 121. Wu, J. et al. The proinflammatory myeloid cell
75. Mulder, K. et al. Cross-tissue single-cell landscape detail/574/cytodyn-announces-cancer-update- receptor TREM-1 controls Kupffer cell activation and
of human monocytes and macrophages in health 12-month-analysis-of-28 (2021). development of hepatocellular carcinoma. Cancer Res.
and disease. Immunity 54, 1883–1900.e5 (2021). 98. Guo, F. et al. CXCL12/CXCR4: a symbiotic bridge 72, 3977–3986 (2012).
76. Luca, B. A. et al. Atlas of clinically distinct cell states linking cancer cells and their stromal neighbors in 122. Barkal, A. A. et al. CD24 signalling through
and ecosystems across human solid tumors. Cell 184, oncogenic communication networks. Oncogene 35, macrophage Siglec-10 is a target for cancer
5482–5496.e28 (2021). 816–826 (2016). immunotherapy. Nature 572, 392–396 (2019).
77. Lin, E. Y. et al. Colony-stimulating factor 1 promotes 99. Hughes, R. et al. Perivascular M2 macrophages 123. Klapdor, R. et al. Characterization of a novel
progression of mammary tumors to malignancy. stimulate tumor relapse after chemotherapy. third-generation anti-CD24-CAR against ovarian
J. Exp. Med. 193, 727–740 (2001). Cancer Res. 75, 3479–3491 (2015). cancer. Int. J. Mol. Sci. 20, 660 (2019).
78. Ries, C. H. et al. Targeting tumor-associated 100. Seo, Y. D. et al. Mobilization of CD8+ T cells via CXCR4 124. Maliar, A. et al. Redirected T cells that target
macrophages with anti-CSF-1R antibody reveals a blockade facilitates PD-1 checkpoint therapy in human pancreatic adenocarcinoma antigens eliminate tumors
strategy for cancer therapy. Cancer Cell 25, 846–859 pancreatic cancer. Clin. Cancer Res. 25, 3934–3945 and metastases in mice. Gastroenterology 143,
(2014). (2019). 1375–1384.e5 (2012).
79. Pfirschke, C. et al. Macrophage-targeted therapy 101. Bockorny, B. et al. BL-8040, a CXCR4 antagonist, in 125. De Henau, O. et al. Overcoming resistance to
unlocks antitumoral crosstalk between IFN-𝛾-secreting combination with pembrolizumab and chemotherapy checkpoint blockade therapy by targeting PI3Kγ
lymphocytes and IL-12-producing dendritic cell. for pancreatic cancer: the COMBAT trial. Nat. Med. in myeloid cells. Nature 539, 443–447 (2016).
Cancer Immunol. Res. 10, 40–55 (2021). 26, 878–885 (2020). 126. Kaneda, M. M. et al. PI3Kγ is a molecular switch that
80. Pyonteck, S. M. et al. CSF-1R inhibition alters 102. & Thomas, R. P. et al. CXCR4 blockade at the end controls immune suppression. Nature 539, 437–442
macrophage polarisation and blocks glioma of irradiation to improve local control of glioblastoma (2016).
progression. Nat. Med. 19, 1264–1272 (2013). (GBM) [abstract]. J. Clin. Oncol. 36 (Suppl. 15), 2019 127. Furman, R. R. et al. Idelalisib and rituximab in
81. Cassier, P. A. et al. CSF1R inhibition with (2018). relapsed chronic lymphocytic leukemia. N. Engl. J.
emactuzumab in locally advanced diffuse-type 103. Hidalgo, M. et al. A multi-center phase 2a trial Med. 370, 997–1007 (2014).
tenosynovial giant cell tumours of the soft tissue: of the CXCR4 inhibitor motixafortide (BL-8040)(M) 128. Vonderheide, R. H. The immune revolution: a case for
a dose-escalation and dose-expansion phase 1 study. in combination with pembrolizumab (P) and priming, not checkpoint. Cancer Cell 33, 563–569
Lancet Oncol. 16, 949–956 (2015). chemotherapy (C), in patients with metastatic (2018).

