You are on page 1of 14

RevIewS

B cells, plasma cells and antibody


repertoires in the tumour
microenvironment
George V. Sharonov   1,2,3, Ekaterina O. Serebrovskaya   2,3, Diana V. Yuzhakova   1,
Olga V. Britanova   1,2,3 and Dmitriy M. Chudakov   1,2,3,4*
Abstract | Recent data show that B cells and plasma cells located in tumours or in tumour-draining
lymph nodes can have important roles in shaping antitumour immune responses. In tumour-
associated tertiary lymphoid structures, T cells and B cells interact and undergo cooperative
selection, specialization and clonal expansion. Importantly, B cells can present cognate tumour-
derived antigens to T cells, with the functional consequences of such interactions being shaped
by the B cell phenotype. Furthermore, the isotype and specificity of the antibodies produced by
plasma cells can drive distinct immune responses. Here we summarize our current knowledge
of the roles of B cells and antibodies in the tumour microenvironment. Moreover, we discuss
the potential of using immunoglobulin repertoires as a source of tumour-specific receptors for
immunotherapy or as biomarkers to predict the efficacy of immunotherapeutic interventions.

The importance of T cells in tumour immunosurveil- repertoires that are found in the tumour microenviron-
lance is well established1–4, but the relative contribution ment and consider their potential roles as prognostic or
of B cells has been studied to a much lesser extent, predictive markers and as agents and targets of cancer
and mostly only during the past decade. Compared immunotherapies.
with T cells, relatively few B cells are usually found in
tumour infiltrates5–8; however, several studies indicate Tumour-infiltrating B cells
that their presence and functionality can be consid- B cells may concentrate at tumour margins7,30 or form
ered as an important prognostic factor in cancer9–19. tumour-associated immune aggregates of various com-
Furthermore, plasma cells are present in tumour infil- plexity, ranging from small unorganized clusters to
trates, and even low counts of these cells are able to structured TLS18,27 (Box 1). The cellular composition of
produce large amounts of cytokines20 and antibodies. such structures can differ considerably depending on the
1
Laboratory of Genomics of
These antibodies can promote antitumour immunity stage and origin of the tumour, as well as other factors,
Antitumor Adaptive Immunity,
Privolzhsky Research Medical
by driving antibody-dependent cellular cytotoxicity and has prognostic value in various metastatic and pri-
University, Nizhny Novgorod, (ADCC) and phagocytosis21,22, complement activa- mary tumours29. In particular, antigen-specific interac-
Russia. tion and enhancing antigen presentation by dendritic tions between T cells and B cells in the TLS and in less
2
Genomics of Adaptive cells23. Moreover, B cells themselves can present anti- organized clusters of tumour-infiltrating lymphocytes
Immunity Department, gens to CD4 + and CD8 + T cells and thereby shape seem to be crucial, and the protective role of T cells in the
Shemyakin and Ovchinnikov
antigen-specific immune responses within the tumour tumour microenvironment often depends on coopera-
Institute of Bioorganic
Chemistry, Moscow, Russia.
microenvironment24–26. B cells may also contribute to tion with B cells. The presence of CD20+ B cells close to
3
Institute of Translational
the formation of tumour-associated tertiary lymphoid CD8+ T cells31 and the colocalization of B cells and T cells
Medicine, Pirogov Russian structures (TLS), which support the further matura- in breast cancer, in malignant melanoma and within
National Research Medical tion and isotype switching of tumour-specific B cells intraepithelial infiltrates from ovarian cancer have
University, Moscow, Russia. as well as the development of tumour-specific T cell been found to be a positive prognostic marker12,32–34.
4
Center of Life Sciences, responses27–29. Elimination of B cells was shown to suppress the ability
Skolkovo Institute of Science At the same time, tumour-infiltrating B cells may of an anti-GITR antibody to activate an antitumour T cell
and Technology, Moscow,
Russia.
exert both protumour and antitumour effects depend- response in a mouse model of colorectal and breast car-
ing on the composition of the tumour microenviron- cinoma and of a CD73 inhibitor to activate T helper 1
*e-mail: chudakovdm@
gmail.com ment and on the phenotypes of B cells present and the (TH1) and TH17 cells in melanoma35,36. Furthermore,
https://doi.org/10.1038/ antibodies that they produce (Figs 1,2; Supplementary adoptive co-transfer of ex vivo activated B cells and
s41577-019-0257-x Table 1). In this Review, we focus on the B cell and antibody T cells in the 4T1 mouse model of breast cancer achieved

Nature Reviews | Immunology


Reviews

Tumour antigen

IgG1 antibody Neutral cell


FcγR
TAM Tumour-attacking cell

TH1 cell
IFNγ
NK cell
B cell

‘M1-like’ Granzyme B
macrophage ADCC
CD4 +

T cell IgG1 B cell/


MHC
plasmablast/
class II
plasma cell
Phagocytosis
IFNγ, Complement
IL-12

TCR
MHC
class I
CD8+
B cell T cell
IFNγ,
IL-12

CD8+
T cell
Tumour cells

Fig. 1 | Antitumour roles of tumour-infiltrating B cells and intratumourally produced antibodies. B cells and
plasma cells can support antitumour immune responses through several mechanisms. Plasma cell secretion of tumour
cell-specific IgG1 antibodies can mediate antibody-dependent cell cytotoxicity (ADCC) and phagocytosis of tumour cells.
B cells also participate in the presentation of tumour-derived antigens to CD4+ and CD8+ T cells. First, they can directly
present tumour-associated antigens they have captured via B cell receptors. Second, produced antibodies can support
the uptake of tumour antigens by tumour-associated macrophages (TAMs) and dendritic cells. In addition, B cells may
promote antitumour immunity through the release of cytokines that drive cytotoxic immune responses, such as IFNγ and
IL-12. B cells can also directly attack tumour cells using granzyme B and TRAIL (also known as TNFSF10). Immune cells with
antitumour potential are coloured red. FcγR , Fcγ receptor ; NK , natural killer ; TCR , T cell receptor ; TH1 cell, T helper 1 cell.

better tumour regression than the transfer of either also suggests their active participation in the antitumour
population alone37. response14,18,29,31,40–43.
The ability of long-lived B cells to serve as antigen- Analysis of RNA sequencing (RNA-seq) data from
presenting cells (APCs)38 can essentially explain the pos- The Cancer Genome Atlas (TCGA) database revealed that
itive outcome of intratumoural B cell–T cell cooperation. high levels of expression of B cell and plasma cell sig-
Human ovarian and liver cancer samples contain atypical nature genes correlated with increased overall survival
populations of mature CD27−IgG+ memory B cells that in patients with melanoma, lung adenocarcinoma, pan-
express surface markers associated with APCs (namely creatic adenocarcinoma, and head and neck squamous
MHC class II, CD40, CD80 and CD86) and colocalize cell carcinoma. By contrast, high levels of expression of
with CD8+ T cells30,32. Although dendritic cells are the these genes correlated with poorer clinical outcomes
major APCs that provide initial T cell activation in in patients with glioblastoma and clear cell renal cell
The Cancer Genome Atlas
(TCGA) database the  lymph nodes, B cells can maintain additional carcinoma. High levels of immunoglobulin expression
The most comprehensive T cell population expansion intratumourally by acting in tumours is also associated with increased survival in
cancer genomics database; as APCs25,26, or through CD27–CD70 interactions39, patients with melanoma42–44, in the proximal proliferative
it contains multiple types of which is important during prolonged inflammatory subtype of lung adenocarcinoma45 and in head and neck
genomic data with histological
information and clinical records
response. The evidence that TLS-located B cells may squamous cell carcinoma, but it is associated with poor
for more than 11,000 patients undergo clonal expansion, somatic hypermutation, iso- prognosis in patients with glioblastoma and renal cell
and 33 cancer types. type switching and tumour-specific antibody production carcinoma44. These findings are consistent with studies

www.nature.com/nri
Reviews

Cryptic peptide antigens


associating plasma cell infiltration with longer survival in include overexpressed or aberrantly expressed self anti-
Antigens that originate from patients with melanoma and non-small-cell lung carci- gens, unconventionally modified proteins and normal
translation of sequences noma13,46. In breast cancer, reports on tumour-infiltrating intracellular molecules. In addition, neoantigens and
outside annotated open B cells and plasma cells are to some extent contradic- cryptic peptide antigens that can drive a humoral immune
reading frames. They may
tory, with some studies suggesting association with response have been identified from immunoscreening of
derive from non-annotated
open reading frames, poor prognosis19,47 and some studies suggesting associ- cDNA libraries from tumour tissues49,50. Such antigens
non-coding genomic regions, ation with better prognosis33 (Supplementary Table 1). may originate from a frameshift or replacement muta-
alternative start codons, A meta-analysis of non-TCGA transcriptomic data from tion, an alternative start codon or alternative splicing
frameshift mutations,
~18,000 human tumours found that the relative abun- events50–52. Tumour-specific and self-specific antibodies
alternative splicing or
ribosomal frameshifting.
dance of intratumoural plasma cells is one of the most have been detected in serum from patients with cancer in
Protein splicing and significant positive prognostic factors for patient survival numerous studies53–58. Furthermore, estimates of the lev-
post-translational modifications in the case of various non-brain solid tumours, except for els of serum autoantibodies against tumour-associated
can also be classified as cryptic large-cell lung carcinoma. On the other hand, memory and self antigens have been proposed for early detection
peptide antigens.
B cells had negative prognostic value for lung squamous of cancer59–63. At later stages of disease, serum antibodies
cell carcinoma, colon cancer and gastric cancer48. can serve as potent prognostic markers56,57,64–70.
One well-known tumour-associated antigen, mucin 1
Tumour-specific antibodies (MUC1), is overexpressed in tumours in an underglyco-
In both the serum and the tumour microenvironment, sylated form71–73. Antibodies against MUC1 are detected
antibodies are frequently found that recognize a broad in serum from patients at early stages of pancreatic, breast,
array of tumour-expressed and self antigens. These gastric, lung and ovarian cancer and serve as a marker

CD4+ TAM
Treg cell
T cell

Tumour cells IL-10


TGFβ
TGFβ

IgA B cell/ IgA


B cell plasmablast/
plasma cell
‘M2-like’
IL-10, macrophage
IL-35
PDL1
PD1
TGFβ IL-10 Effector
Tumour- T cell
associated
neutrophil

Neutral cell
FcR
PMN-MDSC
IgG1 Tumour-
attacking cell

Immunosuppressive
cell
Bispecific IgG4

Fig. 2 | Protumour roles of tumour-infiltrating B cells and intratumourally produced antibodies. B cells and plasma cells
may promote tumour growth through several mechanisms. They can release immunosuppressive cytokines — such as IL-10,
IL-35 and transforming growth factor-β (TGFβ) — that promote immunosuppressive phenotypes in myeloid cells, promote
regulatory T cell (Treg cell) development (thereby reciprocally supporting immunosuppressive B cell formation via TGFβ
production) and suppress or misdirect effector T cell responses. The latter processes may be associated with presentation
of tumour-derived antigens by B cells, resulting in antigen-specific boosting of Treg cells and immunosuppression of effector
T cell responses, potentially strengthened by PDL1 expression. B cells can also produce antibodies that are ineffective
in mediating antitumour responses — that is, antibody classes that do not facilitate antigen presentation or mediate
antibody-dependent cell cytotoxicity and phagocytosis of tumour cells, or IgG1 antibody specificities that do not elicit
efficient T cell response or innate cells attack. At the same time, such antibodies can form immune complexes with
tumour or non-tumour antigens, promoting chronic inflammation, tissue remodelling and eventually immunosuppressive
phenotypes in myeloid cells. FcR , Fc receptor ; PNM-MDSC polymorphonuclear myeloid-derived suppressor cell;
TAM, tumour-associated macrophage.

