You are on page 1of 15

REVIEWS

Microbiota: a key orchestrator


of cancer therapy
Soumen Roy and Giorgio Trinchieri
Abstract | The microbiota is composed of commensal bacteria and other microorganisms that live
on the epithelial barriers of the host. The commensal microbiota is important for the health and
survival of the organism. Microbiota influences physiological functions from the maintenance of
barrier homeostasis locally to the regulation of metabolism, haematopoiesis, inflammation,
immunity and other functions systemically. The microbiota is also involved in the initiation,
progression and dissemination of cancer both at epithelial barriers and in sterile tissues. Recently,
it has become evident that microbiota, and particularly the gut microbiota, modulates the
response to cancer therapy and susceptibility to toxic side effects. In this Review, we discuss
the evidence for the ability of the microbiota to modulate chemotherapy, radiotherapy and
immunotherapy with a focus on the microbial species involved, their mechanism of action and the
possibility of targeting the microbiota to improve anticancer efficacy while preventing toxicity.

Germ-free animals
The human microbiota is the ensemble of bacteria and haematopoietic cells of yolk sac origin such as the
Animals raised in strict sterile other microorganisms (archaea, fungi, protozoa, as well microglia in the central nervous system14–16. The gut
conditions that have no as human, fungal, bacterial and protozoan viruses) that microbiota interacting with epithelial and stromal intes-
microorganisms living in or on inhabit the epithelial barrier surfaces of our body 1. The tinal cells regulates barrier functions, mucosal immune
them.
microbiota affects physiological functions, particularly homeostasis7–9, host–microbiota symbiosis, prevention
Commensalism metabolism, neurological and cognitive functions, hae- of infestation by pathogens, control of overgrowth by
A symbiotic relationship matopoiesis, inflammation and immunity 2,3. Germ-free pathobionts, metabolism of indigestible dietary fibre,
between two species in which animals are functionally immature in many physiolog- synthesis of vitamins and regulation of metabolism,
one species benefits without ical systems, including innate immunity, and they are including the prevention of obesity 7–9,17–19. An intact
causing harm to the other.
highly susceptible to infection by pathogens4. However, and functional gut epithelium maintains a healthy
Pathobionts the germ-free condition in itself is not lethal. Germ-free body, and gut epithelial homeostasis is maintained
Resident commensal rodents maintained in a sheltered sterile environment by continuous crosstalk between the gut microbiota,
microorganisms that under and fed with a controlled diet containing factors, such immune cells and the mucosal barrier 20.
certain conditions may acquire
as vitamins that are normally supplied by the intesti- The composition of the microbiota is shaped by host
pathogenic potential.
nal microbiota, can survive significantly longer than genetics, colonization at the time of birth, type of birth
Mutualism conventionally raised animals4–6. The local microbiota delivery, an individual’s lifestyle, incidence of diseases
A symbiotic relationship affects the functions and regulates the immunity of the and exposure to antibiotics21–23. Microbiota composition
between two species that is epithelial barrier on which it resides7–10. In addition, evolves during the first few years of human life before
beneficial for both species.
the microbiota also exerts systemic effects10,11 (FIG. 1) maturation into an adult-like microbiota22. After that,
through mechanisms that are less well understood than the composition of the microbiota in the gut and other
those mediating the local effects. epithelial barriers remains relatively constant through-
The gut microbiota comprises approximately 3 × 1013 out adult life, although it could still be affected by diet,
Cancer and Inflammation bacterial cells that mostly exhibit commensalism with the changes in lifestyle, disease and disease treatment 24,25.
Program, Center for Cancer host 12. However, when the intestinal ecology is altered, The microbiota present at the epithelial barrier and
Research, National Cancer
commensal bacteria that are referred to as pathobionts particularly in the gut influences local and systemic
Institute, National Institutes
of Health, Bethesda, (for example, Clostridium difficile or vancomycin- metabolic functions, inflammation and adaptive immu-
Maryland 20892, USA. resistant Enterococcus) may expand and acquire nity, which modulate cancer initiation, progression
Correspondence to G.T. pathogenic characteristics13. The gut microbiota also and response to anticancer treatment. Host genetics and
trinchig@mail.nih.gov exhibits mutualism with the host that modulates immu- environmental factors, including nutrition, modify the
doi:10.1038/nrc.2017.13 nity, promotes bone marrow haematopoiesis and also composition of the microbiota both in humans and
Published online 17 Mar 2017 regulates maturation and function of tissue-resident in the mouse26–29. In the mouse, the pro-carcinogenic

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Local effects of Systemic effects of lifestyle in part may indirectly affect carcinogenesis and
gastrointestinal microbiota gastrointestinal microbiota
response to cancer therapy through modification of
microbiota composition.
• Translocation of bacteria or Owing to its ability to modulate host metabolism,
bacterial products and toxins
• Metabolites (e.g. small and inflammation and immunity, the microbiota is involved
medium chain fatty acids, in the initiation and/or progression of various types of
choline derivatives, secondary
bile acids, vitamins, hormones cancer both at the epithelial barriers and in sterile tis-
and nutrients) sues3 (BOX 1). Although much progress has been made
• Innate and adaptive immune in cancer therapy — and in particular recent advances
cell migration
• Cytokines in immunotherapy are very promising — a significant
• Endocrine (cortisol) and neural proportion of cancer patients still do not respond to
(vagus and enteric nervous therapy. In this Review we discuss the body of data in
system) pathways
experimental models and increasingly in clinical stud-
ies that suggests that the composition of the microbiota
regulates the efficacy of anticancer therapy and that tar-
• Nutrient absorption • Metabolism geting the microbiota may improve drug efficacy and/or
• Synthesis of vitamins • Neurological, behavioural and cognitive reduce adverse effects.
Physiological

• Metabolism of bile and functions


functions

hormones • Cardiovascular and musculoskeletal functions


• Fermentation of carbohydrates • Haematopoiesis and myeloid cell functions Chemotherapy and microbiota
• Morphogenesis • Circadian rhythm After the discovery of the cytotoxic effects of nitro-
• Barrier strengthening • Ageing gen mustards during the Second World War, cytotoxic
• Mucosal immunity • Inflammation and immunity
chemotherapeutic agents were developed that still today
Non-neoplastic

• Inflammatory bowel disease • Obesity, insulin resistance and type II diabetes remain a major staple of cancer therapy 34. Cytotoxic
• Gastritis and gastric ulcers • Cardiovascular diseases
pathology

drugs are classified according to their mechanisms of


• Autoimmunity: CNS, eye and joints
• Non-alcoholic steatohepatitis action, as alkylating agents, heavy metals including plati­
• Gout num, antimetabolites, cytotoxic antibiotics and spindle
• Stomach cancer (Helicobacter • MALT, ocular and skin lymphoma poisons. Most forms of chemotherapy display antitu-
pylori) • Thymic lymphoma mour activity by targeting DNA integrity and cell divi-
• Colorectal carcinoma (Escherichia • Hepatocellular carcinoma
Cancer

coli, Fusobacterium spp. • Mammary carcinoma


sion in the dividing cancer cells. Cytotoxicity may also
and enterotoxigenic Bacteroides • Pancreatic cancer be induced by the effect of drugs on other cellular com-
fragilis) • Prostate cancer partments such as mitochondria and cell membranes35.
• Gallbladder carcinoma • Sarcoma
(Salmonella enterica Typhi) • Ovarian cancer Chemotherapy is not specific and its use is always asso-
ciated with significant toxicity for tissues that have a high
• Cancer therapy gastrointestinal • Cancer chemotherapy, immunotherapy
therapy

rate of cell replacement and division36.