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 419

0123456789();:
Reviews

129. Beatty, G. L. et al. CD40 agonists alter tumor stroma macrophage response on tumor cells that have shut xenograft models of nasopharyngeal carcinoma
and show efficacy against pancreatic carcinoma in down tumor antigen expression. Cancer Res. 71, [abstract]. Cancer Res. 81 (Suppl. 13), 948 (2021).
mice and humans. Science 331, 1612–1616 (2011). 5697–5706 (2011). 174. Kamber, R. A. et al. Inter-cellular CRISPR screens
130. Kashyap, A. S. et al. Optimized antiangiogenic 151. Chmielewski, M. & Abken, H. CAR T cells releasing reveal regulators of cancer cell phagocytosis. Nature
reprogramming of the tumor microenvironment IL-18 convert to T-bethigh FoxO1low effectors that 597, 549–554 (2021).
potentiates CD40 immunotherapy. Proc. Natl Acad. exhibit augmented activity against advanced solid 175. Zhai, K. et al. Pharmacological inhibition of BACE1
Sci. USA 117, 541–551 (2020). tumors. Cell Rep. 21, 3205–3219 (2017). suppresses glioblastoma growth by stimulating
131. Garris, C. S. et al. Successful anti-PD-1 cancer 152. Kuhn, N. F. et al. CD40 ligand-modified chimeric macrophage phagocytosis of tumor cells. Nat. Cancer
immunotherapy requires T cell-dendritic cell crosstalk antigen receptor T cells enhance antitumor function 2, 1136–1151 (2021).
involving the cytokines IFN-γ and IL-12. Immunity 49, by eliciting an endogenous antitumor response. 176. Klug, F. et al. Low-dose irradiation programs
1148–1161.e7 (2018). Cancer Cell 35, 473–488.e6 (2019). macrophage differentiation to an iNOS+/M1
132. Siwicki, M. et al. Resident Kupffer cells and neutrophils 153. De Palma, M. et al. Tumor-targeted interferon-α phenotype that orchestrates effective T cell
drive liver toxicity in cancer immunotherapy. delivery by Tie2-expressing monocytes inhibits tumor immunotherapy. Cancer Cell 24, 589–602 (2013).
Sci. Immunol. 6, eabi7083 (2021). growth and metastasis. Cancer Cell 14, 299–311 177. Virtakoivu, R. et al. Systemic blockade of Clever-1
133. Dahan, R. et al. Therapeutic activity of agonistic, (2008). elicits lymphocyte activation alongside checkpoint
human anti-CD40 monoclonal antibodies requires 154. Klichinsky, M. et al. Human chimeric antigen molecule downregulation in patients with solid
selective FcγR engagement. Cancer Cell 29, 820–831 receptor macrophages for cancer immunotherapy. tumors: results from a phase I/II clinical trial. Clin.
(2016). Nat. Biotechnol. 38, 947–953 (2020). Cancer Res. 27, 4205–4220 (2021).
134. Machiels, J. P. et al. Phase Ib study of anti-CSF-1R 155. CARISMA Therapeutics. CARISMA Therapeutics 178. Bates, S. E. Epigenetic therapies for cancer. N. Engl. J.
antibody emactuzumab in combination with CD40 announces U.S. Food and Drug Administration grants Med. 383, 650–663 (2020).
agonist selicrelumab in advanced solid tumor patients. fast track designation to CT-0508 for the treatment 179. Guerriero, J. L. et al. Class IIa HDAC inhibition reduces
J. Immunother. Cancer 8, e001153 (2020). of patients with solid tumors. CISION https://prn.to/ breast tumours and metastases through anti-tumour
135. Grilley-Olson, J. E. et al. SEA-CD40, a non-fucosylated 3kvshij (2021). macrophages. Nature 543, 428–432 (2017).
CD40 agonist: interim results from a phase 1 study in 156. Zhang, W. et al. Chimeric antigen receptor 180. Burgess, M. et al. HDAC7 is an actionable driver of
advanced solid tumors [abstract]. J. Clin. Oncol. 36 macrophage therapy for breast tumours mediated therapeutic antibody resistance by macrophages from
(Suppl. 15), 3093 (2018). by targeting the tumour extracellular matrix. Br. J. CLL patients. Oncogene 39, 5756–5767 (2020).
136. O’Hara, M. H. et al. CD40 agonistic monoclonal Cancer 121, 837–845 (2019). 181. Georgoudaki, A. M. et al. Reprogramming
antibody APX005M (sotigalimab) and chemotherapy, 157. Kaczanowska, S. et al. Genetically engineered myeloid tumor-associated macrophages by antibody targeting
with or without nivolumab, for the treatment of cells rebalance the core immune suppression program inhibits cancer progression and metastasis. Cell Rep.
metastatic pancreatic adenocarcinoma: an open-label, in metastasis. Cell 184, 2033–2052.e21 (2021). 15, 2000–2011 (2016).