Nature Reviews | Immunology


Reviews

Box 1 | Tumour-associated tertiary lymphoid structures and antibody-producing plasma cells41,74,80,81. These
plasma cells may reside locally and produce high titres
tertiary lymphoid structures (tLs) are typically found at sites of chronic inflammation of tumour-specific antibodies, as has been shown for
and share similarities with lymph nodes. Mature tumour-associated tLs contain breast cancer, melanoma and high-grade serous ovar-
follicle-like structures that have one or more germinal centres with follicular dendritic ian cancer31,80,82. In medullary carcinoma of the breast,
cells and proliferating B cells. Germinal centres are surrounded by more disperse T cells,
tumour-infiltrating B cells were also shown to produce
mature dendritic cells and plasma cells, along with lymphatics and blood vessels18,27,29,176.
some reports indicate that B cells themselves may be involved in the formation of tLs by oligoclonal, somatically mutated anti­bodies that bind
producing CXC-chemokine ligand 13 (CXCL13) and lymphotoxin177–179. tLs have been self antigens such as cell surface-exposed actin with
found in different types of human cancers and usually — but not always — have positive high affinity83. Such antibodies can mediate opsoni-
prognostic value9,29,107,180,181. TLS can be localized in the peritumoural zone and, less zation, complement-mediated lysis of cancer cells and
frequently, within the tumour. The latter location has been reported to have a stronger ADCC executed by natural killer (NK) cells or promote
association with patient survival in pancreatic cancer155. a high density of tumour- antibody-mediated phagocytosis of tumour cells by
infiltrating B cells was associated with increased survival of patients with pancreatic macrophages and granulocytes. In vitro studies have
adenocarcinoma, but only if they were organized within TLS16. in non-small-cell lung demonstrated that B cells from patients with melanoma
carcinoma, stromal infiltration by B cells10,17, a B cell-mediated immune response15 and a can produce IgG that is capable of eliminating cancer
high density of TLS germinal centre B cells, in combination with dendritic cell density,
cells by triggering ADCC21,22.
were found to be positive prognostic markers9,14,29. High density of tLs B cells was also
associated with increased CD4+ T cell clonality28. in anti-PD1 therapy for non-small-cell Reports showing that infiltration of melanoma and
lung carcinoma, TLS and plasma cells were observed within tumour regression sites159, non-small-cell lung carcinoma by plasma cells13,46 and high
and the presence of PD1+CD8+ T cells in TLS was associated with clinical response182. intratumoural immunoglobulin expression in mela-
In high-grade serous ovarian cancer, CD8+ T cell tumour infiltration had no prognostic noma42–44 and head and neck squamous cell carcinoma44
value unless combined with the presence of TLS and a high count of plasma cells, correlate with longer survival indicate the important role
CD4+ T cells and CD20+ B cells. the presence of all these factors simultaneously was of intratumourally localized plasma cells. Clonal expan-
associated with markedly increased survival, with ~65% of patients alive after 10 years18. sion of tumour-infiltrating B cells is also associated with
the presence of tLs — alongside evidence for intense intratumoural proliferation and a more efficient antitumour response42,43,80,83, further
hypermutation of B cells — suggests enhanced local production of effector/memory strengthening the idea that focused humoral response
B cells and plasma cells that have been trained to recognize tumour antigens29,107.
can have important antitumour effects.
Consequently, TLS may potentially provide a microenvironment for maturation of an
efficient antitumour humoral response. At the same time, the role of TLS remains
ambiguous, as in an immunosuppressive environment — for example, in the crosstalk Protumour effects of B cells and antibodies
with regulatory T cells, which may be intensified along with tumour progression — the It is important to note that a number of studies in mouse
TLS may also manufacture immunosuppressive cytokines, as well as B cell and TH cell models have described protumorigenic roles for B cells.
subsets that are immunosuppressive or suboptimal for antitumour response29,107,183. In particular, the growth of syngeneic thymoma, colon
carcinoma and melanoma was significantly slower
in B cell-deficient mice than in wild-type mice or in
of improved prognosis67–70. High serum levels of IgG B cell-deficient mice that had received adoptively trans-
antibodies specific for the Thomsen–Friedenreich antigen ferred B cells. B cell-deficient mice were also reported
(Galβ1–3GalNAcα-O-Ser/Thr) were associated with to show increased T cell infiltration into tumours and
longer survival in patients with breast and gastric can- higher levels of TH1 cell-type cytokine production84.
cer64,68. In general, antibodies against known tumour Another study demonstrated an immunosuppressive role
antigens are detectable in the serum of approximately of B cells and B cell-derived IL-10 in mouse models of
50% of patients with breast cancer74. lymphoma, melanoma and sarcoma85. In three different
There is no universal single origin for tumour-specific mouse models of prostate cancer, B cell depletion made
antibodies that are detected in the serum. These antibod- mice responsive to the chemotherapy agent oxaliplatin86,
ies may be produced by plasma cells residing in classical whereas adoptive transfer of B cells restored tumour
niches, such as the bone marrow or spleen. However, growth87. Additionally, antigen-specific anti­tumour vac-
there is also evidence that plasma cells may occupy cines were shown to be more effective in the absence of
local tissue niches, especially in the context of chronic B cells in mice88,89.
inflammation75,76, and in particular in the tumour micro­ For a number of human cancer types, the presence
environment46,74. If plasma cells in tumour-associated of B cells in tumours has also been shown to be an indi-
TLS produce high levels of tumour-specific antibodies cator of negative outcome (Supplementary Table 1).
in situ, these antibodies can also be detected in serum. High levels of B cell and/or plasma cell infiltration into
On the other hand, there is also evidence of systemic tumours has been associated with increased invasiveness
B cell responses to tumour antigens, as tumour-specific in bladder cancer90, an increased risk of prostate carci-
B cells, plasmablasts and plasma cells can be found in noma relapse after prostatectomy91 and reduced survival
Thomsen–Friedenreich peripheral blood21,77. of patients with renal cell carcinoma19,44. In some stud-
antigen
Intratumoural antibodies were first detected in the ies, high rates of plasma cell and CD138+ B cell infil-
A tumour-associated
carbohydrate antigen highly 1970s78. In 1996, the presence of IgA1 in preparations tration into tumours were also associated with shorter
expressed by approximately from primary breast cancers and their metastases was recurrence-free survival in patients with invasive breast
90% of human carcinomas. reported79. Multiple lines of evidence now leave no doubt carcinoma47. The tumour-promoting effects of B cells
It is believed to facilitate that B cells undergo clonal proliferation, selection for are generally connected with the presence of immuno-
tumour growth by allowing
increased interaction of the
high-affinity antibody and isotype switching within suppressive B cell subsets, often designated as ‘regulatory
tumour cells with carbohy- tumour-associated TLS and in less organized struc- B cells’, that may support tumour progression through
drate-binding lectins. tures, ultimately converting to effector/memory B cells various mechanisms (Box 2; Fig. 2; Table 1).

www.nature.com/nri
Reviews

Immune complexes
Paradoxically, locally or systemically produced From the perspective of antitumour cytotoxic responses,
Antigen–antibody complexes, tumour-specific antibodies may also drive tumorigenic the human IgG1 antibody class is of primary importance,
may include multiple antigen effects. In particular, it has been observed that persistent as these antibodies can bind to Fcγ receptor (FcγR)
and antibody molecules, immune complexes formed by tumour-specific antibod- and trigger ADCC and antibody-mediated phagocy-
as well as complement
ies may be associated with unfavourable clinical out- tosis and mediate complement-based cytotoxicity101.
proteins. They may modulate
activity of myeloid cells, comes65,92,93, potentially due to modulation of activity of Furthermore, FcγR expressed on dendritic cells and
triggering chronic inflammation Fc receptor-bearing myeloid cells. This includes initiation macrophages may capture IgG-bound tumour antigens
and tissue remodelling of chronic inflammation, tissue remodelling and angio- and present peptides derived from these antigens to
processes, and facilitating
genesis programmes94–97, and favouring — directly or T cells23,102–105 (Fig. 1).
formation of myeloid-derived
suppressor cells.
indirectly — of myeloid-derived suppressor cell phenotype98. Maturation of high-affinity antibodies and isotype
It can be hypothesized that antibody response to switching to IgG1 (ref.106) may occur in TLS located
Myeloid-derived antigens that, first, are not exposed and thus cannot be within or in close to the tumour or in tumour-draining
suppressor cell recognized on the surface of tumour cells and, second, lymph nodes, and this results in the generation of
An immunosuppressive
myeloid cell that develops
do not contain immunogenic peptides suitable for pres- plasma cells that may be released into the blood or
under chronic inflammatory entation to T cells on MHC molecules98 may mislead the reside locally and produce tumour-specific cytotoxic
conditions. These cells can immune response and thus favour protumour processes. antibodies29,107. The IgG antibody isotypes, and IgG1
be subdivided into monocytic This could explain the reported correlation of antibodies in particular, are often associated with antitumour B
and polymorphonuclear
specific for certain tumour antigens, such as p53, with a cell activity. The correlation of MUC1-specific and
myeloid-derived suppressor
cells.
negative prognosis56,65. Certain antigenic specificities of Thomsen–Friedenreich-specific antibodies with longer
serum antibodies were also associated with poor prog- survival is mainly associated with IgG isotypes67,68,70.
nosis in ovarian and pancreatic cancer57. Generally, high Tumour-specific IgG production by plasma cells present
levels of immunoglobulin production44 and active isotype in metastases of patients with high-grade serous ovarian
switching99 have been associated with poor prognosis in cancer was also associated with an antitumour response82.
renal cell carcinoma (Supplementary Table 1). Furthermore, effector and memory B cells expressing
Altogether, there is more than sufficient evidence to membrane IgG may also execute direct antitumour func-
show that, in some settings, tumour-infiltrating B cells, tions. In hepatocellular carcinoma, it has been shown
plasma cells and locally produced antibodies may exert that IgG+ memory B cells concentrated in the tumour
prominent immunosuppressive effects, thereby protect- invasive margin can produce granzyme B and TRAIL.
ing the tumour against immune-mediated elimination. These cells also express surface markers characteristic of
An important determinant of whether antibodies have APCs, can produce IFNγ and can cooperate with CD8+
protumour or antitumour effects is the antibody isotype T cells, and are correlated with favourable prognosis30.
present, as we discuss in the following section. Likewise, several reports showed that B cell infiltration is
associated with longer survival in patients with hepato-
Antibody isotypes in tumours cellular carcinoma (Supplementary Table 1). At the same
Human IgG1 antibodies. Antibody functionality is time, accumulation of immunosuppressive B cells may
determined by the constant fragment (Fc) and may be be a negative factor in the same cancer type108,109.
modulated by various post-translational modifications100. Using human RNA-seq datasets from TCGA, we
showed that a high proportion of IgG1 isotype RNA
among all intratumourally expressed immunoglobulin
Box 2 | Immunosuppressive B cells heavy chain (IgH) sequences positively correlates with
non-silent mutation burden in lung adenocarcinoma,
immunosuppressive or regulatory B cells are characterized by the expression of
further supporting the role of IgG1+ B cells in antigen
inhibitory ligands and cytokines such as IL-10, transforming growth factor-β (tGFβ),
PDL1 (potentially including soluble splice variants184) or lymphotoxin, as well as IL-35,
presentation45. Such a high IgG1 proportion was also
IL-6, PD1, CD80, CD86, latency-associated peptide (LAP)–TGFβ, FASL, CD40L and strongly associated with longer survival in melanoma43,
OX40L185–188. immunosuppressive B cells may support tumour progression by inhibiting KRAS-mutant lung adenocarcinoma, and non-papillary
the ability of CD4+ and cytotoxic T cells to eliminate tumours84,189, inhibiting natural killer subtype of bladder cancer45.
cell response85, causing regulatory T cell proliferation and generation from naive or
effector CD4+ T cells190,191, modulating the activity of tumour-associated macrophages IgA, IgD and IgE antibodies. Notably, in the same human
and myeloid-derived suppressor cells, and directly promoting tumorigenesis and melanoma study of TCGA RNA-seq data, high propor-
angiogenesis (reviewed in ref.187). In renal clear cell carcinoma, a malignancy in which tions of IgA, IgD or IgE were associated with a poor
all studies currently indicate a negative role of activated tumour-infiltrating B cells prognosis43. A high intratumoural proportion of the
(Supplementary Table 1), IL-10-producing B cells were shown to inhibit tumour-
IgA isotype was also associated with negative prognosis
infiltrating T cell function192. B cell-produced iL-35 was reported to be upregulated in
serum from patients with pancreatic cancer and to stimulate the proliferation of
in the KRAS-mutant subtype of lung adenocarcinoma45.
pancreatic cancer cell lines, and more specifically KRASG12D-harbouring neoplastic These observations are in line with those from sev-
lesions193. immunosuppressive B cells may be generated within tumour-associated eral other publications. High intratumoural IgA level
tertiary lymphoid structures41 or attracted by the tumour microenvironment — for was found in various human cancers and is associated
example, by the chemokine CXCL13 in prostate and pancreatic cancers87,193. with shorter survival in patients with bladder cancer110.
immunosuppressive B cells have been identified in a variety of different human solid In mouse models, IgA+ plasma cells expressing PDL1 and
tumours, including tongue squamous cell carcinoma194, hepatocellular carcinoma108 IL-10 were shown to impede T cell-dependent responses
and gastric cancer195, and are generally associated with a negative prognosis. Advanced on immunogenic chemotherapy for prostate cancer87.
colorectal carcinomas and metastases also contain increased numbers of cells with a In general, the IgA isotype is often characteristic of the
regulatory B cell phenotype, reflecting immune escape196.
immunosuppressive B cells involved in the regulatory