Cancer

toxicity and radiotherapy efficacy


• Cancer therapy toxicity
• Graft-versus-host disease Drug metabolism. The gut microbiota affects drug
Nature Reviews
Figure 1 | Local and systemic effects of the gastrointestinal microbiota. The | Cancer pharmacokinetics, anticancer activity and toxicity at
abundant microbiota present on the gastrointestinal mucosa affects local mucosal various levels3,37. A schematic representation of the role
homeostasis, functions and immunity7–9. Many of the mechanisms by which various of the microbiota on the metabolism of drugs following
bacterial species and their products and metabolites affect mucosal physiology and enteral (for example, oral) or parenteral (for example,
pathology have been described7–10. However, the presence and composition of the gut intravenous) administration is presented in FIG. 2. The
microbiota also systemically affects the functions of most physiological systems, the rate of absorption and bioavailability of many oral drugs
pathology and the response to therapy in distant organs10. Some of the demonstrated or depends on their exposure in the gut to both host and
proposed mechanisms by which the gastrointestinal microbiota can achieve this are
bacterial enzymes before entering the circulation38,39.
listed in the box at the top of the figure205. The microbiota and specific microbial species
Xenobiotics induce changes in the composition and
also affect neoplastic pathology both at the local level in the gastrointestinal
tract145,188,206,207 and systemically in organs that are not normally associated with the gut shape the physiology and gene expression of the gut
microbiota192–194,208–212. Although these mechanisms have been studied primarily for the microbiota40,41, further modulating its effect on drug
most abundant intestinal microbiota organisms, microorganisms colonizing other metabolism. Biotransformation of drugs mediated by the
epithelial barriers, for example, the mouth and the skin, are also expected to mediate gut microbiota includes many chemical reactions, with
both local and systemic effects195,196. In addition to its association with cancer reduction and hydrolysis affecting the largest number
development, the microbiota also has both a local and a systemic role in modulating the of drugs42. Other reactions include functional group
efficacy and toxicity of cancer therapy3. CNS, central nervous system; MALT, removal, N‑oxide cleavage, proteolysis, denitration,
mucosa-associated lymphoid tissue. deconjugation, amine formation and/or hydrolysis,
thiazole ring opening, acetylation and isoxazole scis-
phenotype expressed by some genetically mutated mice sion43. Commensal microorganisms also decrease the
has been shown to be transferred to wild-type mice by absorption of certain drugs by physical binding and
microbiota transfer 30–32, and the transfer of the faecal segregation44. More than 40 drugs have been shown
Xenobiotics microbiota of patients who are responsive to cancer ther- to be metabolized by the gut microbiota43. However,
Foreign chemical substances,
including drugs, that are not
apy into germ-free mice has been shown to endow those among anticancer drugs, only the nitroreduction of the
naturally produced by the animals with an ability to respond efficiently to the ther- radiation sensitizer misonidazole, the hydrolysis of
organism. apy 33. Thus, it can be speculated that host genetics and the antimetabolite methotrexate and the deconjugation

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Biotransformation of the liver-detoxified form of the topoisomerase I overexpress the genes encoding xenobiotic-sensing
The chemical alteration of a inhibitor irinotecan (also known as CPT‑11) (discussed receptors and transcription factors such as constitutive
xenobiotic, such as a drug, in more detail below) have clearly been shown to be androstane receptor (Car; also known as Nr1i3), CAR-
within the body. affected by the gut microbiota43. regulated gene P450 oxidoreductase (Por), aryl hydro-
Probiotic
In addition to the direct effect of microorgan- carbon receptor (Ahr), peroxisome proliferator-activated
Live microorganisms that are isms and microbial enzymes on drug absorption and receptor‑α (Ppara) and nuclear factor erythroid 2‑related
consumed by humans and metabo­lism39–41, the gut microbiota indirectly affects factor 2 (Nrf2; also known as Nfe2l2)45–49. This altered
animals as food supplements the metabolism of both orally and systemically delivered gene expression results in the faster metabolism of many
for their potential
drugs through modulation of gene expression and the xenobiotics in germ-free mice, supporting the role of the
health-promoting qualities.
physiology of the local mucosal barrier and of distant microbiota in regulating drug metabolism and detoxi-
organs such as the liver 45–47. In the livers of germ-free fication45. Interestingly, although colonization of germ-
mice compared with specific pathogen-free mice, some free mice with microbiota taken from conventionally
members of the large cytochrome P450 (Cyp450) gene raised mice re-establishes normal gene expression, colo-
family are increased (for example, the Cyp2a, Cyp2b and nization of germ-free mice with the probiotic preparation
Cyp3a families involved in xenobiotic steroid metabo- VSL#3 normalizes only a proportion of the genes altered
lism) whereas others are decreased (for example, the in germ-free mice48. Thus, the heterogeneity of response
Cyp4a family involved in fatty acid or arachidonic acid to drug therapy and susceptibility to toxic effects in can-
metabolism)45–49. Germ-free mice also consistently cer patients may be due at least in part to differences
in gut microbiota composition and activity between
individual patients50.
Box 1 | Microbiota effects on cancer initiation and progression In addition to oral drugs, injected drugs are also
The health of the organism requires maintenance of epithelial barriers on which the
in part firstly metabolized in the liver, before biliary
microbiota resides. The barriers maintain a peaceful relationship with commensal excretion into the gut, where they are exposed to the
microorganisms while protecting the host from pathogens and pathobionts. gut microbiota and can be further metabolized and
Dysbiosis, which is an alteration in the composition of the microbiota associated with reabsorbed (FIG. 2). Irinotecan is an intravenous chemo­
pathology, disrupts the physiological interaction between epithelial cells and the therapeutic drug used for colorectal cancer treat-
microbiota, results in breach of the barriers, induces inflammatory pathologies and ment 51. Irinotecan is transformed into its active form,
may contribute to cancer initiation and progression182–184. SN‑38, by liver and small intestine tissue carboxyl­
The cytokine interleukin‑18 (IL‑18) mediates IL‑22‑dependent intestinal mucosal esterase and then detoxified in the liver by host UDP-
protection. Mice unable to produce, process or respond to IL‑18 (for example, mice glucuronosyltransferases into inactive SN‑38‑G before
genetically deficient in the genes encoding IL‑18, IL‑18 receptor (IL‑18R), myeloid
being secreted into the gut 51. In the gut, SN‑38‑G can
differentiation primary response 88 (MYD88), inflammasomes or inflammasome
signalling molecules) display a dysbiosis that results in enhanced susceptibility to
be reconverted by bacterial β‑glucuronidases into active
chemically induced colitis and colon carcinogenesis32,170,185,186. This susceptibility can SN‑38, which induces significant intestinal toxicity and
be transferred to wild-type mice by cohousing or faecal transfer32,170. Deficiency for diarrhoea52. In tumour-bearing rats, irinotecan chemo-
other immunologically relevant genes may also enhance the susceptibility to colon therapy increased the abundance of Clostridium cluster XI
carcinogenesis owing to a dysbiosis that can be transferred to wild-type mice187. Some and Enterobacteriaceae in the proximal colon, indicat-
of the mechanisms by which microbial species either in humans or in experimental ing induction of an altered microbiota composition that
animals induce colorectal carcinogenesis have been characterized187–189. For example, could directly affect inflammation or change the propor-
Fusobacterium nucleatum, associated with human colorectal cancer, expands tion of bacterial species expressing β‑glucuronidase53.
tumour-promoting myeloid cell populations188,189, activates host β‑catenin–WNT In the human gut, β‑glucuronidase activity was found
signalling by the binding of its FadA adhesin to E‑cadherin188,189 and modulates
mostly in the Firmicutes phylum, particularly within
anticancer immune response by triggering the inhibitory receptor T cell
immunoreceptor with immunoglobulin (Ig) and ITIM domains (TIGIT) on T and natural
Clostridium clusters XIVa and IV54,55. Antibiotic treat-
killer cells via the microbial Fap2 protein190. However, among the various bacteria ments that decrease the number of β‑glucuronidase-
described to be associated with human cancer, only Helicobacter pylori has been positive bacterial species or the use of an inhibitor
proved to be a human carcinogen causing gastric cancer, as its elimination from the specific for bacterial β‑glucuronidase effectively treat
host prevents disease187,191. intestinal inflammation induced by irinotecan therapy
The gut microbiota also affects cancer at distant sites. Colonic infection with in experimental animals56,57. Use of probiotics in clini-
Helicobacter hepaticus not only enhances colon carcinogenesis but also favours the cal trials has shown a modest decrease in irinotecan‑
generation of mammary and prostate carcinoma in the ApcMin/+ mouse model of induced diarrhoea58. As the intestinal toxicity of irino-
colorectal cancer192 and augments chemical and viral transgenic liver tecan in humans is severe and dose limiting, there is
carcinogenesis184,193. In a mouse model of ataxia telangiectasia, the gut microbiota
much interest in the development of selective bacterial
composition observed in different mouse colonies or upon experimental
perturbation determines thymic lymphoma incidence and survival194. In humans, a
β‑glucuronidase inhibitors to be used in clinical trials
history of periodontal disease and antibodies against oral bacteria have been found to reduce toxicity 59.
to be associated with increased risk of pancreatic cancer195. Specifically, the
presence in the oral microbiota of Porphyromonas gingivalis and Aggregatibacter Response to chemotherapy. Direct interaction with
actinomycetemcomitans was observed to be significantly associated with increased bacteria can affect the efficacy of chemotherapeutic
risk of pancreatic cancer, and the presence of the phylum Fusobacterium was drugs60. Among 30 drugs tested in vitro in the presence
associated with decreased risk196. The effects of the microbiota on carcinogenesis in of non-pathogenic Gram-negative Escherichia coli or
distant organs are mediated in part by the modulation of the systemic inflammatory Gram-positive Listeria welshimeri, the activity of 10
tone, oxidative stress and inflammation-induced genotoxicity towards leukocytes of them was found to be inhibited by one or both spe-
and epithelial cells192–194,197.
cies whereas the efficacy of 6 others was enhanced60.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Enteral drug metabolism b Parenteral drug metabolism