multicentre, phase 1b study. Lancet Oncol. 22, 158. Zhang, L. et al. Pluripotent stem cell-derived 182. Eisinger, S. et al. Targeting a scavenger receptor on
118–131 (2021). CAR-macrophage cells with antigen-dependent tumor-associated macrophages activates tumor cell
137. Irenaeus, S. M. M. et al. First-in-human study with anti-cancer cell functions. J. Hematol. Oncol. 13, 153 killing by natural killer cells. Proc. Natl Acad. Sci. USA
intratumoral administration of a CD40 agonistic (2020). 117, 32005–32016 (2020).
antibody, ADC-1013, in advanced solid malignancies. 159. Kang, M. et al. Nanocomplex-mediated in vivo 183. La Fleur, L. et al. Targeting MARCO and IL37R on
Int. J. Cancer 145, 1189–1199 (2019). programming to chimeric antigen receptor-M1 immunosuppressive macrophages in lung cancer blocks
138. Garris, C. S. et al. Dendritic cell targeting with macrophages for cancer therapy. Adv. Mater. 33, regulatory T cells and supports cytotoxic lymphocyte
Fc-enhanced CD40 antibody agonists induces durable e2103258 (2021). function. Cancer Res. 81, 956–967 (2021).
antitumor immunity in humanized mouse models of 160. Sloas, C., Gill, S. & Klichinsky, M. Engineered 184. Lam, K. C. et al. Microbiota triggers STING-type I
bladder cancer. Sci. Transl. Med. 13, eabd1346 (2021). CAR-macrophages as adoptive immunotherapies for IFN-dependent monocyte reprogramming of the
139. Labiano, S. et al. CD40 agonist targeted to fibroblast solid tumors. Front. Immunol. 12, 783305 (2021). tumor microenvironment. Cell 184, 5338–5356.e21
activation protein α synergizes with radiotherapy 161. Andtbacka, R. H. et al. Talimogene laherparepvec (2021).
in murine HPV-positive head and neck tumors. improves durable response rate in patients with 185. Spencer, C. N. et al. Dietary fiber and probiotics
Clin. Cancer Res. 27, 4054–4065 (2021). advanced melanoma. J. Clin. Oncol. 33, 2780–2788 influence the gut microbiome and melanoma
140. Rodell, C. B. et al. TLR7/8-agonist-loaded (2015). immunotherapy response. Science 374, 1632–1640
nanoparticles promote the polarisation of 162. Ramelyte, E. et al. Oncolytic virotherapy-mediated (2021).
tumour-associated macrophages to enhance cancer anti-tumor response: a single-cell perspective. 186. Gopalakrishnan, V. et al. Gut microbiome modulates
immunotherapy. Nat. Biomed. Eng. 2, 578–588 Cancer Cell 39, 394–406.e4 (2021). response to anti-PD-1 immunotherapy in melanoma
(2018). 163. Chiocca, E. A. et al. Regulatable interleukin-12 gene patients. Science 359, 97–103 (2018).
141. Geisse, J. et al. Imiquimod 5% cream for the therapy in patients with recurrent high-grade glioma: 187. Irmisch, A. et al. The Tumor Profiler Study: integrated,
treatment of superficial basal cell carcinoma: results results of a phase 1 trial. Sci. Transl. Med. 11, multi-omic, functional tumor profiling for clinical
from two phase III, randomized, vehicle-controlled eaaw5680 (2019). decision support. Cancer Cell 39, 288–293 (2021).
studies. J. Am. Acad. Dermatol. 50, 722–733 (2004). 164. Desjardins, A. et al. Recurrent glioblastoma treated 188. Maas, R. R. et al. An integrated pipeline for
142. Hilf, N. et al. Actively personalized vaccination trial for with recombinant poliovirus. N. Engl. J. Med. 379, comprehensive analysis of immune cells in human
newly diagnosed glioblastoma. Nature 565, 240–245 150–161 (2018). brain tumor clinical samples. Nat. Protoc. 16,
(2019). 165. Samson, A. et al. Intravenous delivery of oncolytic 4692–4721 (2021).
143. Migliorini, D. et al. Phase I/II trial testing safety reovirus to brain tumor patients immunologically 189. Oh, J. et al. Rapid serial immunoprofiling of the tumor
and immunogenicity of the multipeptide IMA950/ primes for subsequent checkpoint blockade. immune microenvironment by fine needle sampling.
poly-ICLC vaccine in newly diagnosed adult malignant Sci. Transl. Med. 10, eaam7577 (2018). Clin. Cancer Res. 27, 4781–4793 (2021).
astrocytoma patients. Neuro Oncol. 21, 923–933 166. Cohn, D. E. et al. Randomized phase IIB evaluation 190. Voabil, P. et al. An ex vivo tumor fragment platform
(2019). of weekly paclitaxel versus weekly paclitaxel with to dissect response to PD-1 blockade in cancer.