Nature Reviews | Immunology


Reviews

Table 1 | B cell and plasma cell roles in tumours


Cell types Associated effector Roles in the tumour microenvironment Possible therapeutic approaches Refs
molecules
Immunosuppressive IL-10; TGFβ; Immunosuppression via conversion of Remove tumour-infiltrating B cells, 200,204

B cells and plasma membrane and soluble CD4+ T cells to Treg cells; suppression of TH1 including CD20− ones (e.g. using
cells (often IgA+ PDL1; lymphotoxin; cells, CD8+ T cells and NK cells (via PDL1, anti-CD19 antibodies); remove IgA+
plasma cells, mostly IL-35; IgA with IL-10 and IL-35), including in the course of B cells, including membrane IgA+
CD20−/CD19+) unfocused specificity antigen presentation; induce angiogenesis plasma cells
(via lymphotoxin); promote macrophage
conversion to protumoural M2 phenotype;
stimulation of PMN-MDSC development; no
identified function for produced IgA
IgG1+ plasma cells IgG1 with focused Tumour cell killing via opsonization, complement Keep and support tumour-infiltrating 29,160,

specificity (large fixation, ADCC, antibody-mediated phagocytosis; B cells; promote TLS formation; stimulate 161

hypermutating promotion of antigen presentation by dendritic NK cells (e.g. by anti-NKG2A , anti-PD1


clones); cytokines cells; drive cytotoxic T cell responses and anti-PDL1); clone IgG1 for antibody
or CAR T cell therapy ; tumour-associated
antigen/neoantigen vaccination
Effector B cells with Granzyme B, TRAIL; Direct tumour cell killing; antigen-specific Tumour-associated antigen/neoantigen 29,160,

antitumour effects MHC class I, MHC antigen presentation; polarization and education vaccination; clone tumour cell-specific 161

(often IgG+) class II; IFNγ, IL-12 of CD4+ T cells; TLS formation; dendritic cell IgG1 for CAR T cell therapy
migration; macrophage conversion to tumoricidal
M1 phenotype
ADCC, antigen-specific cell cytotoxicity ; CAR , chimeric antigen receptor ; NK , natural killer ; PMN-MDSC, polymorphonuclear myeloid-derived suppressor cell;
TGFβ, transforming growth factor-β; TH1 cells, T helper 1 cells; TLS, tertiary lymphoid structures; Treg cells, regulatory T cells.

loop in which B cells promote expansion of regula- lung squamous cell carcinoma122, as well as in STK11-
tory T cell (Treg cell) populations, while Treg cells produce mutant and proximal proliferative subtypes of lung ade-
transforming growth factor-β (TGFβ), which mediates nocarcinoma45. The explanation for this positive effect of
isotype class switching to IgA29,111,112 (Fig. 2). This loop IgG4 in particular tumour microenvironments remains
maintains gut homeostasis113–115 and may be naturally unclear, but we hypothesize that it could be associated
engaged by the immune system to regulate inflammation with the inability of IgG4 to form immune complexes123,
of various causes. For example, it has been shown that IgA+ which could otherwise stimulate chronic inflammation
B cells producing IL-10 and expressing PDL1 may accu- and tissue remodelling processes and eventually induce
mulate in the liver during chronic inflammation and sup- immunosuppressive myeloid-derived suppressor cell
press cytotoxic CD8+ T lymphocytes capable of preventing phenotype94,97,98 (Fig. 2).
hepatocellular carcinoma109. Correspondingly, high intra-
tumoural IgA expression levels may be considered as a IgG3 antibodies. RNA-seq data have suggested that
biomarker of B cell involvement in the IL-10, TGFβ and the relative proportions of IgG3 can be either neutral
Treg cell pathways, in at least some cancer types or subtypes. or negatively associated with survival in human mela-
A high proportion of IgE may reflect high intra­ noma43, although the reasons for this are unclear. This
tumoural IL-4 levels and TH cell polarization towards is in contrast to IgG1, which has a positive association
the TH2 cell fate, which is commonly associated with inef- with survival for this malignancy, even though both
ficient tumour control due to suppression of TH1 cell and of these antibody isotypes are capable of triggering
cytotoxic T cell response116. IgD antibodies have been ADCC, phagocytosis, complement activation and anti-
shown to bind to basophils and can drive the production gen cross-presentation. A possible explanation could
of factors such as IL-4, IL-5, IL-13, BAFF and APRIL117,118; be that although IgG3 antibodies can interact with
as a consequence, high IgD levels may be associated activating Fc receptors, there is evidence that the IgG3
with IgE and IgA class switching and the generation response is early and transient and that IgG3 antibodies
of TH2 cells. These effects could potentially explain the generally have a lower number of accumulated hyper-
correlation of a high proportion of intratumoural IgE mutations and lower affinity106. Faster turnover of IgG3
and IgD with negative prognosis in melanoma43. antibodies124 can also result in relatively lower local con-
centrations of IgG3 than IgG1, even if both isotypes are
IgG4 antibodies. It was shown that IgG4 antibodies, produced at comparable levels.
which are also associated with type 2 immunity119, An alternative explanation comes from the high affin-
lack antitumoural effector functions and may block ity of monomeric IgG3 for FcγRI (which is expressed
tumour-specific IgG1 response120. A high proportion of on macrophages) and FcγRIIIA (which is expressed on
IgG4 of all IgG in serum is associated with a poor prog- both macrophages and NK cells), and especially for the
nosis in patients with melanoma, and tumour-specific variant FcγRIIIA-158V (ref.125). Therefore, non-tumour-­
IgG4 antibodies have been shown to be produced in situ specific, antigen-free IgG3 antibodies may efficiently
by plasma cells located within the melanoma lesions121. occupy vacant FcγRs of NK cells and macro­phages, thus
A high proportion of IgG4 has also been associated blocking their interaction with cytotoxic antibodies
with favourable prognosis in some cancer types, such as bound to the surface of tumour cells (Fig. 3).

www.nature.com/nri
Reviews

By contrast, non-tumour-specific, antigen-free IgG1 identifying prognostic and predictive cancer biomarkers,
antibodies do not occupy FcγRs, leaving them vacant such as clonality metrics43,77,128. Furthermore, such analysis
and thus allowing IgG1 or IgG3 bound to the surface allows one to identify therapeutic T cell receptor and anti-
of tumour cells to trigger their recognition and elimi- body variants specific for tumour-associated antigens or
nation via ADCC and antibody-mediated phagocytosis. neoantigens21,22,31,77,80,83,129. A number of molecular and bio-
On the surface of tumour cells, avidity increases due to informatic tools have been developed to this end, and the
engagement of multiple receptors (Fig. 3). Furthermore, appropriate choice depends on the available material,
allosteric increase of antigen-bound IgG1 affinity for time, cost-efficiency and required quality and depth of
FcγRs126, still disputable, could enhance productive immunoglobulin repertoire profiling42,43,130–140 (Table 2).
ADCC or phagocytic attack127. Of note, immunoglobulin repertoires produced at the
In conclusion, the relative abundance of distinct tumour site are often very focused. Up to ~70% of immu-
antibody isotypes in the tumour and tumour-associated noglobulin mRNA molecules may belong to a single
lymphoid structures is an important parameter of the clonal group, as has been shown for breast cancer31,40,80,
tumour microenvironment. At the same time, the corre- medullary carcinoma of the breast83, ovarian cancer18 and
lation of distinct antibody isotypes with poor or favour- melanoma41–43,129, which reflects the presence of clonal
able prognoses or response to therapy does not allow one plasma cells producing particular antibody variants.
to discriminate the passive role of isotype as a biomarker At the same time, effector B cells may also have a number
of ongoing processes, in which antibody specificity is of of antigen-specific roles (Table 1), which makes analysis of
low importance, and an active role in which antigen spec- their repertoire an important part of the whole picture.
ificity determines the efficiency of cancer surveillance or The technical complication is that such analysis requires
promotion. A way to look deeper into this picture is to cell sorting to plasma and memory/effector B cells.
analyse intratumoural immunoglobulin repertoires and Otherwise, plasma cell immuno­g lobulin sequences
clonality, as we discuss next. will dominate the repertoire at the RNA level, owing to
their markedly higher expression levels. Alternatively,
Analysing immunoglobulin repertoires a combination of DNA-based and RNA-based immuno-
The RNA-based or DNA-based analysis of intratu- globulin repertoire profiling would be the informative
moural T cell receptor and immunoglobulin repertoires way to estimate both B cell clonal and immunoglobulin
has received increasing attention as a powerful tool for functional composition within a tumour sample.

High proportion of IgG3 antibodies High proportion of IgG1 antibodies

FcγR

NK cell
‘M1-like’
macrophage
‘M1-like’ NK cell
ADCC macrophage
ADCC
Phagocytosis

Phagocytosis

Tumour
antigen
Tumour cell

IgG1 IgG3 Tumour-specific Tumour-


IgG1 and IgG3 attacking cell

Fig. 3 | Potential advantages of Igg1-biased over Igg3-biased humoral response. Left panel: due to the high affinity of
monomeric IgG3 for Fcγ receptors (FcγRs) expressed on macrophages and natural killer (NK) cells, high levels of production
of IgG3 (shown in blue) block antigen-specific cell cytotoxicity (ADCC) and antibody-mediated phagocytosis, even
though tumour-specific IgG1 (shown in red) and IgG3 antibodies may also be present in the tumour microenvironment.
Right panel: by contrast, in conditions dominated by the IgG1 isotype, antigen-free IgG1 antibodies do not occupy
FcγRs. The avidity of macrophages and NK cells towards IgG1 bound on the surface of tumour cells increases due to the
engagement of multiple receptors.

Nature Reviews | Immunology


Reviews

Table 2 | Pros and cons of methods for extracting immunoglobulin repertoires from cancer samples
Feature Advantages Disadvantages
Starting tissue
Bulk tumour No sorting required; no decrease in sensitivity due to cell loss during tissue Low reproducibility if tumour infiltration is low;
preparation and sorting unpaired repertoirea
Sorted B cells Good reproducibility ; ability to combine B cell RNA-seq and repertoire Requires cell sorting; sorting may decrease
profiling sensitivity due to cell loss; unpaired repertoirea
Single B cells Allows pairing of full-length heavy and light chains; semiquantitative in Low throughput, superficial profiling; expensive;
terms of cells; can be combined with single-cell RNA-seq; full-length chain low reproducibility
extraction available for single-cell RNA-seq with MiXCR
Starting template
DNA Provides information on clonal composition at the level of cells Information on expression levels lost; isotype
information lost
RNA Provides information on immunoglobulin expression levels; isotype No information on clonal composition at the level
information preserved of cells
Profiling method
RNA-seq No additional efforts and costsb; ability to work with TCGA and other tumour Prominent tumour infiltration is a prerequisite,
or B cell RNA-seq data; full-length chain extraction available for the large although even minor presence of plasma cells may
clonotypes with MiXCR be sufficient to capture highly represented CDR3
RNA reads; superficial profiling
Multiplex PCR High sensitivity, works with medium-quality RNA Quantitative biases
5′ RACE Unbiased amplification; high reproducibility Lower sensitivity than multiplex PCR; requires
high-quality RNA
UMI
With UMI Improved quality in long paired-end sequencing; high efficiency in full-length Additional step in data analysis (e.g. with MiGEC137
V(D)J profiling; elimination of most PCR and sequencing errors and biases; or MiNNN)
elimination of solid-phase artefacts and contaminants; quantification of
starting molecules offers a control for technical bottlenecks; accurate
normalization of samples in terms of unique molecules analysed
Without UMI No additional step in data analysis Lots of errors; low efficiency in full length V(D)J
profiling; uncontrolled depth of profiling
CDR3, complementarity determining region 3; RACE, rapid amplification of cDNA ends; RNA-seq, RNA sequencing; TCGA , The Cancer Genome Atlas; UMI, unique
molecular identifier. aMost highly represented clones can be paired by frequency (ref.145). bIf RNA-seq data are used for general analysis of transcriptome profiles
for tumour or sorted (for example, CD19+) tumour-infiltrating B cells.