E5 P2
E1
Approximately 10% of the drug Shortly after administration
Enteral drug enters the circulation via the >90% of the drug is
administration hepatic veins, and reaches available in the circulation
E2 for the target tumour
target tumours and other tissues
100% of the drug reaches the
GI tract and is absorbed into P3
the liver via the portal circulation
Systemic toxicity
Heart
First pass metabolism Lung
P4
E3 in the liver Hepatic vein
Phase I Each minute, 29% of
Altered gene Liver the circulating blood
expression Gall bladder P1 and drugs reach the
(CYP450 enzymes) Portal vein liver via the splanchnic
Parenteral drug circulation
Activation Bile duct administration
Stomach
Detoxification
Small intestine
Phase II Large intestine
Deconjugation
E4
~90% of the drug metabolites Gut microbiota Gut microbiota
from the liver are excreted • Biotransformation of xenobiotics Reactivation of liver detoxified xenobiotics
with bile into the intestinal affecting absorption and bioavailability
tract where they are excreted • Reactivation of liver-detoxified xenobiotics
in stools or reabsorbed
through the portal circulation Drug elimination by kidney Drug elimination by kidney
excretion and via the biliary tract excretion and via the biliary tract

Figure 2 | Major pathways of drug metabolism and the role of microbiota following enteral Nature
(forReviews
example,| Cancer
oral)
or parenteral (for example, intravenous) administration. a | Enteral drug metabolism. Orally administered drugs
(E1) sit in the stomach for 30–45 minutes before reaching the intestine and being absorbed into the liver by the
portal circulation (E2). In the intestine, host and microbial enzymes induce metabolic alterations to the drug that
together with direct binding to bacterial products and segregation control intestinal absorption43. In the liver,
following phase I and phase II processing (first pass metabolism; E3), approximately 90% of the oral drug is
metabolized and destroyed or eliminated through biliary secretion (E4). The drugs secreted into the intestine via the
biliary duct can be reabsorbed via the portal circulation or excreted in stools. As a consequence, only 10% of the oral
drug enters the circulation through the hepatic veins and is available to reach the target tumours and other tissues
(E5). Phase I and phase II processing are also affected by the gut microbiota through the regulation of the level of
host enzymes involved in drug processing. b | Parenteral drug metabolism. Following intravenous administration (P1)
close to 100% of the drug enters the circulation and is available to reach the target tumours (P2); however, the drug
is also distributed systemically, inducing adverse toxic reactions (P3). Any remaining drug not retained in tissues can
be rapidly excreted by the kidney. Each minute 29% of the circulating drug is transported via the splanchnic
circulation (hepatic, mesenteric and splenic arteries) to the liver (P4), where the drug is processed similarly to
enterally administered drugs. The detoxified drugs that are secreted from the liver to the intestine through the
biliary excretion route can be reactivated by bacterial enzymes, inducing intestinal toxicity. CYP450, cytochrome
P450; GI, gastrointestinal.

High-performance liquid chromatography (HPLC) and Platinum-based antineoplastic drugs such as


mass spectrometry analysis showed that contact with oxaliplatin and cisplatin kill tumour cells by inhibiting
the bacteria resulted in biotransformation of the drugs. DNA replication and by targeting cellular membranes
The ability of bacteria to inhibit the antitumour effect of and mitochondria63. They form intra-strand platinum–
gemcitabine and to increase that of the prodrug CB1954 DNA adducts that lead to DNA double-strand breaks
was confirmed in vivo in mice by intratumour inocu- (DSBs). In addition to killing tumour cells, plati-
lation of E. coli 60. In vivo, doxorubicin maintains its num drugs cause severe intestinal toxicity, nephro-
efficacy against subcutaneous tumours in microbiota- toxicity, disruption of blood–brain barrier integrity,
depleted mice; however, in one study, overgrowth oto­toxicity leading to hearing loss and peripheral neu-
of Parabacteroides distasonis in mice treated with ropathy 64–69. Like other chemotherapeutic agents, the
broad-spectrum antibiotics was observed to abrogate genotoxic platinum compounds are toxic for the rap-
its antitumour effect 47. Thus, the results in experimen- idly regenerating intestinal mucosal cells, leading to
tal animals suggest a complex and variable interaction disruption of barrier functions70,71. Barrier breaching
between several chemotherapeutic agents and micro- enables commensal microorganisms and pathogens
biota. However, detailed mechanistic studies in vivo to access the mesenteric lymph nodes and also the
have been reported only for platinum compounds and blood circulation, leading to septicaemia and systemic
cyclophosphamide (CTX)61,62. inflammation70.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The antitumour effect of oxaliplatin or cisplatin innate immune receptor, formyl peptide receptor 1
treatment on subcutaneous transplantable tumours (FPR1) on dendritic cells, not by microbial products
is dramatically decreased in germ-free mice or in but through interaction with the endogenous ligand
mice in which gut commensals have been depleted by annexin A1 from dying cancer cells, which promotes
broad-spectrum antibiotics62 (FIG. 3). In microbiota- stable contacts between the dying cancer cells and the
depleted mice, oxaliplatin induces a lower level of dendritic cells79 (FIG. 4). Unlike oxaliplatin, cisplatin
inflammatory gene expression in tumours than does not induce immunogenic cell death and in mouse
observed in conventionally raised mice62. The pres- tumour models mediates an initial effective and rapid
ence of gut microbiota is not necessary for the drug antitumour effect with almost complete destruction
to penetrate the tumour and to form platinum–DNA of the tumours; however, this effect is short lived and
adducts. However, oxaliplatin-induced DNA damage tumours grow back within about a week, killing most
in the tumour cells is decreased in microbiota-depleted of the animals62. In microbiota-depleted mice, the early
mice62. Known components of the cytotoxic effects of genotoxic and cytotoxic effects of both platinum com-
platinum drugs that mediate the DNA and other cellu- pounds as well as the oxaliplatin-induced long-term
lar damage are reactive oxygen species (ROS)72,73. The survival are decreased62. These findings suggest that
source of the ROS, required for induction of DNA the microbiota composition modulates both the early
damage and apoptosis in tumour and tissue cells was cytotoxic effect and the adaptive immune response,
expected to be the cancer cells themselves upon inter- possibly by independent mechanisms.
action with the drug 73. However, the absence of gut Similarly to oxaliplatin, the alkylating agent CTX
microbiota in mice was shown to prevent the paracrine also induces immunological cell death80. The anti­
production of ROS by tumour-infiltrating myeloid cells tumour activity of CTX in tumour-bearing mice
via NADPH oxidase 2 (NOX2)62. Consistently, oxalip- induces alteration of the gut mucosa within 1–2 days,
latin is not effective in mice that are genetically defi- with increased mucosal permeability and transloca-
cient for Cybb (which encodes the gp91phox chain of tion of gut bacteria into the mesenteric lymph nodes61.
NOX2) or in mice in which myeloid cells are depleted Furthermore, CTX treatment alters the composition of
by antibody treatment 62. Similarly, re‑association commensals in the small intestine of tumour-bearing
of antibiotic-treated mice with the probiotic bac- mice within a week, similar to what has been observed
terial species Lactobacillus acidophilus restores the in cancer patients61,81. More specifically, this chemo-
cisplatin antitumour effect and recovers some of therapeutic agent selectively reduces the abundance of
the cisplatin-induced inflammatory gene expression several bacterial phyla in the small intestinal mucosa
that is observed in conventionally raised mice74. The while it enhances that of Gram-positive bacteria, such
mechanism by which the intact gut microbiota or as Enterococcus hirae, which translocate into mesenteric
L. acidophilus prime intratumour myeloid cells for lymph nodes61,82. Following CTX-induced immuno­
ROS production in response to platinum drugs is genic tumour cell death, these translocated bacteria
dependent on signalling through myeloid differentia- increase the intratumoural CD8+ T cell/T regulatory
tion primary response 88 (MYD88)-associated innate (Treg) cell ratio and activate pathogenic T helper 17 cells
immune receptors (also known as pattern recognition (pTH17 cells) and memory TH1 cell immune responses,
receptors (PRRs)), although the precise receptors resulting in the induction of an antitumour adaptive
involved remain to be determined62 (FIG. 4). immune response that can be detected in the spleen of
Oxaliplatin induces immunogenic cancer cell death, the treated animals61,82,83. The pTH17 cell responses and
a cell death modality that is preceded by autophagy and the antitumour effect of CTX treatment are reduced in
that exposes cell surface-associated and soluble immuno­ germ-free mice and in mice depleted of Gram-positive
stimulatory signals, thus stimulating an antitumour bacteria by treatment with antibiotics61. Moreover, the
immune response through the activation of antigen- antitumour effect of CTX can be restored in microbiota-
presenting cells 75. The adaptive T  cell-mediated depleted mice by adoptive transfer of pTH17 cells61. In
cytotoxic response against oxaliplatin-treated tumours addition, another bacterial species, the Gram-negative
results in long-lasting tumour regression and cure in Barnesiella intestinihominis accumulates in the colon
most mouse tumour models62,75,76. The activation of and promotes the infiltration of interferon‑γ (IFNγ)-
T cells following immunogenic cell death is dependent producing γδ T cells in cancer lesions of CTX-treated
on an inflammatory response involving the induction of mice82. The immune sensor nucleotide-binding oli-
interleukin‑1β (IL‑1β). This is mediated by the dual acti- gomerization domain-containing 2 (NOD2) limits the
vation of the innate immune receptor Toll-like recep- ability of these microorganisms to translocate and to
Pathogenic T helper 17 cells tor 4 (TLR4) by nuclear high mobility group protein modulate CTX anticancer activity. It is noteworthy that
(pTH17 cells). A CD4+ T cell
B1 (HMGB1), a damage-associated molecular pattern the presence of E. hirae- and B. intestinihominis-specific
subset that simultaneously
expresses markers of TH1 cells (DAMP) released by dying cells, and of the NOD-like TH1 cells correlated with a more favourable prognosis in
(T‑bet transcription factor, receptor, pyrin domain-containing 3 (NLRP3) inflam- patients with advanced lung or ovarian cancer treated
interferon–γ (IFN–γ) and CXC masome77. DAMPs react with germline-encoded PRRs with chemo-immunotherapy 82.
chemokine receptor 3 (CXCR3)) and activate inflammation, innate resistance and adap-
and of TH17 cells (RORγT
transcription factor,
tive immunity 78 (FIG. 4). The ability of another chemo- Drug-induced toxicity. There is little information cur-
interleukin‑17 (IL‑17) and C-C therapeutic agent, anthracycline, to elicit antitumour rently available regarding the ability of probiotics to pre-
chemokine receptor 6 (CCR6)). T cell immunity requires the activation of another vent platinum-compound toxicity. Oral treatment with a