144. Okada, H. et al. Induction of CD8+ T-cell responses oncolytic reovirus (Reolysin®) in recurrent ovarian, Nat. Med. 27, 1250–1261 (2021).
against novel glioma-associated antigen peptides and tubal, or peritoneal cancer: an NRG Oncology/ 191. Wensink, G. E. et al. Patient-derived organoids as a
clinical activity by vaccinations with α-type 1 polarized Gynecologic Oncology Group study. Gynecol. Oncol. predictive biomarker for treatment response in cancer
dendritic cells and polyinosinic-polycytidylic acid 146, 477–483 (2017). patients. NPJ Precis. Oncol. 5, 30 (2021).
stabilized by lysine and carboxymethylcellulose in 167. Xu, B. et al. An oncolytic virus expressing a full-length 192. Kim, H. R. et al. Mouse-human co-clinical trials
patients with recurrent malignant glioma. J. Clin. antibody enhances antitumor innate immune response demonstrate superior anti-tumour effects of buparlisib
Oncol. 29, 330–336 (2011). to glioblastoma. Nat. Commun. 12, 5908 (2021). (BKM120) and cetuximab combination in squamous
145. Ribas, A. et al. Overcoming PD-1 blockade resistance 168. De Palma, M. & Naldini, L. Angiopoietin-2 TIEs up cell carcinoma of head and neck. Br. J. Cancer 123,
with CpG-A Toll-like receptor 9 agonist vidutolimod in macrophages in tumor angiogenesis. Clin. Cancer Res. 1720–1729 (2020).
patients with metastatic melanoma. Cancer Discov. 17, 5226–5232 (2011). 193. Ignatiadis, M., Sledge, G. W. & Jeffrey, S. S. Liquid
https://doi.org/10.1158/2159-8290.CD-21-0425 169. Yu, X. et al. Suppression of KSHV-induced biopsy enters the clinic – implementation issues and
(2021). angiopoietin-2 inhibits angiogenesis, infiltration future challenges. Nat. Rev. Clin. Oncol. 18, 297–312
146. Sun, L. et al. Activating a collaborative innate-adaptive of inflammatory cells, and tumor growth. Cell Cycle (2021).
immune response to control metastasis. Cancer Cell 15, 2053–2065 (2016). 194. Taube, J. M. et al. The Society for Immunotherapy
39, 1361–1374.e9 (2021). 170. Vergote, I. et al. Trebananib or placebo plus of Cancer statement on best practices for multiplex
147. Rodriguez-Garcia, A. et al. CAR-T cell-mediated carboplatin and paclitaxel as first-line treatment for immunohistochemistry (IHC) and immunofluorescence
depletion of immunosuppressive tumor-associated advanced ovarian cancer (TRINOVA-3/ENGOT-ov2/ (IF) staining and validation. J. Immunother. Cancer 8,
macrophages promotes endogenous antitumor GOG-3001): a randomised, double-blind, phase 3 e000155 (2020).
immunity and augments adoptive immunotherapy. trial. Lancet Oncol. 20, 862–876 (2019). 195. Ji, A. L. et al. Multimodal analysis of composition
Nat. Commun. 12, 877 (2021). 171. Kloepper, J. et al. Ang-2/VEGF bispecific antibody and spatial architecture in human squamous cell
148. Johnson, L. R. et al. The immunostimulatory RNA reprograms macrophages and resident microglia carcinoma. Cell 182, 497–514.e22 (2020).
RN7SL1 enables CAR-T cells to enhance autonomous to anti-tumor phenotype and prolongs glioblastoma 196. Rao, A. et al. Exploring tissue architecture using spatial
and endogenous immune function. Cell 184, survival. Proc. Natl Acad. Sci. USA 113, 4476–4481 transcriptomics. Nature 596, 211–220 (2021).
4981–4995.e14 (2021). (2016). 197. Bera, K. et al. Artificial intelligence in digital
149. Spear, P. et al. Chimeric antigen receptor 172. Kienast, Y. et al. Ang-2-VEGF-A CrossMab, a novel pathology – new tools for diagnosis and precision
T cells shape myeloid cell function within the tumor bispecific human IgG1 antibody blocking VEGF-A oncology. Nat. Rev. Clin. Oncol. 16, 703–715 (2019).
microenvironment through IFN-γ and GM-CSF. and Ang-2 functions simultaneously, mediates potent 198. Pai, S. I. et al. New technology on the horizon: fast
J. Immunol. 188, 6389–6398 (2012). antitumor, antiangiogenic, and antimetastatic efficacy. analytical screening technique FNA (FAST-FNA)
150. Chmielewski, M. et al. IL-12 release by engineered Clin. Cancer Res. 19, 6730–6740 (2013). enables rapid, multiplex biomarker analysis in head
T cells expressing chimeric antigen receptors can 173. Wong, E. C. et al. Preclinical evaluation of VEGF Ang2 and neck cancers. Cancer Cytopathol. 128, 782–791
effectively muster an antigen-independent bispecific nanobody BI836880 in patient-derived (2020).