Most current work on immunoglobulin repertoire of intratumoural immunoglobulin repertoires33,44,142. In


profiling is based on RNA derived either from the particular, assessment of the degree of clonality — that
bulk tumour or from sorted B cell subsets. Targeted is, the presence of large clonal expansions — in the intra-
IgH and Igλ/Igκ cDNA libraries can be obtained by tumoural immunoglobulin repertoire, as well as the
multiplex amplification or 5′ rapid amplification of amount of somatic hypermutations in immunoglobulin
cDNA ends, with or without use of unique molecular gene segments, can offer prognostic value for some
identifiers (UMIs; also known as UIDs)130–139. These types of cancer. Analysis of IgH repertoires extracted
can be designed to obtain full-length (from frame- from TCGA RNA-seq data from melanoma samples has
work region 1 to framework region 4) sequences or revealed that survival is highest for patients with high
only complementarity-determining region 3 (CDR3) immunoglobulin expression and clonality42,43, where
sequences, or to preserve or discard information about clonality reflects mainly the presence of plasma cell
antibody isotype. These methods differ in terms of sen- clonal expansion, as discussed above. Clonality had a
sitivity, accuracy and informativity of obtained reper- much stronger association with survival: the combination
toires and should be properly selected for each particular of high immunoglobulin expression (that is, numerous
task (Table 2). plasma cells at the site) and low clonality, which could
One recent study of intratumoural immunoglobulin be interpreted as absence of a distinct antigen-specific
repertoires with UMI-based CDR3 profiling demon- response, resulted in an equally poor prognosis as low
strated that there is high clonal overlap, but with nota- immunoglobulin expression, while low immunoglobu-
ble differences in the frequency of IgH CDR3 variants, lin expression and high clonality resulted in an average
between the tumour site and adjacent normal mucosal prognosis. Although high immunoglobulin expression
tissue in colorectal advanced adenoma and colorectal can- and a high proportion of the IgG1 isotype has been asso-
cer. In comparison, much greater CDR3 repertoire homo­ ciated with a better prognosis, the combination of these
geneity was observed across different segments of healthy two parameters with high IgH clonality was not associ-
colon141. These results may reflect clonal proliferation and ated with further improvements in survival prognosis for
plasma cell generation at the tumour site. TCGA patients with melanoma43. This can be explained
On the basis of non-targeted tumour or sorted B cell by the fact that a high IgG1 proportion correlates with
RNA-seq data, one can also identify prognostic signatures the presence of large clonal expansions, such that the two

www.nature.com/nri
Reviews

parameters are essentially dependent. In contrast, a high important role in cancer surveillance, the RNA-seq-
IgA proportion correlates with low clonality, suggesting based approach could be directly used in cancer immu-
that, in melanoma, B cell switching to the IgA isotype is a notherapy using autologous or allogeneic cytotoxic
passive consequence of the intratumoural cytokine envi- antibodies or chimeric antigen receptor (CAR) T cells.
ronment and is not driven by particular antigens. Large Implementation of microfluidic emulsion-based
clusters of hypermutating immunoglobulin subvariants approaches can provide a comprehensive analysis of
are also predominantly formed by IgG1 antibodies43. paired heavy and light immunoglobulin chains expressed
IgG1 repertoires are characterized by high clonality and by hundreds and thousands of individual B cells146.
intense hypermutation in samples either with or without The combination of molecular barcoding and droplet
lymph node material43, reflecting the presence of TLS and barcoding is crucial for providing increased accuracy
correlating with increased AID expression, as has been of analysis. This approach was recently applied to the
reported in melanoma129. analysis of tumour-infiltrating lymphocytes in ovarian
Intratumoural expression levels, proportions, clon- cancer147, revealing ~6,000 immunoglobulin pairs per
ality and extent of hypermutation for the IgG1, IgA, 400,000 unsorted tumour cells that entered the emulsion
IgE and IgD isotypes in melanoma are thus compo- reaction. In general, the rapid development of single-cell
nents of the same picture. A focused immune response analysis approaches, especially those based on emulsion
driven by the appropriate environment and available techniques148, is revealing new avenues for both immuno­
immunogenic tumour antigens leads to the genera- globulin repertoire and transcriptomic profiling of
tion of large plasma cell clones producing high-affinity, tumour-infiltrating B cells, allowing pairing of heavy and
tumour-specific IgG1 antibodies, while an immunosup- light chains of immunoglobulins146,149 as well as linkage
pressive environment leads to B cells switching to IgA of the repertoire with antigenic specificity150 and the
and other isotypes that usually exhibit less focused spec- single-cell transcriptome.
ificity but with higher potential for cytokine-mediated A remarkable example is a recent work in which
immunosuppression. New York oesophageal squamous cell carcinoma 1
Of note, this scenario may be typical of human mela- (NY-ESO-1)-specific antibodies were successfully
noma, but could be different for other cancers. Although identified in breast cancer by single-cell analysis of
a high IgG1 proportion is specifically beneficial for B  cells taken from tumour-draining lymph nodes.
KRAS-mutant lung adenocarcinoma, high IgG1 clonality Several reconstituted antibodies were cloned and shown
remains a neutral parameter45. This observation may indi- to be affinity maturated against NY-ESO-1 and were also
cate that IgG1+ B cells play a positive role in this cancer found in the circulation by affinity chromatography
subtype via antigen presentation or direct attack of tumour enrichment and mass spectrometry151.
cells, rather than through ADCC or antibody-mediated The microfluidic methods are still relatively laborious
phagocytosis, since the latter would require a highly and expensive and thus are currently limited to relatively
clonal IgG1 repertoire produced by plasma cells at the small numbers of samples and cells. Nevertheless, these
RNA level, as seen with melanoma. As KRAS-specific powerful approaches could help reveal the functional
B cells143 and CD4+ T cells144 have been recently identi- heterogeneity of tumour-infiltrating B cells and sug-
fied, an attractive interpretation that still requires fur- gest rational therapeutic approaches that canalize their
ther investigation is that in KRAS-mutant tumours, the functionality towards desirable antitumour action. In the
IgG1 shift of tumour-infiltrating B cells could lead to future, direct single cell-based clinical applications
more efficient presentation of the mutant KRAS peptide may become possible that allow rapid identification
itself — along with other tumour-associated antigens and of tumour-specific IgG1 antibodies for use in cancer
neoantigens — thus attacking the tumour in the most immunotherapy.
vulnerable spot.
Therapeutic targeting of B cells in tumours
Recovering functional antitumour antibodies As discussed already, a number of studies indicate that
Both tumour-infiltrating and circulating B cells and B cells and plasma cells may often play a protumour role
plasma cells have been shown to be a potent source of in the tumour microenvironment, and this has prompted
antibodies that recognize autologous tumours as well as the development of therapeutic approaches based on
some heterologous tumours — predominantly those of B cell depletion in solid cancers93,152,153 (Box 3). However,
the same tissue origin21,22,31,77,80,82,83. To recover functional as we have also discussed in this Review, B cells may exert
antitumour antibodies via high-throughput sequencing, potent antitumour effects, with notable interspecies dif-
it is crucial to obtain full-length and error-free immuno­ ferences that can make such effects challenging to model
globulin sequences and to accurately pair light and in mice. In relatively short-term tumour transplantation
heavy chains from each antibody of interest. For the models, TLS may not form or may have defective struc-
largest intratumoural immunoglobulin-producing tures154, which may in turn account for the prevalence of
plasma cell clones, information on full-length IgH protumoural functions of B cells reported in mice mod-
and Igκ or Igλ light chains can be extracted from tar- els. By contrast, in human cancers, immune-activating,
geted immunoglobulin-sequencing data139 or RNA-seq tumour-suppressing B cells are often organized in intra-
data42,43. The latter option was recently upgraded in tumoural TLS, the appearance of which is associated
MiXCR. The resulting heavy and light chains can be with favourable prognoses for many cancers9,14,29,155.
paired on the basis of frequency matching145. Because Thus, B cell depletion may have deleterious conse-
some of the largest IgG1 clonotypes could play an quences for a large subset — and potentially a majority

Nature Reviews | Immunology


Reviews

Box 3 | B cell depletion in solid cancers clonality, dominance of the IgG1 isotype, hypermutation
intensity and the spectrum of cytokines and costimu-
For those types and subtypes of solid cancers (for example, clear cell renal cell latory molecules they produce, as well as abundance
carcinoma; supplementary table 1) or narrowly specified cohorts of patients where of tumour-specific antibodies associated with positive
B cell depletion could have clinical benefit, the method used to eliminate or suppress
clinical outcome57,64,67–70.
B cells should be chosen carefully. Early animal studies provided motivation for the use
Considering ADCC as one of the direct mechanisms
of B cell-depleting anti-CD20 antibodies such as rituximab or ofatumumab as an
adjuvant for treatment of solid tumours. However, clinical data have shown that of antigen-specific antitumour B cell response, immuno­
neoadjuvant therapy with rituximab had no effect following iL-2 therapy for therapeutic agents that stimulate NK cells —such as
metastatic renal cell carcinoma and melanoma197. anti-CD20 depleting antibodies monalizumab, a monoclonal antibody to NKG2A,
were also used in an adjuvant setting to treat 10 patients with metastatic melanoma198. which serves as a checkpoint inhibitor for NK cells156
this treatment significantly decreased overall inflammation and tumour infiltration — should contribute to a more pronounced response
by CD8+ T cells and macrophages, and did not suppress tumour progression. Several for patients with a highly focused IgG1 repertoire of
other clinical trials involving patients with melanoma treated with rituximab tumour-associated plasma cells. Activation of (probably
(Clinicaltrials.gov identifiers NCt01032122 and NCt02142335) were either clonal) NK cell populations with anti-PD1 therapy157,158
terminated or completed without results being reported. One more trial of rituximab suggests that the combination of B cell-promoting and
for treatment of solid tumours is now in progress, with the aim of depleting
PD1-blocking therapies could be beneficial owing to
tumour-infiltrating B cells in prostate cancer (NCt01804712). intratumoural
immunosuppressive B cells are typically classified either as regulatory B cells (typically, synergistic effects on B and T cells as well as B and NK
as producing iL-10 and tGFβ) or iL-10-producing plasma B cells (often iga+, also cells. Notably, immune-mediated tumour clearance in
expressing PDL1 and tGFβ). Regardless, these cells are usually CD20– (refs87,199), neoadjuvant anti-PD1 therapy for non-small-cell lung
and thus the clinical failure of anti-CD20-based efforts to deplete intratumoural carcinoma was associated with local formation of TLS
B cells197,200 is unsurprising. Bispecific T cell-engaging anti-CD19/anti-CD3 antibodies and the presence of plasma cells159. More recent stud-
such as blinatomumab or CD19-targeting chimeric antigen receptor T cells, both of ies have also demonstrated prominent association of
which are now widely used in B cell malignancies, could be a superior method for responsiveness to immune checkpoint blockade with
eliminating all B cells, including immunosuppressive B cells and plasma cells. the presence of TLS and clonal B cell expansion in
Long-living plasma cells may lose expression of CD19 as well201. However, melanoma160,161, and of TLS and B cells in soft-tissue
immunosuppressive CD19− tumour-associated plasma cells have not yet been
sarcomas162. Anti-CTLA4 therapy was shown to nota-
reported. Several alternative targets to CD19 remain as well, including CD38
(refs202,203), CD138 and surface IgA, as human IgA+ plasma cells (unlike igG+ plasma
bly increase the number of circulating plasma cells
cells) retain a functional membrane B cell receptor204. small synthetic drugs such as in non-progressing patients with metastatic cancer77.
ibrutinib are another option for suppressing B cells. Ibrutinib targets BTK, a critical In the context of the largely CD4+ T cell-supporting out-
mediator of signalling by B cell receptor and other receptors (FcγRs, Toll-like come of anti-CTLA4 therapy3, this effect suggests that
receptors, G protein-coupled receptors) as well as B cell survival, migration and CD4+ T cell-mediated stimulation of antibody affinity
differentiation into plasma cells. ibrutinib has been used in several clinical trials as a maturation and plasma cell generation could induce
monotherapy or in combination with other therapies to treat various solid tumours205. tumour-associated B cells that produce tumour-specific
ibrutinib significantly increases survival in combination with chemotherapy or antibodies in local and draining lymph nodes and TLS.
immunotherapy. However, the role of B cells in this effect is not clear, as ibrutinib has Vaccination trials have also demonstrated the induction
also been shown to affect intratumoural myeloid cells and enhance t helper 1 cell
of TLS formation29,163,164.
skewing of CD4+ T cells206,207. inhibitors of the BaFF/aPriL system208 also represent
unexplored options for immunotherapeutic intervention in solid cancers where Analysis of intratumoural compositions of immuno­
suppression of B cell activity could be desirable. there is also interest in rational globulin repertoires could help in patient stratification
combination of B cell-depleting therapy with other immunotherapeutic approaches. for different types of B cell-targeting therapy, either
The existence of a natural loop between IgA-producing B cells and regulatory T cells individually or at the level of particular cohorts. For
(Fig. 2) suggests that simultaneous targeting of the two subsets could be beneficial — melanoma, the intratumoural presence of large hyper-
for example, by combining cytotoxic anti-GITR antibodies targeting regulatory T cells mutating clonal IgG1 expansion — reflecting affin-
with anti-CD19 therapy. On the other hand, a study on a mouse model suggested ity maturation and antigen-driven proliferation of
that anti-GITR therapy activates B cells, which was a crucial part of response to this immunoglobulin repertoires produced in situ within
immunotherapy35. this observation confirms once again that total B cell depletion
tumour-associated TLS and draining lymph nodes —
is a 1D intervention and should be applied to only very specific cohorts of patients for
is associated with longer patient survival14,18,31,40–43. This
whom the role of B lymphocytes is shown to be clearly negative.
most probably results from efficient tumour control by
ADCC or antibody-mediated phagocytosis, suggesting
— of patients, and strategies for rational patient stratifi- that therapeutic approaches that support these mecha-
cation for B cell-related therapies are therefore required. nisms, promote TLS function and shift B cells towards
Such stratification could be based on the abundance of the IgG1 isotype should be generally beneficial for
IL-10-producing B cells in tumours or tumour-draining patients with melanoma.
lymph nodes, as estimated by immunohistochemistry or However, the whole picture for different cancers
flow cytometry. Measuring the serum or tumour abun- seems to be more complicated. The established mode
dance of tumour-specific antibodies or B cells that are of tumour-immunity interactions may differ for dis-
associated with a negative clinical outcome56,57,65,66 could tinct functional subtypes of a certain cancer, as cur-
also potentially inform B cell-depleting therapies. rently classified on the basis of tumour mutational
Alternative strategies should prevail for the cate- load, extent of infiltration by immune cells, abundance
gories of patients in which tumour-associated B cells, of cancer-associated fibroblasts, angiogenesis, tumour
plasma cells and antibody specificities reinforce the proliferation rate, cancer signalling pathways and other
immune system against cancer. For some cancer types, gene signatures and histological features165–168. Notably,
this can be determined on the basis of high antibody the established type of tumour–immune interaction may