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

L. acidophilus Gram-negative L. johnsonii A. shahii Burkholderiales B. breve


bacteria E. hirae B. thetaiotaomicron B. longum
B. intestinihominis B. fragilis B. adolescentis
Commensals

Intestinal
epithelium

MYD88

Mechanism
of anticancer CTLA4
therapy

ROS
PD1 or
PDL1
DNA damage TNF

T cell-mediated
• Pt-DNA adduct Promotion of Licensing of
Tumours T cell-mediated killing following Haemorrhagic
• Genotoxicity antitumour T cell-mediated
tumour killing immunogenic necrosis
and cell death immunity killing
cell death

Cisplatin or TBI and adoptive CpG-ODN and


Treatments Untreated Cyclophosphamide Anti-CTLA4 Anti-PDL1
oxaliplatin T cell transfer anti-IL-10R

Apoptotic cell Bacteria Blood vessels CTL Dendritic Fibroblast


cell

Macrophage Monocyte Neutrophil pTH17 or TH1 cell Tumour cells TLR4 TLR9

Nature Reviews | Cancer


probiotic combination of lyophilized live L. acidophilus ROS production by tissue myeloid cells85. Thus, the gut
and Bifidobacterium bifidum was shown to prevent intes- microbiota may regulate both the antitumour effect
tinal toxicity in cancer patients treated with both radio­ and the toxicity of platinum compounds by similarly
therapy and cisplatin84. Combined with experimental modulating tissue and tumour ROS production62,85.
data that L. acidophilus supports the anticancer effect of The intestinal chemotoxicity of methotrexate is medi-
cisplatin, these clinical data suggest the possibility that ated in part by activation of TLR4 by either microbial
probiotic bacterial species such as L. acidophilus may products or endogenous DAMPs86,87 (FIG. 4). Activation
favour the antitumour effect while preventing some of of TLR2 protects the mucosa against methotrexate-
the toxic side effects of the drug 74,84. Inhibition of NOX induced damage by increasing the expression of the ABC
proteins by acetovanillone protects mice from cisplatin transporter multidrug resistance protein 1 (MDR1; also
nephrotoxicity by blocking early ROS production in known as P‑glycoprotein and ABCB1), which regulates
drug-damaged proximal tubular cells and subsequent the efflux of xenobiotics from intestinal epithelial cells86–88.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