420 | June 2022 | volume 19 www.nature.com/nrclinonc

0123456789();:
Reviews

199. Wculek, S. K. et al. Dendritic cells in cancer 209. Cloughesy, T. F. et al. Neoadjuvant anti- ImmuneOncia, KSQ Therapeutics, Merck, Siamab
immunology and immunotherapy. Nat. Rev. Immunol. PD-1immunotherapy promotes a survival benefit Therapeutics, Third Rock Ventures and Tidal. O.M. has been
20, 7–24 (2020). with intratumoral and systemic immune responses a consultant for Amgen, BMS, GSK, MSD, Novartis, Pierre
200. Siwicki, M. & Pittet, M. J. Versatile neutrophil in recurrent glioblastoma. Nat. Med. 25, 477–486 Fabre and Roche, and has received research support from
functions in cancer. Semin. Immunol. https://doi.org/ (2019). Amgen, BMS, MSD, Neracare and Pierre Fabre. D.M. is an
10.1016/j.smim.2021.101538 (2021). inventor on patents related to CAR T cell therapy, filed by the
201. Coffelt, S. B., Wellenstein, M. D. & de Visser, K. E. Acknowledgements University of Pennsylvania and the University of Geneva, and
Neutrophils in cancer: neutral no more. Nat. Rev. M.J.P. receives support from the ISREC Foundation, Ludwig has been a consultant for Limula Therapeutics and MPC
Cancer 16, 431–446 (2016). Cancer Research, and NIH grants P01-CA240239 and Therapeutics.
202. Derakhshani, A. et al. Mast cells: a double-edged R01-CA218579. O.M. receives support from the ISREC
sword in cancer. Immunol. Lett. 209, 28–35 (2019). Foundation, the Ludwig Cancer Research, the Swiss National Peer review information
203. Khazaie, K. et al. The significant role of mast Science Foundation (SNSF) 31003A_176168, the Nature Reviews Clinical Oncology thanks L. Akkari and the
cells in cancer. Cancer Metastasis Rev. 30, 45–60 Swiss National Cancer Ligue KFS-5034-02-2020, the Swiss other, anonymous, reviewer(s) for their contribution to
(2011). Personalized Health Network (SPHN), and the Personalized the peer review of this work.
204. Geirsdottir, L. et al. Cross-species single-cell analysis Health and Related Technologies (PHRT). D.M. receives sup-
reveals divergence of the primate microglia program. port from the ISREC Foundation, Association Frédéric Fellay, Publisher’s note
Cell 179, 1609–1622.e16 (2019). Ernst and Lucie Schmidheiny Foundation, Fondation Dr Henri Springer Nature remains neutral with regard to jurisdictional
205. Evren, E. et al. Distinct developmental pathways Dubois-Ferrière Dinu Lipatti (DFDL), Ligue Genevoise contre claims in published maps and institutional affiliations.
from blood monocytes generate human lung le cancer, Personalized Health and Related Technologies
macrophage diversity. Immunity 54, 259–275.e7 (PHRT) ETH fund, Swiss Bridge Foundation, and Swiss Supplementary information
(2021). Innovation Agency (Innosuisse). The online version contains supplementary material available
206. Rongvaux, A. et al. Development and function of at https://doi.org/10.1038/s41571-022-00620-6.
human innate immune cells in a humanized mouse Author contributions
model. Nat. Biotechnol. 32, 364–372 (2014). All the authors contributed to all aspects of preparation
207. Tuveson, D. & Clevers, H. Cancer modeling meets of this manuscript. Related links
human organoid technology. Science 364, 952–955 ClinicalTrials.gov: https://clinicaltrials.gov/
(2019). Competing interests
208. Yuki, K. et al. Organoid models of tumor immunology. M.J.P. has been a consultant for Aileron Therapeutics,
Trends Immunol. 41, 652–664 (2020). AstraZeneca, Cygnal Therapeutics, Elstar Therapeutics, © Springer Nature Limited 2022, corrected publication 2022

nATuRe RevIeWS | ClinicAl Oncology volume 19 | June 2022 | 421

0123456789();:

You might also like