www.nature.com/nri
Reviews

also be influenced by driver mutations associated with or CAR T cells of deficient specificities. Thus, identifica-
particular modes of immune response169–172. tion and demonstration of clinical utility of novel thera-
Furthermore, there may be other specific tumour peutic tumour-specific antibodies capable of recognizing
features, such as particular mechanisms of immune tumour antigens in a relatively broad spectrum of patients
suppression and escape, as well as the presence and — with the aim of depleting tumour cells and/or boost-
abundance of immunogenic tumour antigens that elicit ing efficient tumour-specific T cell response — remains a
efficient antitumour responses or, conversely, that mis- highly attractive and potentially feasible goal173–175.
lead the immune system, triggering prominent but sub-
optimal B cell and T cell responses. Dominant isotypes Conclusion
of antibodies specific to particular tumour antigens and In this Review, we have emphasized that B cells are not
the presence of tumour antigen-specific clonal popu­ simply bystander cells in the tumour microenvironment
lations of T cells and B cells of a particular functionality — but are instead active participants that can fundamen-
including Treg cells and regulatory B cells — further add tally orchestrate the immune response. B cells can pro-
to the complexity of the whole picture. This latter con- tect against tumours under certain conditions, mainly
sideration makes the ability to identify tumour-specific through the production of tumour-specific antibodies
immunosuppressive T and B cells highly desirable. and presentation of tumour antigens, but certain B cell
Correspondingly, we should expect that the roles of subsets and antibody specificities can also suppress
tumour-infiltrating B cells, and thus the prognostic and anticancer immunity and promote tumour growth. The
predictive significance of immunoglobulin repertoires, existence of such opposing effects can be explained by
will differ for different cancer subtypes. Indeed, the classifi- the influence of different intratumoural contexts, which
cation of tumours into molecular subtypes often correlates polarize B cells to distinct functional phenotypes, as well
with the prognostic value of B cell and immunoglobulin as the distinct consequences of immune reactions to dif-
repertoire features (Supplementary Table 1). ferent types of immunogenic tumour antigen. Different
A remarkable example is the distinct prognostic functional populations of B cells, antibody isotypes and
role of immunoglobulin repertoires in subtypes of lung antigenic specificities could enhance or diminish ADCC
cancer. As already mentioned, a high ratio of intra- and antibody-dependent phagocytosis, provide desira-
tumoural IgG1 to IgA is specifically associated with ble or undesirable modes of antigen presentation, or
longer survival in KRAS-mutant adenocarcinoma but contribute to the generation of immune complexes with
not in wild-type KRAS adenocarcinoma, while abun- potentially immunosuppressive impacts.
dance of IgG4 is associated with a favourable prognosis Systematic characterization of antibody expression,
in squamous cell carcinoma122 and in STK11-mutant spatial localization and functional phenotypes of intra-
and proximal proliferative adenocarcinoma45. Positive tumoural and nearby B cells and their interactions with
prognosis was also associated with high IgH expression other cells may provide new insights into the role of
level and high IgH to MS4A1 (which encodes CD20) B cells in tumour immunology and expand our ability
expression ratio specifically for proximal proliferative to rationally manage these roles in distinct, properly
lung adenocarcinoma45. classified cancer subtypes.
Immune infiltration of breast and ovarian cancers The ambiguous role of B cells in the tumour environ-
is highly dependent on molecular subtype. Activated ment predetermines the multidirectional development
B cells and IgG gene signatures are enriched and have of immunotherapeutic approaches, either supporting
significant prognostic value only in those subtypes that positive B cell types and specificities or providing defi-
have high overall immune infiltration (namely basal and cient specificities or else suppressing negative B cell types
HER2 enriched for breast cancer, and immunoreactive and their associated processes and antibody specificities.
and mesenchymal for ovarian cancer), reflecting tight As a consequence, decisions related to the application
cooperation of B cells with other immune cells33. of B cell-targeted therapies and their combination with
Altogether, it seems that an appropriate classification other therapies should be based on a deep understand-
strategy for tumour subtypes needs to be developed — ing of the prevailing nature of B cell participation in
considering immunoglobulin repertoires as one of the tumour–immune interactions in each cancer subtype
critical parameters — wherein distinct B cell phenotype or even in each patient. Our ability to analyse, rationally
and antibody isotype content, clonality and specificity interpret and use immunoglobulin repertoire data from
will ultimately dictate the rationale for different com- tumour-associated B cells is becoming an important part
binations of immunotherapeutic interventions. Where of this story.
possible and appropriate, one component of such com-
Published online xx xx xxxx
bination therapies could include antitumour antibodies

1. Schumacher, T. N. & Schreiber, R. D. Neoantigens and immunotherapy. Nat. Rev. Immunol. 18, 635–647 7. Bindea, G. et al. Spatiotemporal dynamics of
in cancer immunotherapy. Science 348, 69–74 (2018). intratumoral immune cells reveal the immune
(2015). 5. Schoorl, R., Riviere, A. B., Borne, A. E. & landscape in human cancer. Immunity 39, 782–795
2. Wherry, E. J. & Kurachi, M. Molecular and cellular Feltkamp-Vroom, T. M. Identification of T and (2013).
insights into T cell exhaustion. Nat. Rev. Immunol. 15, B lymphocytes in human breast cancer with 8. Chevrier, S. et al. An immune atlas of clear cell
486–499 (2015). immunohistochemical techniques. Am. J. Pathol. 84, renal cell carcinoma. Cell 169, 736–749 e718
3. Wei, S. C. et al. Distinct cellular mechanisms underlie 529–544 (1976). (2017).
anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 6. Jackson, P. A. et al. Lymphocyte subset infiltration 9. Dieu-Nosjean, M. C. et al. Long-term survival for
170, 1120–1133 (2017). patterns and HLA antigen status in colorectal patients with non-small-cell lung cancer with
4. Borst, J., Ahrends, T., Babala, N., Melief, C. J. M. & carcinomas and adenomas. Gut 38, 85–89 intratumoral lymphoid structures. J. Clin. Oncol. 26,
Kastenmuller, W. CD4+ T cell help in cancer immunology (1996). 4410–4417 (2008).