◀ Figure 3 | The gut microbiota regulates anticancer therapies. In physiological pharmacokinetics in these patients103,104. Although the
conditions, the commensals in the intestinal lumen are prevented from translocating mechanism underlying cachexia in these conditions
through the intestinal mucosa by an intact epithelial barrier covered by a mucus layer remains poorly understood, the close relationship between
that is poorly permeable to microorganisms. Treatment with platinum (Pt) drugs, total the gut microbiota and energy metabolism raises the pos-
body irradiation (TBI), cyclophosphamide (CTX) and anti-cytotoxic T lymphocyte-
sibility that the composition of the microbiota could be
associated antigen 4 (CTLA4) all cause damage to the mucus layer, which disrupts
barrier integrity and enables bacteria to penetrate the lamina propria, which lies
involved in the pathogenesis of this condition and there-
beneath the epithelium. Translocated bacteria activate innate immune cells and fore, could be targeted for therapy98,105,106. Use of probiotics
initiate local and systemic inflammation. Mechanisms of gut-associated toxicity and alone or together with a nutritional supplement-enriched
tumour clearance vary based on treatment type, and the microbial species that have diet was shown to improve body weight in patients and
been demonstrated to affect these mechanisms are listed at the top of the figure. mice with cancer-associated cachexia107,108. Recent stud-
Gut commensals, through myeloid differentiation primary response 88 (MYD88)- ies in mice have shown that the colonization of the gut
associated receptors, prime myeloid cells for the generation of reactive oxygen by the E. coli strain O21:H+ prevents muscle wasting
species (ROS), which, in the presence of Pt–DNA adducts formed in response to induced by infection or intestinal damage by engaging
cisplatin or oxaliplatin, cause DNA damage62,74. TBI used to condition patients before the NLR family CARD domain-containing protein 4
they receive adoptive T cell therapy induces mucosal damage and translocation of
(NLRC4) inflammasome and the insulin-like growth fac-
commensals, which through Toll-like receptor 4 (TLR4) signalling activate dendritic
cells to sustain proliferation and cytotoxic functions of the transferred T cells153.
tor 1 (IGF1) signalling pathway109. Additional mechanistic
CTX induces immunological cell death of tumour cells, which elicits the generation studies and rigorous clinical trials are needed to estab-
of antitumour pathogenic T helper 17 (pTH17) cells, TH1 cells and cytotoxic lish whether modulation of gut microbiota homeo­stasis
T lymphocytes (CTLs); CTX also induces damage to the mucosa and translocation could become an effective clinical approach to treat cancer
of commensal bacteria that activate tumour antigen-presenting dendritic cells, co‑morbidities such as anorexia and cachexia as well as
enhancing the antitumour immune response61,82. During CpG-oligodeoxynucleotide cancer therapy adverse events.
(ODN)–anti-interleukin‑10 receptor (IL‑10R) therapy, the gut microbiota, through
TLR4 signalling, primes tumour-infiltrating myeloid cells to respond to the TLR9 Radiotherapy and microbiota
agonist CpG-ODN, producing tumour necrosis factor (TNF) and other inflammatory Many cancer patients receive ionizing radiation therapy
cytokines that induce haemorrhagic necrosis of the tumour and an antitumour
(RTX) that is genotoxic for tumour cells and may be cura-
immune response62. Anti‑CTLA4 immunotherapy promotes both antitumour and
anti-commensal immunity; the anti-commensal immunity against specific genera,
tive for localized cancers. The classic paradigm in radi-
such as Burkholderiales and Bacteroidales (Bacteroides thetaiotaomicron and ation biology assumed that the cellular nucleus was the
Bacteroides fragilis), results in mucosal damage and bacterial translocation but only target of radiation and DNA damage was induced by
also serves as an adjuvant for the antitumour response33. The efficacy of anti- direct deposition of energy or production of ROS through
programmed cell death protein 1 ligand 1 (PDL1) therapy in generating antitumour radiation-induced dissociation of intracellular water mol-
immunity by preventing programmed cell death protein 1 (PD1) interaction with ecules. However, ionizing radiation also induces non-
PDL1 is enhanced by the presence in the gut microbiota of Bifidobacterium spp. targeted effects on non-irradiated cells such as the
(Bifidobacterium breve, Bifidobacterium longum and Bifidobacteri‑um adolescentis)152. bystander effect on nearby cells; systemic radio-adaptive
A. shahii, Alistipes shahii; B. intestinihominis, Barnesiella intestinihominis; E. hirae, responses including inflammatory and immune reactiv-
Enterococcus hirae; L. acidophilus, Lactobacillus acidophilus; L. johnsonii, Lactobacillus
ity; and genomic instability 110,111. Bystander and systemic
johnsonii.
effects are secondary to DNA damage and are mediated
by the disruption of gap junction proteins involved in
Doxorubicin induces severe side effects such as cardio­ cell–cell interactions112 and by the release of extracellular
myopathy and intestinal mucositis associated with mediators, including ROS, nitric oxide (NO), cytokines
major changes in both the oral and the intestinal and exosomes78,113–116. Thus, radiation, similarly to the
microbiota89–91. Although these results suggest that the tissue damage associated with infection by pathogens,
alterations in microbiota composition contribute to tox- induces the release of DAMP stress signals.
icity, the innate immune receptors responsible for the
microbiota-induced toxicity have not been fully char- Response to RTX. Relatively little is known about
acterized89–91. However, NOD2 stimulation by bacterial whether and how the microbiota regulates the response
Cachexia
A wasting syndrome with muramyl dipeptide has a protective effect and prevents to RTX. Local irradiation can induce immunogenic
muscle atrophy and loss of mucosal damage by doxorubicin92 (FIG. 4). tumour cell death and promote systemic inflammation
weight and adipose tissue, The gut microbiota regulates pancreatic beta-cell mass, and immunity 80,117. However, the effects of radiation
often associated with cancer adipose tissue inflammation and uptake of lipids. It ulti- are complex, activating both immunostimulating and
and cancer therapy.
mately promotes diet-induced obesity through the bile immunosuppressive responses and may be insufficient
Bystander effect acid nuclear receptor farnesoid X receptor (FXR) path- to activate a protective anticancer immune response80,117.
In radiobiology, collateral way 93,94. Fat metabolism and adipose tissue are affected Ionizing radiation exerts antitumour responses outside
damage exhibited by in many patients with tumours as well as in platinum the field of radiation — known as the abscopal effect
unirradiated cells in response
drug-treated individuals, resulting in cachexia95–98. Not — that are immune mediated and require the activa-
to signals received from nearby
irradiated cells. only can cancer therapy aggravate the devastating effects tion of antigen-presenting dendritic cells and immune
of cancer-associated cachexia99,100, but also many chemo- T cells118. As the gut microbiota has been shown to affect
Abscopal effect therapeutic agents, including cisplatin, can directly induce the immune response induced by immunogenic cell
In radiotherapy, a multi-organ failure and muscle wasting that resembles the death in both chemotherapy and immunotherapy (dis-
phenomenon whereby local
radiotherapy induces tumour
properties of cancer cachexia100–102. In addition, the over- cussed below)61,62,119, it can be hypothesized that the gut
regression at sites distant from all toxicity of cancer treatment is augmented in patients microbiota also fulfils a role in the immunostimulatory
the irradiated site. with cachexia in part as a consequence of altered drug effects of RTX.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

TLR4 TLR4 TLR2 TLR1 or FPR1 or


? ? TLR6 FPR2

NLRP3
TRAM MAL
+ Gα
MYD88 TRIF Gγ

Oxaliplatin or Methotrexate Oxaliplatin or CTX Radiation Mucosal homeostasis


cisplatin • Hyperexpression • Induction of IL-1β • Migration of COX2+ mesenchymal
Priming of tumour- • Exacerbated mucosal injury • Immunogenic stem cells to the crypt base Ligand: bacterial and
infiltrating cell death • Mucosal protection cellular formyl peptides
myeloid cells for Ligand: bacterial LPS • NOX1-dependent ROS production and other DAMPs
NOX2-dependent inducing the NRF2 system
ROS production Ligand: cellular
CpG-ODN HMGB1 Anthracycline
Priming of tumour infiltrating Ligand: MAMPs from L. rhamnosus Induction of T cell
Ligand: MAMP myeloid cells for TNF production immunity
from L. acidophilus
Ligand: LPS (A. shaii) Methotrexate Ligand: endogenous
Increased detoxification by MDR1 annexin-A1
TBI and adoptive T cell transfer expressed on dying
Intestinal mucosal bacterial Ligand: MAMPs from L. reuteri cancer cells
translocation
Ligand: LPS NOD2
Radiation
Induction of caspase
Endosome 1-mediated intestinal and
haematopoietic cell death
Oxaliplatin or
TLR3 TLR3 Double-strand doxorubicin
Radiation DNA breaks • Protection against
Induction of intestinal damage
massive cell • Bacterial translocation
death AIM2
Ligand: bacterial
Leakage of muramyl dipeptide
cellular RNA Nucleus

Figure 4 | Microbiota-triggered innate immune receptors. Microbial radiation86–88,130–132. Radiation activates TLR3 and the inflammasome
Nature Reviews | Cancer
products (microorganism-associated molecular patterns; MAMPs) and absent in melanoma 2 (AIM2) by inducing leakage of cellular RNA and
endogenous ligands, often released following tissue damage double-strand DNA breaks, respectively, resulting in massive cell death
(damage-associated molecular patterns; DAMPs) interact with and tissue damage 127–129 . The cytoplasmic nucleotide-binding
membrane-bound and cytoplasmic innate immune receptors regulating oligomerization domain-containing 2 (NOD2) receptor recognizing
nutrition, metabolism, tissue homeostasis, inflammation, innate and bacterial muramyl dipeptides participates in the regulation of intestinal
adaptive immunity and, to a lesser extent, morphogenesis213–217. The gut mucosa homeostasis and protects against chemotherapy-induced
microbiota promotes platinum cancer therapy by signalling through mucosal damage and bacterial translocation82,92. The N‑formyl peptide
myeloid differentiation primary response 88 (MYD88), an adaptor for receptors (FPRs) recognize bacterial peptides as well as endogenous
both the Toll-like receptor (TLR) and the interleukin‑1 receptor (IL‑1R) ligands 218. FPR2 expressed on apical and lateral membranes of the
families61,62. ? denotes that the identity of the receptors are yet to be colonic crypt has a crucial role in regulating intestinal homeostasis and
determined. Activation of TLR4 by MAMPs primes tumour myeloid cells inflammation219. The ability of anthracycline to elicit antitumour T cell
to respond to the TLR9 agonist CpG-oligodeoxynucleotide (ODN), immunity requires the interaction of FPR1 on dendritic cells with the
induces mucosal bacterial translocation following total body irradiation endogenous ligand annexin‑A1, which promotes stable contacts
(TBI), which favours optimal anticancer activity of adoptive T cell transfer between dying cancer cells and dendritic cells79. A. shahii, Alistipes shahii;
and mediates mucosal toxicity by methotrexate62,86,87. In response to COX2, cyclooxygenase 2; HMGB1, high mobility group protein B1;
immunogenic cell death mediated by oxaliplatin or cyclophosphamide L. acidophilus, Lactobacillus acidophilus; L. reuteri, Lactobacillus reuteri;
(CTX), activation of TLR4 by DAMPs in combination with NOD-like L. rhamnosus, Lactobacillus rhamnosus; LPS, lipopolysaccharide; MDR1,
receptor, pyrin domain-containing 3 (NLRP3) inflammasome activation multi-drug resistance protein 1; NOX, NADPH oxidase; NRF2, nuclear
induces antitumour T cell activation77. Activation of TLR2 by MAMPs factor erythroid 2‑related factor 2; ROS, reactive oxygen species;
protects against mucosal damage induced by chemotherapy or TNF, tumour necrosis factor.