Nature Reviews | Immunology


Reviews

10. Al-Shibli, K. I. et al. Prognostic effect of epithelial and ductal carcinoma of the breast. J. Immunol. 169, 53. Sahin, U. et al. Human neoplasms elicit multiple
stromal lymphocyte infiltration in non-small cell lung 1829–1836 (2002). specific immune responses in the autologous host.
cancer. Clin. Cancer Res. 14, 5220–5227 (2008). 32. Nielsen, J. S. et al. CD20+ tumor-infiltrating Proc. Natl Acad. Sci. USA 92, 11810–11813 (1995).
11. Lund, F. E. & Randall, T. D. Effector and regulatory lymphocytes have an atypical CD27- memory 54. Stockert, E. et al. A survey of the humoral immune
B cells: modulators of CD4+ T cell immunity. Nat. Rev. phenotype and together with CD8+ T cells promote response of cancer patients to a panel of human
Immunol. 10, 236–247 (2010). favorable prognosis in ovarian cancer. Clin. Cancer tumor antigens. J. Exp. Med. 187, 1349–1354
12. Ladanyi, A. et al. Prognostic impact of B-cell density in Res. 18, 3281–3292 (2012). (1998).
cutaneous melanoma. Cancer Immunol. Immunother. This study described antigen-experienced 55. Brichory, F. M. et al. An immune response manifested
60, 1729–1738 (2011). tumour-infiltrating B cells that express molecules by the common occurrence of annexins I and II
13. Erdag, G. et al. Immunotype and immunohistologic associated with antigen presentation and colocalize autoantibodies and high circulating levels of IL-6
characteristics of tumor-infiltrating immune cells with activated CD8+ T cells. in lung cancer. Proc. Natl Acad. Sci. USA 98,
are associated with clinical outcome in metastatic 33. Iglesia, M. D. et al. Prognostic B-cell signatures using 9824–9829 (2001).
melanoma. Cancer Res. 72, 1070–1080 (2012). mRNA-seq in patients with subtype-specific breast 56. Reuschenbach, M., von Knebel Doeberitz, M. &
14. Germain, C. et al. Presence of B cells in tertiary and ovarian cancer. Clin. Cancer Res. 20, 3818–3829 Wentzensen, N. A systematic review of humoral
lymphoid structures is associated with a protective (2014). immune responses against tumor antigens.
immunity in patients with lung cancer. Am. J. Respir. 34. Milne, K. et al. Systematic analysis of immune Cancer Immunol. Immunother. 58, 1535–1544
Crit. Care Med. 189, 832–844 (2014). infiltrates in high-grade serous ovarian cancer reveals (2009).
15. Hernandez-Prieto, S. et al. A 50-gene signature is a CD20, FoxP3 and TIA-1 as positive prognostic factors. This work summarizes data on elevated levels
novel scoring system for tumor-infiltrating immune PLoS One 4, e6412 (2009). of tumour-associated antigen-specific antibodies
cells with strong correlation with clinical outcome of 35. Zhou, P. et al. Mature B cells are critical to T-cell- in the serum of patients with cancer and their
stage I/II non-small cell lung cancer. Clin. Transl. Oncol. mediated tumor immunity induced by an agonist association with prognosis.
17, 330–338 (2015). anti-GITR monoclonal antibody. J. Immunother. 33, 57. Gnjatic, S. et al. Seromic profiling of ovarian and
16. Castino, G. F. et al. Spatial distribution of 789–797 (2010). pancreatic cancer. Proc. Natl Acad. Sci. USA 107,
B cells predicts prognosis in human pancreatic 36. Forte, G. et al. Inhibition of CD73 improves 5088–5093 (2010).
adenocarcinoma. Oncoimmunology 5, e1085147 B cell-mediated anti-tumor immunity in a mouse model 58. Amornsiripanitch, N. et al. Complement factor H
(2016). of melanoma. J. Immunol. 189, 2226–2233 (2012). autoantibodies are associated with early stage
17. Kinoshita, T. et al. Prognostic value of tumor-infiltrating 37. Li, Q. et al. Adoptive transfer of tumor reactive B cells NSCLC. Clin. Cancer Res. 16, 3226–3231 (2010).
lymphocytes differs depending on histological type and confers host T-cell immunity and tumor regression. 59. Chapman, C. J. et al. EarlyCDT®-Lung test: improved
smoking habit in completely resected non-small-cell Clin. Cancer Res. 17, 4987–4995 (2011). clinical utility through additional autoantibody assays.
lung cancer. Ann. Oncol. 27, 2117–2123 (2016). 38. Rubtsov, A. V. et al. CD11c-expressing B cells are Tumour Biol. 33, 1319–1326 (2012).
18. Kroeger, D. R., Milne, K. & Nelson, B. H. Tumor- located at the T cell/B cell border in spleen and 60. Macdonald, I. K., Parsy-Kowalska, C. B. &
infiltrating plasma cells are associated with tertiary are potent APCs. J. Immunol. 195, 71–79 (2015). Chapman, C. J. Autoantibodies: opportunities for early
lymphoid structures, cytolytic T-cell responses, and 39. Deola, S. et al. Helper B cells promote cytotoxic T cell cancer detection. Trends Cancer 3, 198–213 (2017).
superior prognosis in ovarian cancer. Clin. Cancer Res. survival and proliferation independently of antigen 61. Dai, L. et al. Autoantibodies against tumor-associated
22, 3005–3015 (2016). presentation through CD27/CD70 interactions. antigens in the early detection of lung cancer.
This study links together the presence of TLS, J. Immunol. 180, 1362–1372 (2008). Lung Cancer 99, 172–179 (2016).
plasma cells, CD20+ B cells, CD8+ and CD4+ T cells, 40. Nzula, S., Going, J. J. & Stott, D. I. Antigen-driven 62. Chen, H., Werner, S., Tao, S., Zornig, I. & Brenner, H.
IgG oligoclonality, tumour-associated antigens and clonal proliferation, somatic hypermutation, and Blood autoantibodies against tumor-associated
prognostic benefit. selection of B lymphocytes infiltrating human ductal antigens as biomarkers in early detection of colorectal
19. Charoentong, P. et al. Pan-cancer immunogenomic breast carcinomas. Cancer Res. 63, 3275–3280 cancer. Cancer Lett. 346, 178–187 (2014).
analyses reveal genotype-immunophenotype (2003). 63. Zayakin, P. et al. Tumor-associated autoantibody
relationships and predictors of response to checkpoint 41. Cipponi, A. et al. Neogenesis of lymphoid structures signature for the early detection of gastric cancer.
blockade. Cell Rep. 18, 248–262 (2017). and antibody responses occur in human melanoma Int. J. Cancer 132, 137–147 (2013).
20. Dang, V. D., Hilgenberg, E., Ries, S., Shen, P. & metastases. Cancer Res. 72, 3997–4007 (2012). 64. Kurtenkov, O. et al. IgG immune response to
Fillatreau, S. From the regulatory functions of B cells 42. Mose, L. E. et al. Assembly-based inference of B-cell tumor-associated carbohydrate antigens (TF, Tn,
to the identification of cytokine-producing plasma cell receptor repertoires from short read RNA sequencing alphaGal) in patients with breast cancer: impact of
subsets. Curr. Opin. Immunol. 28, 77–83 (2014). data with V’DJer. Bioinformatics 32, 3729–3734 neoadjuvant chemotherapy and relation to the survival.
21. Gilbert, A. E. et al. Monitoring the systemic human (2016). Exp. Oncol. 27, 136–140 (2005).
memory B cell compartment of melanoma patients 43. Bolotin, D. A. et al. Antigen receptor repertoire 65. Kumar, S., Mohan, A. & Guleria, R. Prognostic
for anti-tumor IgG antibodies. PLoS One 6, e19330 profiling from RNA-seq data. Nat. Biotechnol. 35, implications of circulating anti-p53 antibodies in lung
(2011). 908–911 (2017). cancer–a review. Eur. J. Cancer Care 18, 248–254
22. Kurai, J. et al. Antibody-dependent cellular cytotoxicity The work reports an association of high (2009).
mediated by cetuximab against lung cancer cell lines. intratumoural IgG1 proportions and clonality 66. Garaud, S. et al. Antigen specificity and clinical
Clin. Cancer Res. 13, 1552–1561 (2007). with increased survival in human melanoma. significance of IgG and IgA autoantibodies produced
23. Carmi, Y. et al. Allogeneic IgG combined with dendritic 44. Iglesia, M. D. et al. Genomic analysis of immune cell in situ by tumor-infiltrating B cells in breast cancer.
cell stimuli induce antitumour T-cell immunity. Nature infiltrates across 11 tumor types. J. Natl Cancer Inst. Front. Immunol. 9, 2660 (2018).
521, 99–104 (2015). 108, djw144 (2016). 67. Hamanaka, Y. et al. Circulating anti-MUC1 IgG
24. Rivera, A., Chen, C. C., Ron, N., Dougherty, J. P. 45. Isaeva, O. I. et al. Intratumoral immunoglobulin antibodies as a favorable prognostic factor for
& Ron, Y. Role of B cells as antigen-presenting cells isotypes predict survival in lung adenocarcinoma pancreatic cancer. Int. J. Cancer 103, 97–100 (2003).
in vivo revisited: antigen-specific B cells are essential subtypes. J. Immunother. Cancer 7, 279 (2019). 68. Kurtenkov, O. et al. Humoral immune response to
for T cell expansion in lymph nodes and for systemic This work reports the association of high MUC1 and to the Thomsen-Friedenreich (TF)
T cell responses to low antigen concentrations. intratumoural IgG1 and IgG4 proportions with glycotope in patients with gastric cancer: relation to
Int. Immunol. 13, 1583–1593 (2001). increased survival in KRAS-mutant and STK11- survival. Acta Oncol. 46, 316–323 (2007).
25. Bruno, T. C. et al. Antigen-presenting intratumoral mutant lung adenocarcinomas, respectively, thereby 69. Hirasawa, Y. et al. Natural autoantibody to MUC1
B cells affect CD4+ TIL phenotypes in non-small cell linking driver mutations and B cell response. is a prognostic indicator for non-small cell lung cancer.
lung cancer patients. Cancer Immunol. Res. 5, 46. Lohr, M. et al. The prognostic relevance of tumour- Am. J. Respir. Crit. Care Med. 161, 589–594 (2000).
898–907 (2017). infiltrating plasma cells and immunoglobulin kappa C 70. Fremd, C. et al. Mucin 1-specific B cell immune
26. Rossetti, R. A. M. et al. B lymphocytes can be activated indicates an important role of the humoral immune responses and their impact on overall survival in
to act as antigen presenting cells to promote anti-tumor response in non-small cell lung cancer. Cancer Lett. breast cancer patients. Oncoimmunology 5,
responses. PLoS One 13, e0199034 (2018). 333, 222–228 (2013). e1057387 (2016).
27. Pitzalis, C., Jones, G. W., Bombardieri, M. & Jones, S. A. 47. Mohammed, Z. M., Going, J. J., Edwards, J., 71. Brockhausen, I., Yang, J. M., Burchell, J., Whitehouse, C.
Ectopic lymphoid-like structures in infection, cancer Elsberger, B. & McMillan, D. C. The relationship & Taylor-Papadimitriou, J. Mechanisms underlying
and autoimmunity. Nat. Rev. Immunol. 14, 447–462 between lymphocyte subsets and clinico-pathological aberrant glycosylation of MUC1 mucin in breast cancer
(2014). determinants of survival in patients with primary cells. Eur. J. Biochem. 233, 607–617 (1995).
28. Zhu, W. et al. A high density of tertiary lymphoid operable invasive ductal breast cancer. Br. J. Cancer 72. Nath, S. & Mukherjee, P. MUC1: a multifaceted
structure B cells in lung tumors is associated with 109, 1676–1684 (2013). oncoprotein with a key role in cancer progression.
increased CD4+ T cell receptor repertoire clonality. 48. Gentles, A. J. et al. The prognostic landscape of genes Trends Mol. Med. 20, 332–342 (2014).
Oncoimmunology 4, e1051922 (2015). and infiltrating immune cells across human cancers. 73. Haddon, L. & Hugh, J. MUC1-mediated motility in
29. Sautes-Fridman, C., Petitprez, F., Calderaro, J. Nat. Med. 21, 938–945 (2015). breast cancer: a review highlighting the role of the
& Fridman, W. H. Tertiary lymphoid structures in the 49. Zhou, S. et al. Mapping the high throughput SEREX MUC1/ICAM-1/Src signaling triad. Clin. Exp. Metastasis
era of cancer immunotherapy. Nat. Rev. Cancer 19, technology screening for novel tumor antigens. Comb. 32, 393–403 (2015).
307–325 (2019). Chem. High Throughput Screen. 15, 202–215 (2012). 74. Pimenta, E. M. & Barnes, B. J. Role of tertiary
30. Shi, J. Y. et al. Margin-infiltrating CD20+ B cells display 50. Fischer, E. et al. Cryptic epitopes induce high-titer lymphoid structures (TLS) in anti-tumor immunity:
an atypical memory phenotype and correlate with humoral immune response in patients with cancer. potential tumor-induced cytokines/chemokines that
favorable prognosis in hepatocellular carcinoma. J. Immunol. 185, 3095–3102 (2010). regulate TLS formation in epithelial-derived cancers.
Clin. Cancer Res. 19, 5994–6005 (2013). 51. Ishikawa, T. et al. Tumor-specific immunological Cancers 6, 969–997 (2014).
This study reports tumour-infiltrating antigen- recognition of frameshift-mutated peptides in colon 75. Radbruch, A. et al. Competence and competition:
experienced IgG+ B cells that produce IFNγ, cancer with microsatellite instability. Cancer Res. 63, the challenge of becoming a long-lived plasma cell.
interleukin-12 subunit p40, granzyme B and TRAIL 5564–5572 (2003). Nat. Rev. Immunol. 6, 741–750 (2006).
and cooperate with CD8+ T cells. 52. Kahles, A. et al. Comprehensive analysis of alternative 76. Wilmore, J. R. & Allman, D. Here, there, and anywhere?
31. Coronella, J. A. et al. Antigen-driven oligoclonal splicing across tumors from 8,705 patients. Cancer Cell Arguments for and against the physical plasma cell
expansion of tumor-infiltrating B cells in infiltrating 34, 211–224 (2018). survival niche. J. Immunol. 199, 839–845 (2017).

www.nature.com/nri
Reviews

77. DeFalco, J. et al. Non-progressing cancer patients 99. Hu, X. et al. Landscape of B cell immunity and related 125. Bruhns, P. et al. Specificity and affinity of human
have persistent B cell responses expressing shared immune evasion in human cancers. Nat. Genet. 51, Fcgamma receptors and their polymorphic variants
antibody paratopes that target public tumor antigens. 560–567 (2019). for human IgG subclasses. Blood 113, 3716–3725
Clin. Immunol. 187, 37–45 (2018). 100. Vidarsson, G., Dekkers, G. & Rispens, T. IgG subclasses (2009).
This study reports high levels of blood and allotypes: from structure to effector functions. This study shows the high affinity of monomeric
plasmablasts in patients with cancer, further Front. Immunol. 5, 520 (2014). IgG3, but not IgG1, for FcγRIIIA receptors
increasing in response to anti-CTLA4 therapy. 101. Schroeder, H. W. Jr. & Cavacini, L. Structure and expressed on macrophages and NK cells.
Antibodies cloned form these plasmablasts function of immunoglobulins. J. Allergy Clin. Immunol. 126. Zhao, J., Nussinov, R. & Ma, B. Antigen binding
recognized tumour tissues from other patients. 125, S41–S52 (2010). allosterically promotes Fc receptor recognition.
78. Gerstl, B., Eng, L. F. & Bigbee, J. W. Tumor-associated 102. Baker, K. et al. Neonatal Fc receptor expression in MAbs 11, 58–74 (2019).
immunoglobulins in pulmonary carcinoma. Cancer dendritic cells mediates protective immunity against 127. Bowen, A. & Casadevall, A. Revisiting the
Res. 37, 4449–4455 (1977). colorectal cancer. Immunity 39, 1095–1107 (2013). immunoglobulin intramolecular signaling hypothesis.
This study was one of the first to directly report 103. Rafiq, K., Bergtold, A. & Clynes, R. Immune complex- Trends Immunol. 37, 721–723 (2016).
the presence of IgG, IgA, and IgM in human solid mediated antigen presentation induces tumor immunity. 128. Tumeh, P. C. et al. PD-1 blockade induces responses
tumour tissues using an immunohistochemical J. Clin. Invest. 110, 71–79 (2002). by inhibiting adaptive immune resistance. Nature 515,
method. 104. Noujaim, A. A., Schultes, B. C., Baum, R. P. & 568–571 (2014).
79. Streets, A. J., Brooks, S. A., Dwek, M. V. & Madiyalakan, R. Induction of CA125-specific B 129. Saul, L. et al. IgG subclass switching and clonal
Leathem, A. J. Identification, purification and analysis and T cell responses in patients injected with expansion in cutaneous melanoma and normal skin.
of a 55 kDa lectin binding glycoprotein present in MAb-B43.13–evidence for antibody-mediated Sci. Rep. 6, 29736 (2016).
breast cancer tissue. Clin. Chim. Acta 254, 47–61 antigen-processing and presentation of CA125 in vivo. 130. Vollmers, C., Sit, R. V., Weinstein, J. A., Dekker, C. L. &
(1996). Cancer Biother. Radiopharm. 16, 187–203 (2001). Quake, S. R. Genetic measurement of memory B-cell
80. Pavoni, E. et al. Tumor-infiltrating B lymphocytes 105. Platzer, B., Stout, M. & Fiebiger, E. Antigen cross- recall using antibody repertoire sequencing. Proc. Natl
as an efficient source of highly specific immunoglobulins presentation of immune complexes. Front. Immunol. Acad. Sci. USA 110, 13463–13468 (2013).
recognizing tumor cells. BMC Biotechnol. 7, 70 5, 140 (2014). 131. Jiang, N. et al. Lineage structure of the human
(2007). 106. Collins, A. M. & Jackson, K. J. A temporal model of antibody repertoire in response to influenza
81. Nelson, B. H. CD20+ B cells: the other tumor-infiltrating human IgE and IgG antibody function. Front. Immunol. vaccination. Sci. Transl Med. 5, 171ra119 (2013).
lymphocytes. J. Immunol. 185, 4977–4982 (2010). 4, 235 (2013). 132. Ye, J., Ma, N., Madden, T. L. & Ostell, J. M.
82. Montfort, A. et al. A strong B-cell response is part of 107. Colbeck, E. J., Ager, A., Gallimore, A. & Jones, G. W. IgBLAST: an immunoglobulin variable domain
the immune landscape in human high-grade serous Tertiary lymphoid structures in cancer: drivers of sequence analysis tool. Nucleic Acids Res. 41,
ovarian metastases. Clin. Cancer Res. 23, 250–262 antitumor immunity, immunosuppression, or W34–W40 (2013).
(2017). bystander sentinels in disease? Front. Immunol. 8, 133. Laserson, U. et al. High-resolution antibody dynamics
83. Hansen, M. H., Nielsen, H. V. & Ditzel, H. J. 1830 (2017). of vaccine-induced immune responses. Proc. Natl
Translocation of an intracellular antigen to the surface 108. Shao, Y. et al. Regulatory B cells accelerate Acad. Sci. USA 111, 4928–4933 (2014).
of medullary breast cancer cells early in apoptosis hepatocellular carcinoma progression via CD40/ 134. Kaplinsky, J. et al. Antibody repertoire deep
allows for an antigen-driven antibody response CD154 signaling pathway. Cancer Lett. 355, sequencing reveals antigen-independent selection in
elicited by tumor-infiltrating B cells. J. Immunol. 169, 264–272 (2014). maturing B cells. Proc. Natl Acad. Sci. USA 111,
2701–2711 (2002). 109. Shalapour, S. et al. Inflammation-induced IgA+ cells E2622–E2629 (2014).
84. Shah, S. et al. Increased rejection of primary tumors in dismantle anti-liver cancer immunity. Nature 551, 135. Khan, T. A. et al. Accurate and predictive antibody
mice lacking B cells: inhibition of anti-tumor CTL and 340–345 (2017). repertoire profiling by molecular amplification
TH1 cytokine responses by B cells. Int. J. Cancer 117, 110. Welinder, C. et al. Intra-tumour IgA1 is common fingerprinting. Sci. Adv. 2, e1501371 (2016).
574–586 (2005). in cancer and is correlated with poor prognosis in 136. Vander Heiden, J. A. et al. pRESTO: a toolkit for
85. Inoue, S., Leitner, W. W., Golding, B. & Scott, D. bladder cancer. Heliyon 2, e00143 (2016). processing high-throughput sequencing raw reads of
Inhibitory effects of B cells on antitumor immunity. 111. Stavnezer, J. & Kang, J. The surprising discovery lymphocyte receptor repertoires. Bioinformatics 30,
Cancer Res. 66, 7741–7747 (2006). that TGF beta specifically induces the IgA class switch. 1930–1932 (2014).
86. Kroemer, G., Galluzzi, L., Kepp, O. & Zitvogel, L. J. Immunol. 182, 5–7 (2009). 137. Shugay, M. et al. Towards error-free profiling of
Immunogenic cell death in cancer therapy. Annu. Rev. 112. Park, K.-H., Seo, G.-Y., Jang, Y.-S. & Kim, P.-H. TGF-β immune repertoires. Nat. Methods 11, 653–655
Immunol. 31, 51–72 (2013). and BAFF derived from CD4+CD25+Foxp3+ T cells (2014).
87. Shalapour, S. et al. Immunosuppressive plasma cells mediate mouse IgA isotype switching. Genes Genomics 138. Gupta, N. T. et al. Change-O: a toolkit for analyzing
impede T-cell-dependent immunogenic chemotherapy. 34, 619–625 (2012). large-scale B cell immunoglobulin repertoire
Nature 521, 94–98 (2015). 113. Cong, Y., Feng, T., Fujihashi, K., Schoeb, T. R. & sequencing data. Bioinformatics 31, 3356–3358
This study reveals immunosuppressive tumour- Elson, C. O. A dominant, coordinated T regulatory (2015).
infiltrating IgA+ plasma cells that express IL-10 and cell-IgA response to the intestinal microbiota. Proc. 139. Turchaninova, M. A. et al. High-quality full-length
PDL1. Natl Acad. Sci. USA 106, 19256–19261 (2009). immunoglobulin profiling with unique molecular
88. Perricone, M. A. et al. Enhanced efficacy of 114. Wang, L. et al. T regulatory cells and B cells barcoding. Nat. Protoc. 11, 1599–1616 (2016).
melanoma vaccines in the absence of B lymphocytes. cooperate to form a regulatory loop that maintains 140. Canzar, S., Neu, K. E., Tang, Q., Wilson, P. C. &
J. Immunother. 27, 273–281 (2004). gut homeostasis and suppresses dextran sulfate Khan, A. A. BASIC: BCR assembly from single cells.
89. Oizumi, S. et al. Surmounting tumor-induced immune sodium-induced colitis. Mucosal Immunol. 8, Bioinformatics 33, 425–427 (2017).
suppression by frequent vaccination or immunization 1297–1312 (2015). 141. Zhang, W. et al. Characterization of the B cell
in the absence of B cells. J. Immunother. 31, 394–401 115. Bauche, D. & Marie, J. C. Transforming growth receptor repertoire in the intestinal mucosa and
(2008). factor β: a master regulator of the gut microbiota and of tumor-infiltrating lymphocytes in colorectal
90. Ou, Z. et al. Tumor microenvironment B cells increase immune cell interactions. Clin. Transl Immunol. 6, adenoma and carcinoma. J. Immunol. 198,
bladder cancer metastasis via modulation of the IL-8/ e136 (2017). 3719–3728 (2017).
androgen receptor (AR)/MMPs signals. Oncotarget 6, 116. Disis, M. L., Watt, W. C. & Cecil, D. L. Th1 epitope 142. Kardos, J. et al. Claudin-low bladder tumors are
26065–26078 (2015). selection for clinically effective cancer vaccines. immune infiltrated and actively immune suppressed.
91. Woo, J. R. et al. Tumor infiltrating B-cells are increased Oncoimmunology 3, e954971 (2014). JCI Insight 1, e85902 (2016).
in prostate cancer tissue. J. Transl Med. 12, 30 117. Chen, K. et al. Immunoglobulin D enhances 143. Meng, Q., Valentini, D., Rao, M. & Maeurer, M.
(2014). immune surveillance by activating antimicrobial, KRAS RENAISSANCE(S) in tumor infiltrating
92. Aziz, M., Das, T. K. & Rattan, A. Role of circulating proinflammatory and B cell-stimulating programs in B cells in pancreatic cancer. Front. Oncol. 8, 384
immune complexes in prognostic evaluation and basophils. Nat. Immunol. 10, 889–898 (2009). (2018).
management of genitourinary cancer patients. 118. Shan, M. et al. Secreted IgD amplifies humoral 144. Cafri, G. et al. Memory T cells targeting oncogenic
Indian J. Cancer 34, 111–120 (1997). T helper 2 cell responses by binding basophils via mutations detected in peripheral blood of
93. Gunderson, A. J. & Coussens, L. M. B cells and their galectin-9 and CD44. Immunity 49, 709–724 e708 epithelial cancer patients. Nat. Commun. 10,
mediators as targets for therapy in solid tumors. (2018). 449 (2019).
Exp. Cell Res. 319, 1644–1649 (2013). 119. Harada, K. & Nakanuma, Y. Cholangiocarcinoma 145. Reddy, S. T. et al. Monoclonal antibodies isolated
94. Barbera-Guillem, E., May, K. F. Jr., Nyhus, J. K. & with respect to IgG4 reaction. Int. J. Hepatol. 2014, without screening by analyzing the variable-gene
Nelson, M. B. Promotion of tumor invasion by 803876 (2014). repertoire of plasma cells. Nat. Biotechnol. 28,
cooperation of granulocytes and macrophages 120. Chiaruttini, G. et al. B cells and the humoral response 965–969 (2010).
activated by anti-tumor antibodies. Neoplasia 1, in melanoma: the overlooked players of the tumor 146. DeKosky, B. J. et al. In-depth determination and
453–460 (1999). microenvironment. Oncoimmunology 6, e1294296 analysis of the human paired heavy- and light-chain
95. de Visser, K. E., Korets, L. V. & Coussens, L. M. (2017). antibody repertoire. Nat. Med. 21, 86–91 (2015).
De novo carcinogenesis promoted by chronic 121. Karagiannis, P. et al. IgG4 subclass antibodies impair 147. Briggs, A. W. et al. Tumor-infiltrating immune
inflammation is B lymphocyte dependent. Cancer Cell antitumor immunity in melanoma. J. Clin. Invest. 123, repertoires captured by single-cell barcoding in
7, 411–423 (2005). 1457–1474 (2013). emulsion. Preprint at bioRxiv https://doi.org/
96. Tan, T. T. & Coussens, L. M. Humoral immunity, 122. Fujimoto, M. et al. Stromal plasma cells expressing 10.1101/134841 (2017).
inflammation and cancer. Curr. Opin. Immunol. 19, immunoglobulin G4 subclass in non-small cell lung 148. Seah, Y. F. S., Hu, H. & Merten, C. A. Microfluidic
209–216 (2007). cancer. Hum. Pathol. 44, 1569–1576 (2013). single-cell technology in immunology and antibody
97. Andreu, P. et al. FcRgamma activation regulates 123. van der Neut Kolfschoten, M. et al. Anti-inflammatory screening. Mol. Asp. Med. 59, 47–61 (2018).
inflammation-associated squamous carcinogenesis. activity of human IgG4 antibodies by dynamic Fab 149. Busse, C. E., Czogiel, I., Braun, P., Arndt, P. F. &
Cancer Cell 17, 121–134 (2010). arm exchange. Science 317, 1554–1557 (2007). Wardemann, H. Single-cell based high-throughput
98. Yuen, G. J., Demissie, E. & Pillai, S. B lymphocytes 124. Morell, A., Terry, W. D. & Waldmann, T. A. Metabolic sequencing of full-length immunoglobulin heavy and
and cancer: a love-hate relationship. Trends Cancer 2, properties of IgG subclasses in man. J. Clin. Invest. light chain genes. Eur. J. Immunol. 44, 597–603
747–757 (2016). 49, 673–680 (1970). (2014).