RTX toxicity. The biggest limitations to the effectiveness radiation exposure122. The observed changes in micro-
and safety of RTX in cancer therapy remain the hetero- biota composition at epithelial surfaces in patients and
geneity in radiation sensitivity of different cancer types mice treated with RTX have been proposed to contrib-
as well as local and systemic toxicity and stress responses ute to the pathogenesis of oral mucositis, diarrhoea,
that cause significant morbidity and may also impair anti- enteritis, colitis and bone marrow failure123–125. RTX
tumour immunity120,121. RTX is associated with damage to induces apoptosis in the intestinal crypts, breach of
healthy tissues and, in particular, actively proliferating tis- the intestinal barrier and alterations in the microbiota
sues such as bone marrow and epithelia (for example, the composition122. These alterations enable pathobionts to
skin and the digestive tract mucosa) are highly affected by gain access to the intestinal immune system, leading

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

to inflammation in the gut as well as changes to the containing L. acidophilus, B. bifidum, Lactobacillus casei
microbiota-mediated homeostatic control of mucosal and particularly the VSL#3 formulation contain-
and systemic innate and adaptive immunity 11,123,126 ing Bifidobacterium, Lactobacillus and Streptococcus
(FIG. 1). spp. have been found to be protective against pelvic
The severity of RTX-induced oral mucositis and radiation-induced gut toxicity, significantly reducing
enteropathy may impose a limitation on therapy the incidence of severe diarrhoea and delaying the
completion. The TLR3 for double-stranded RNA necessity for rescue treatment with loperamide123,133.
(dsRNA) regulates irradiation-mediated intestinal Administration of Lactobacillus brevis CD2 lozenges
toxicity. Tlr3−/− mice are sensitive to p53‑dependent during radiation and chemotherapy treatment of
radiation-induced apoptosis but are protected against patients with head and neck cancer also decreased the
TLR3‑dependent massive cell death following radiation- incidence of therapy-induced mucositis and increased
induced leakage of cellular RNA 127,128. Tlr3 −/− mice the treatment completion rate134.
survive longer when exposed to ionizing radiation Total body irradiation (TBI) is part of the preparative
and show less intestinal toxicity than wild-type mice, regimen for haematopoietic stem cell (or bone marrow)
suggesting that blocking TLR3 signalling may reduce transplantation (BOX 2) and for adoptive T cell transfer
radiation-mediated gastrointestinal damage127,128 (FIG. 4). immunotherapy. Although probiotic treatment amelio-
Radiation-induced DNA DSBs also activate the DNA rates local RTX-induced toxicity, germ-free mice survive
sensor absent in melanoma 2 (AIM2) inflammasome, longer than conventionally raised mice after TBI and
resulting in cell death and tissue damage129 (FIG. 4). In require a higher radiation dose to induce enteropathy
contrast, TLR2‑activating microorganisms such as and 50% mortality 135. After irradiation, germ-free mice
the probiotic Lactobacillus rhamnosus GG have been have fewer apoptotic endothelial cells in the intestinal
shown in mice to protect the intestinal mucosa against mucosa and less lymphocyte infiltration than conven-
chemotherapy- or radiotherapy-induced toxicity by tionally raised mice135. The resistance of germ-free mice
relocating cyclooxygenase 2 (COX2)-expressing cells to the enteric toxicity of TBI could be due to the absence
from the villi to the base of the intestinal crypts130 of gut commensals and pathobionts that in conven-
and inducing ROS, which activate the cytoprotective tionally raised animals may contribute to the mucosal
NRF2 system131,132 (FIG. 4). Indeed, probiotics have been inflammation and damage that follow the initial insult
proved in some clinical studies to be beneficial in pre- mediated by RTX. However, one of the major mecha-
venting radiation-induced enteropathy. Preparations nisms responsible for the resistance of germ-free mice to
TBI is the production of angiopoietin-like 4 (ANGPTL4;
also known as FIAF), a protein inhibitor of lipoprotein
Box 2 | Role of intestinal microbiota in haematopoietic stem cell transplantation lipase that is suppressed by the presence of the micro-
Allogeneic haematopoietic stem cell transplantation (allo-HSCT) and bone marrow biota in conventionally raised mice135. ANGPTL4 has
transplantation are routine procedures for the treatment of haematopoietic pleiotropic effects on lipid metabolism, angiogenesis and
neoplasias198. Before infusing haematopoietic cells, a conditioning regimen using tissue repair 136. Similarly to conventionally raised mice,
high-dose chemoradiotherapy or chemotherapy alone is administered to cancer Angptl4 genetically deficient germ-free mice are suscep-
patients in order to eliminate the malignant cells followed by the graft to restore tible to intestinal damage. The transcription of Angptl4 is
haematopoiesis. Although allo-HSCT represents a curative procedure for some regulated by the PPAR family in response to bacteria that
patients, frequent complications can arise, such as graft-versus-host disease (GVHD), induce the production of small chain fatty acids from
systemic infections and bacteraemia (the presence of bacteria in the blood)199. indigestible carbohydrates137–139. Conversely, the bacte-
Following allo-HSCT, changes in gut microbiota composition and reduced diversity
ria that induce Angptl4 expression include those such as
were observed in both mice and patients199–202, which are probably due to
the Bifidobacterium, Lactobacillus and Streptococcus spp.
therapy-associated mucosal damage, inflammation and immune response against
the commensal bacteria, as well as the use of antibiotics. Depending on the type that mediate protection against RTX-induced mucosi-
of antibiotic used during the treatment, the risk of dominance in the gut of tis and colitis in patients, providing an explanation for
Enterococcus, Streptococcus and various proteobacteria can increase over time after the paradoxical observation that both germ-free mice
allo-HSCT203. Dominance of Enterococcus could evolve into vancomycin-resistant and mice treated with probiotic bacteria are resistant to
Enterococcus bacteraemia whereas dominance of proteobacteria could evolve into radiotherapy toxicity.
Gram-negative bacteraemia203. Mortality is significantly higher in patients with lower Mice are more susceptible to irradiation toxicity
diversity in the gut microbiota after HSCT and the diversity of the microbiota at during daytime than at night-time140–142. The circadian
engraftment is an independent predictor of mortality in allo-HSCT200. In addition, loss rhythm affects radiation-induced apoptosis and activa-
of the genus Blautia as a consequence of the use of antibiotics with specificity for
tion of p53‑responsive genes in the intestine, bone mar-
anaerobic bacteria was associated with increased GVHD and reduced survival204.
row and peripheral blood140–142. As the circadian rhythm
Thus, the use of antibiotics, although required to prevent infections during the
engraftment phase, might affect treatment-related toxicities and favour domination is associated both with diurnal variation of gut microbial