Nature Reviews | Immunology


Reviews

150. Eyer, K. et al. Single-cell deep phenotyping of 171. Cheng, H. et al. Kras(G12D) mutation contributes to 195. Wang, W. W. et al. CD19+CD24hiCD38hiBregs
IgG-secreting cells for high-resolution immune regulatory T cell conversion through activation of the involved in downregulate helper T cells and upregulate
monitoring. Nat. Biotechnol. 35, 977–982 (2017). MEK/ERK pathway in pancreatic cancer. Cancer Lett. regulatory T cells in gastric cancer. Oncotarget 6,
This work presents a microfluidic system that 446, 103–111 (2019). 33486–33499 (2015).
allows high-throughput screening for antigen- 172. Thorsson, V. et al. The immune landscape of cancer. 196. Shimabukuro-Vornhagen, A. et al. Characterization
specific antibody-secreting cells, simultaneously Immunity 48, 812–830 e814 (2018). of tumor-associated B-cell subsets in patients
measuring the antibody secretion rate and affinity. 173. Kuroki, M. & Shirasu, N. Novel treatment strategies with colorectal cancer. Oncotarget 5, 4651–4664
151. McDaniel, J. R. et al. Identification of tumor-reactive for cancer and their tumor-targeting approaches (2014).
B cells and systemic IgG in breast cancer based on using antibodies against tumor-associated antigens. 197. Aklilu, M. et al. Depletion of normal B cells with
clonal frequency in the sentinel lymph node. Cancer Anticancer Res. 34, 4481–4488 (2014). rituximab as an adjunct to IL-2 therapy for renal
Immunol. Immunother. 67, 729–738 (2018). 174. Bacac, M. et al. A novel carcinoembryonic antigen cell carcinoma and melanoma. Ann. Oncol. 15,
In this work, paired VH and VL repertoires are T-cell bispecific antibody (CEA TCB) for the treatment 1109–1114 (2004).
obtained from sentinel lymph node B cells, followed of solid tumors. Clin. Cancer Res. 22, 3286–3297 198. Griss, J. et al. B cells sustain inflammation and
by screening for NY-ESO-1-specific antibody (2016). predict response to immune checkpoint blockade
variants, the presence of which in the patient 175. Trenevska, I., Li, D. & Banham, A. H. Therapeutic in human melanoma. Nat. Commun. 10, 4186
serum was confirmed by mass spectrometry of antibodies against intracellular tumor antigens. (2019).
enriched NY-ESO-1-specific IgG antibodies. Front. Immunol. 8, 1001 (2017). 199. Bodogai, M. et al. Anti-CD20 antibody promotes
152. Kim, S. et al. B-cell depletion using an anti-CD20 176. Goc, J. et al. Dendritic cells in tumor-associated cancer escape via enrichment of tumor-evoked
antibody augments antitumor immune responses tertiary lymphoid structures signal a Th1 cytotoxic regulatory B cells expressing low levels of CD20
and immunotherapy in nonhematopoetic murine immune contexture and license the positive prognostic and CD137L. Cancer Res. 73, 2127–2138
tumor models. J. Immunother. 31, 446–457 (2008). value of infiltrating CD8+ T cells. Cancer Res. 74, (2013).
153. Affara, N. I. et al. B cells regulate macrophage 705–715 (2014). 200. Bodogai, M. et al. Failure of rituximab in solid
phenotype and response to chemotherapy in squamous 177. Silina, K., Rulle, U., Kalnina, Z. & Line, A. Manipulation tumors is due to its inability to eliminate tumor evoked
carcinomas. Cancer Cell 25, 809–821 (2014). of tumour-infiltrating B cells and tertiary lymphoid B regulatory cells. J. Immunol. 188 (Suppl. 1), 165.15
154. Engelhard, V. H. et al. Immune cell infiltration and structures: a novel anti-cancer treatment avenue? (2012).
tertiary lymphoid structures as determinants of Cancer Immunol. Immunother. 63, 643–662 201. Halliley, J. L. et al. Long-lived plasma cells are
antitumor immunity. J. Immunol. 200, 432–442 (2014). contained within the CD19-CD38hiCD138+ subset
(2018). 178. Dubey, L. K. et al. Lymphotoxin-dependent B cell-FRC in human bone marrow. Immunity 43, 132–145
155. Hiraoka, N. et al. Intratumoral tertiary lymphoid crosstalk promotes de novo follicle formation and (2015).
organ is a favourable prognosticator in patients with antibody production following intestinal helminth 202. Krejcik, J. et al. Daratumumab depletes CD38+
pancreatic cancer. Br. J. Cancer 112, 1782–1790 infection. Cell Rep. 15, 1527–1541 (2016). immune regulatory cells, promotes T-cell expansion,
(2015). 179. Litsiou, E. et al. CXCL13 production in B cells via and skews T-cell repertoire in multiple myeloma. Blood
156. Andre, P. et al. Anti-NKG2A mAb is a checkpoint Toll-like receptor/lymphotoxin receptor signaling is 128, 384–394 (2016).
inhibitor that promotes anti-tumor immunity by involved in lymphoid neogenesis in chronic obstructive 203. Manna, A., Lewis-Tuffin, L. J., Ailawadhi, S.,
unleashing both T and NK cells. Cell 175, 1731–1743 pulmonary disease. Am. J. Respir. Crit. Care Med. Chanan-Khan, A. A. & Paulus, A. Using anti-CD38
(2018). 187, 1194–1202 (2013). immunotherapy to enhance anti-tumor T-cell immunity
157. Hsu, J. et al. Contribution of NK cells to immunotherapy 180. Bergomas, F. et al. Tertiary intratumor lymphoid tissue in chronic lymphocytic leukemia (CLL). J. Immunol.
mediated by PD-1/PD-L1 blockade. J. Clin. Invest. 128, in colo-rectal cancer. Cancers 4, 1–10 (2011). 200, 58.17 (2018).
4654–4668 (2018). 181. Coppola, D. et al. Unique ectopic lymph node-like 204. Pinto, D. et al. A functional BCR in human IgA
158. Sanchez-Correa, B. et al. Modulation of NK cells structures present in human primary colorectal and IgM plasma cells. Blood 121, 4110–4114
with checkpoint inhibitors in the context of cancer carcinoma are identified by immune gene array (2013).
immunotherapy. Cancer Immunol. Immunother. 68, profiling. Am. J. Pathol. 179, 37–45 (2011). 205. Molina-Cerrillo, J., Alonso-Gordoa, T., Gajate, P. &
861–870 (2019). 182. Thommen, D. S. et al. A transcriptionally and Grande, E. Bruton’s tyrosine kinase (BTK) as a
159. Cottrell, T. R. et al. Pathologic features of response to functionally distinct PD-1+ CD8+ T cell pool with promising target in solid tumors. Cancer Treat. Rev.
neoadjuvant anti-PD-1 in resected non-small-cell lung predictive potential in non-small-cell lung cancer 58, 41–50 (2017).
carcinoma: a proposal for quantitative immune-related treated with PD-1 blockade. Nat. Med. 24, 206. Stiff, A. et al. Myeloid-derived suppressor cells
pathologic response criteria (irPRC). Ann. Oncol. 29, 994–1004 (2018). express Bruton’s tyrosine kinase and can be depleted
1853–1860 (2018). 183. Joshi, N. S. et al. Regulatory T cells in tumor- in tumor-bearing hosts by ibrutinib treatment.
160. Helmink, B. A. et al. B cells and tertiary lymphoid associated tertiary lymphoid structures suppress Cancer Res. 76, 2125–2136 (2016).
structures promote immunotherapy response. Nature anti-tumor T cell responses. Immunity 43, 579–590 207. Sagiv-Barfi, I. et al. Therapeutic antitumor immunity
https://doi.org/10.1038/s41586-019-1922-8 (2020). (2015). by checkpoint blockade is enhanced by ibrutinib,
161. Cabrita, R. et al. Tertiary lymphoid structures improve 184. Mahoney, K. M. et al. A secreted PD-L1 splice an inhibitor of both BTK and ITK. Proc. Natl Acad.
immunotherapy and survival in melanoma. Nature variant that covalently dimerizes and mediates Sci. USA 112, E966–E972 (2015).
https://doi.org/10.1038/s41586-019-1914-8 (2020). immunosuppression. Cancer Immunol. Immunother. 208. Nakayamada, S. & Tanaka, Y. BAFF- and
162. Petitprez F. et al. B cells are associated with survival 68, 421–432 (2019). APRIL-targeted therapy in systemic autoimmune
and immunotherapy response in sarcoma. Nature 185. Ammirante, M., Luo, J. L., Grivennikov, S., diseases. Inflamm. Regen. 36, 6 (2016).
https://doi.org/10.1038/s41586-019-1906-8 (2020). Nedospasov, S. & Karin, M. B-cell-derived lymphotoxin
163. Lutz, E. R. et al. Immunotherapy converts promotes castration-resistant prostate cancer. Nature Author contributions
nonimmunogenic pancreatic tumors into immunogenic 464, 302–305 (2010). All of the authors contributed to researching data for the
foci of immune regulation. Cancer Immunol. Res. 2, 186. Balkwill, F., Montfort, A. & Capasso, M. B article, the discussion of content and the writing of the article.
616–631 (2014). regulatory cells in cancer. Trends Immunol. 34, D.M.C. and G.V.S. reviewed and edited the manuscript before
164. Maldonado, L. et al. Intramuscular therapeutic 169–173 (2013). submission.
vaccination targeting HPV16 induces T cell responses 187. Schwartz, M., Zhang, Y. & Rosenblatt, J. D. B cell
that localize in mucosal lesions. Sci. Transl Med. 6, regulation of the anti-tumor response and role Competing interests
221ra213 (2014). in carcinogenesis. J. Immunother. Cancer 4, 40 The authors declare no competing interests.
165. Angelova, M. et al. Characterization of the (2016).
immunophenotypes and antigenomes of colorectal 188. Sarvaria, A., Madrigal, J. A. & Saudemont, A. Funding
cancers reveals distinct tumor escape mechanisms B cell regulation in cancer and anti-tumor immunity. The work was supported by grants from the Ministry of
and novel targets for immunotherapy. Genome Biol. Cell Mol. Immunol. 14, 662–674 (2017). Education and Science of the Russian Federation (14.W03.
16, 64 (2015). 189. Qin, Z. et al. B cells inhibit induction of T cell-dependent 31.0005) and Russian Science Foundation (19-14-00317, in
166. Senbabaoglu, Y. et al. Tumor immune microenvironment tumor immunity. Nat. Med. 4, 627–630 (1998). part of antibody repertoire analysis methods).
characterization in clear cell renal cell carcinoma 190. Carter, N. A. et al. Mice lacking endogenous IL-10-
identifies prognostic and immunotherapeutically producing regulatory B cells develop exacerbated
Peer review information
relevant messenger RNA signatures. Genome Biol. 17, disease and present with an increased frequency
Nature Reviews Immunology thanks K. Willard-Gallo and
231 (2016). of Th1/Th17 but a decrease in regulatory T cells.
the other, anonymous, reviewer(s) for their contribution to the
167. Becht, E. et al. Immune and stromal classification J. Immunol. 186, 5569–5579 (2011).
peer review of this work.
of colorectal cancer is associated with molecular 191. Olkhanud, P. B. et al. Tumor-evoked regulatory B cells
subtypes and relevant for precision immunotherapy. promote breast cancer metastasis by converting
Clin. Cancer Res. 22, 4057–4066 (2016). resting CD4+ T cells to T-regulatory cells. Cancer Res. Publisher’s note
168. Miller, L. D. et al. Immunogenic subtypes of breast 71, 3505–3515 (2011). Springer Nature remains neutral with regard to jurisdictional
cancer delineated by gene classifiers of immune 192. Cai, C. et al. Interleukin 10-expressing B cells inhibit claims in published maps and institutional affiliations.
responsiveness. Cancer Immunol. Res. 4, 600–610 tumor-infiltrating T cell function and correlate with
(2016). T cell Tim-3 expression in renal cell carcinoma. Supplementary information
169. Biton, J. et al. TP53, STK11, and EGFR mutations Tumour Biol. 37, 8209–8218 (2016). Supplementary information is available for this paper at
predict tumor immune profile and the response to 193. Pylayeva-Gupta, Y. et al. IL35-producing B cells https://doi.org/10.1038/s41577-019-0257-x.
anti-PD-1 in lung adenocarcinoma. Clin. Cancer Res. promote the development of pancreatic neoplasia.
24, 5710–5723 (2018). Cancer Discov. 6, 247–255 (2016).
170. Schabath, M. B. et al. Differential association of 194. Zhou, X., Su, Y. X., Lao, X. M., Liang, Y. J. & Liao, G. Q. Related links
STK11 and TP53 with KRAS mutation-associated gene CD19+IL-10+ regulatory B cells affect survival of MiNNN: https://minnn.milaboratory.com
expression, proliferation and immune surveillance in tongue squamous cell carcinoma patients and induce MiXCR: https://mixcr.milaboratory.com
lung adenocarcinoma. Oncogene 35, 3209–3216 resting CD4+ T cells to CD4+Foxp3+ regulatory T cells.
(2016). Oral. Oncol. 53, 27–35 (2016). © Springer Nature Limited 2020

www.nature.com/nri

You might also like