by pathogens and pathobionts. It has been proposed that reconstitution of the composition and short chain fatty acid production, and
microbiota lost during allo-HSCT through transplantation after haematopoietic with cyclic expression in the gut of microbiota-detecting
engraftment with autologous faecal preparations collected and stored before the innate immune receptors resulting in oscillation in the
initiation of conditioning, or restoration of specific bacteria, may improve the clinical number of circulating inflammatory cells that may mod-
outcome200. Mouse data showing that administration of Lactobacillus spp. reduces the ulate radiation sensitivity 143–147, it is possible to hypoth-
severity of GVHD201 suggest that the restoration of specific bacterial species through esize that the observed changes in radiation sensitivity
clinical use of probiotic preparations could be effective in ameliorating HSCT-related may be linked to the circadian variation in microbiota
toxicity.
composition.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The data presently available suggest that the micro­ CpG-oligodeoxynucleotide intratumour therapy.
biota composition modulates the response and repair fol- Intratumoural treatment with the TLR9 agonist
lowing irradiation-induced damage. However, the ability CpG-oligodeoxynucleotide (CpG-ODN) induces an
of the microbiota to license the genotoxicity of chemo- antitumour effect that is potentiated when the immuno­
therapeutic agents by priming for ROS production and suppressive effect of the cytokine IL‑10, produced
inflammatory responses62 suggests that the microbiota by tumour-infiltrating Treg cells and myeloid cells, is
may also control the direct nuclear effect of ionizing prevented by using IL‑10 receptor (IL‑10R) antibod-
radiation and/or its bystander effect. Recently, it has ies155–157. CpG-ODNs act by inducing the secretion from
been reported that conventionally raised mice exposed myeloid cells of pro-inflammatory cytokines such as
to high-energy protons are more susceptible than mice tumour necrosis factor (TNF) and IL‑12 that provoke
with a restricted gut microbiota to single-strand and per- a rapid haemorrhagic necrosis and redirect tumour-
sistent dsDNA damage in peripheral blood leukocytes148. infiltrating macrophages and dendritic cells from an anti-
A better understanding of the mechanism of radiation inflammatory to a pro-inflammatory state. In this
non-targeted effects and their regulation by the commen- pro-inflammatory microenvironment, an antigen-
sal microbiota or its therapeutic manipulation will be specific adaptive T cell antitumour immunity is elicited
invaluable in increasing therapeutic effectiveness, reduc- that can clear tumours in most conventionally raised
ing collateral toxicity of radiotherapy and managing mice155. In contrast, in germ-free mice or mice treated orally
the health effects of accidental exposure to radiation. with a cocktail of non-absorbable antibiotics (vanco­mycin,
neomycin and imipenem), treatment of sub­cutaneous
Immunotherapy and microbiota tumours with CpG-ODNs and anti‑IL‑10R is largely
In most patients treated with traditional cancer ther- inefficient and the tumours progress62. In microbiota-
apies, tumours become resistant to therapy and the depleted mice, tumour-infiltrating myeloid cells fail
chances of tumour recurrence are high149. Recently, to produce pro-inflammatory cytokines in response to
immunotherapy approaches have shown potential in CpG-ODNs and neither the TNF-dependent haemor-
treating haematopoietic and solid cancers150 and have for rhagic necrosis nor the antitumour adaptive immunity
the first time achieved long-lasting complete responses are efficiently induced by the treatment62.
in a proportion of patients with metastatic melanoma In tumours of microbiota-depleted mice, the total
and lung cancer who were unresponsive to conventional number of infiltrating inflammatory monocytes before
therapies. However, the efficacy of immunotherapy is treatment is unaltered while the number of monocyte-
still limited by the variability of the immune response derived Ly6C+ major histocompatibility complex (MHC)
in different patients and the different susceptibility of class II+ cells is reduced62. These data suggest impaired
tumour types151. The emerging knowledge of the abil- differentiation of bone marrow-derived infiltrating
ity of the gut microbiota to modulate the response to inflammatory monocytes into cells with the charac-
immuno­therapy offers new possibilities to improve its teristics and functions of macrophages and dendritic
efficacy by targeting the microbiota33,62,152. cells in the tumour microenvironment of microbiota-
depleted mice. However, only after CpG-ODN treatment
Adoptive T cell transfer. The first report of the ability are major differences in gene expression, particularly in
of the gut microbiota to sustain the anticancer effect of genes encoding inflammatory products and markers, for
immune therapy was the observation that the efficacy example, TNF and IL‑12, observed in tumour-infiltrating
of adoptive transfer of tumour-specific cytotoxic T cells myeloid cell subsets between microbiota-depleted and
following TBI was reduced in mice treated with anti- conventionally raised mice. A partial reduction in
biotics153. The proliferation of the transferred T cells in the response of the myeloid cell to CpG-ODNs is also
the tumour and their antitumour cytotoxic activity is observed in TLR4‑deficient mice, and oral administra-
enhanced by the TBI-induced translocation of the gut tion of the TLR4 agonist LPS to microbiota-depleted
microbiota into the mesenteric lymph nodes153. This mice reconstitutes the responsiveness of the myeloid
effect of the translocated microbiota requires TLR4 cells62 (FIG. 4). These results suggest that the gut micro­
signalling, as TLR4‑deficient mice respond poorly to biota, in part through TLR4 signalling, systemically
the treatment whereas the administration of a TLR4 primes myeloid cells, including tumour-infiltrating cells,
ligand, lipopolysaccharide (LPS), can enhance the anti- for responsiveness to TLR9, a phenomenon that resem-
tumour response induced by adoptive T cell transfer in bles the ‘trained’ innate resistance described with various
non-irradiated lymphodepleted mice153 (FIG. 4). These innate effector cell types158.
observations may help to explain why patients with TNF production in response to CpG-ODNs corre-
metastatic melanoma treated with adoptive cell therapy lates both positively and negatively with the frequency
using tumour-infiltrating lymphocytes respond better of various bacterial genera in the faecal microbiota of
if a conditioning regimen including myelo­ablative mice at the time of the treatment (FIG. 3). For example,
radiotherapy is used154. Similarly, morbidity and sur- the frequencies of the Gram-negative Alistipes and the
vival after haematopoietic stem cell transplantation Gram-positive Ruminococcus genera are positively cor-
preceded by a conditioning regimen using TBI and/or related with TNF production whereas the frequencies of
chemotherapy have been shown to be modulated by the Lactobacillus genus, including Lactobacillus murinum,
the composition of the patient’s gut microbiota after Lactobacillus intestinalis and Lactobacillus fermentum
therapy (BOX 2). are negatively correlated59. After mice were exposed to

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

antibiotics, recolonization of their gut microbiota with In patients treated with anti‑CTLA4, it was simi-
Alistipes shahii restored the ability of myeloid cells to larly observed that an increased frequency of the
produce TNF, whereas oral administration of L. fer­ Bacteroidetes phylum correlated with resistance to
mentum to conventionally raised mice impaired TNF colitis whereas underrepresentation of bacterial spe-
production62. These results indicate that, although cies expressing genetic pathways involved in polyamine
complete depletion of the gut microbiota results in abo- transport and vitamin B biosynthesis was associated
lition of the ‘training’ of myeloid cells for response to with an increased risk of colitis164. Individual antibiotics
CpG-ODNs, different bacterial species can have oppo- change the composition of the gut microbiota, affect-
site effects. Thus, changes in gut microbiota composition ing the anti‑CTLA4 response; for example, vanco­mycin
or alteration in the frequency of individual species that enhances the efficacy of CTLA4 blockade in mice by
can be obtained by therapeutic approaches such as the decreasing the abundance of Gram-positive bacteria
use of antibiotics, prebiotics or probiotics, could be used while preserving the Gram-negative Bacteroidales and
to modulate the response to immunotherapies. Burkholderiales33.
In both mice and patients, anti‑CTLA4 induces a
Immune checkpoint inhibitors. In many cancer patients, TH1 response specific for either B. thetaiotaomicron or
antitumour immunity is dormant or suppressed but can B. fragilis 33. The response to anti‑CTLA4 in microbiota-
be reactivated by releasing the immunological brakes that depleted mice can be restored by adoptive transfer of
tumours use to escape antitumour immunity 150,159–161. In B. fragilis-specific T cells33. These results are reminiscent
particular, the immune checkpoint inhibitors (antibod- of the long-lived microbiota-specific TH1 cell response
ies against cytotoxic T lymphocyte-associated antigen 4 that is generated in acute intestinal infections and which
(CTLA4) expressed on activated T effector cells and primes for a rapid TH1 polarized immune response to
Treg cells and programmed cell death protein 1 (PD1) the pathogens during subsequent infections in the same
or its ligand PD1 ligand 1 (PDL1)) have shown strong anatomical location165. How the microbiota-specific TH1
antitumour activity in experimental animal models and cells induced by anti‑CTLA4 can reach distant tumour
substantial and long-lasting clinical efficacy in patients microenvironments and help to mount a tumour-specific
with cancer (melanoma, renal cell cancer, lung cancer immune response are intriguing questions.
and others being tested in ongoing clinical trials)162. Analysis of the faecal microbiota from patients with
Interpatient variability in therapeutic responsiveness and melanoma before and after treatment with anti‑CTLA4
the failure of certain types of tumour to respond remain showed a change in the relative proportions of three
of great concern154. In addition, immune checkpoint dominant enterotypes observed in these patients33.
inhibitors can induce immune-related adverse effects, Enterotype A was dominated by Prevotella spp.
particularly colitis and inflammation of the pituitary whereas enterotypes B and C were driven by different
gland in response to CTLA4 antibodies, and thyroid Bacteroides spp. The number of patients with enterotype
dysfunction and pneumonitis following blockade of the C increased after treatment at the expense of patients
PD1–PDL1 interaction163. Recently, two studies have with enterotype B. Transfer of the faecal microbiota from
reported the association of the gut microbiota in regu- patients with each of the three human enterotypes into
lating the efficacy of anti‑CTLA4 and anti‑PDL1 cancer tumour-bearing, germ-free mice showed that only the
therapy 33,152. transfer of enterotype C resulted in colonization with
Subcutaneous tumours respond poorly to B. thetaiotaomicron or B. fragilis and enhanced respon-
anti‑CTLA4 in antibiotic-treated or germ-free mice33. siveness to anti‑CTLA4. This observation indicates that
CTLA4 antagonism induces T cell-mediated mucosal anti‑CTLA4 may in some patients alter the composi-
damage in the ileum and colon that is associated with tion of the gut microbiota in a direction that favours its
an alteration in the composition of the intestinal and antitumour activity 33.
faecal microbiota33. In general, the relative frequency Unlike anti‑CTLA4, PDL1 blockade does not induce
in tumour-bearing mice of both Bacteroidales and intestinal damage and does not seem to have an absolute
Burkholderiales (for example, Bacteroides thetaiotao­ requirement for the presence of the gut microbiota152,166.
micron, Bacteroides uniformis and Burkholderia cepa­ However, when subcutaneous B16.SIY melanoma
cia) decreases, whereas that of Clostridiales increases, growth was compared between C57BL/6 mice obtained
upon anti‑CTLA4 treatment. However, the frequency from two different vendors, Jackson (JAX) and Taconic,
Prebiotics
Non-digestible food
of the immunoregulatory Bacteroides fragilis remains it was observed that tumours grew more quickly in
ingredients, often containing constant 33. Oral feeding of either B. thetaiotaomicron or Taconic mice than in JAX mice152. This correlated with
fibre, that promote the growth B. fragilis to microbiota-depleted mice restores the ther- the ability of the mice to induce an immune response
of beneficial microorganisms in apeutic response to anti‑CTLA4 by inducing matura- against the tumour as shown by higher infiltration of
the intestines.
tion of intratumoural dendritic cells and a TH1 response CD8+ T cells in the tumour of JAX mice compared with
Enterotypes detected in the tumour-draining lymph nodes33. The Taconic mice152. Although anti‑PDL1 had an antitumour
Classification of individuals feeding of mice with combined B. fragilis and B. cepacia effect in both Taconic and JAX mice, the slower-growing
based on the composition of also restores the therapeutic response to anti‑CTLA4, tumour of JAX mice was more profoundly affected,
their gut bacterial ecosystem, but unlike single administration with either B. theta­ with almost complete arrest of tumour progression152.
each enterotype having distinct
clusters of organisms with
iotaomicron or B. fragilis, the combined treatment also Taconic mice cohoused with JAX mice acquired the
characteristic predominant significantly decreases the extent of intestinal damage same rate of tumour growth, antitumour resistance and
bacterial species. and colitis associated with the antitumour response33. responsiveness to anti‑PDL1 as observed in JAX mice152.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The response to anti‑PDL1 in C57BL/6 mice trans- A microbiota imbalance that in genetically altered mice
planted with the B16.SIY tumours is significantly influences their phenotype is not limited to bacteria
associated with the relative frequency in the gut but can involve fungi, protozoa, viruses and reactiva-
microbiota of the Bifidobacterium genus, includ- tion of endogenous retroviruses171–176. Thus, preclini-
ing Bifidobacterium breve, Bifidobacterium longum cal ana­lysis of cancer drugs in mice can be profoundly
and Bifidobacterium  adolescentis 152. Oral adminis- affected by the composition of the microbiota in differ-
tration to mice of a commercially available probiotic ent animal populations, necessitating great caution in
cocktail of Bifidobacterium spp. (including B. breve and the interpretation of the results and in their translation
B. longum), alone or with anti‑PDL1, enhanced CD8+ to the clinic.
T cell-induced antitumour activity 152. Furthermore, Identification of the most favourable microbiota
the effect of Bifidobacterium was abolished in CD8+ composition in clinical situations will require a very
T cell-depleted mice, indicating that Bifidobacterium extensive human database with careful analysis of the
action is dependent on cytotoxic T cell activity 152. These correlation of different bacterial species with clini-
results indicate that the mechanism by which anti‑PDL1 cal response. It is possible to confirm the correlative
treatment enhances the antitumour immune response results of human studies by transplanting human faecal
does not require microbiota-specific inflammation and microbial communities into germ-free mice177. These
immune activation, unlike that induced by anti‑CTLA4. humanized mice are stably and heritably colonized and
However, the therapeutic effectiveness of anti‑PDL1 at least in part reproduce the bacterial diversity of the
treatment can be facilitated in tumour-bearing animals donor microbiota177,178. They are susceptible to per-
with a pre-existing antitumour immune response when turbations, such as changes in diet, that are known to
Bifidobacterium spp. are well represented in the gut modify the human microbiota177. Furthermore, even if
microbiota. mice are colonized with the human microbiota, their
physiological and immune responses are similar but not
Concluding remarks identical to those of humans4. For example, the genus
The abundant gut microbiota regulates carcinogenesis Bifidobacterium, which has been shown to induce an
and response to antitumour therapy both in the intes- immune response against tumours and facilitate the
tinal tract and in extra-intestinal tissues, although the therapeutic efficacy of anti‑PDL1 in mice152, can acti-
role of the microbiota at other epithelial barriers remains vate immune cells using two innate immune receptors,
poorly understood11. Many of the mechanisms by which TLR2, which is mostly immunosuppressive, and TLR9,
the gut microbiota affects inflammation, immunity, which is strongly immunostimulating, in the mouse179.
carcinogenesis and response to therapy at the local However, the cellular expression of TLR9 in humans
level have been characterized7–10. However, much less is is very different from its expression in the mouse.
known about the distant effects by which the micro­biota Mouse TLR9 is expressed on all myeloid and dendritic
at the epithelial barriers regulates not only immunity cells whereas in humans its expression is restricted to
and carcinogenesis, but also many of the physiological plasmacytoid dendritic cells and B cells180. Thus, it is
functions of the organism11. not possible yet to conclude that activation of TLR9 in
Most studies in the 3‑year-old scientific field humans by Bifidobacterium spp. has the same immuno­
investigating the modulation of cancer therapy by the stimulating activity as observed in the mouse, and
microbiota have been in mice, and the translation of detailed clinical data are required to determine whether
these experimental findings to the clinic remains a Bifidobacterium-containing probiotics would stimulate
challenge. Experimental mouse tumour models using antitumour activity also in patients.
transplantable cell lines, chemical carcinogenesis or Once the most favourable microbiota composition
genetically modified mice have been instrumental in for each clinical condition has been identified, the next
major advances in immune cancer therapy and in the challenge will be how to modify the patient’s micro­biota.
detailed analysis of the molecular mechanisms involved The resilience and stability of the gut microbiota and
in carcinogenesis and cancer therapy, yet they fail to its responsiveness to physiological, pathological
fully reproduce the features of spontaneous human and environmental changes are characteristics that
tumours167. In addition, the composition of the gut would enable us to use the microbiota composition as a
microbiota varies between mice, even in mice with biomarker, a diagnostic tool and possibly a therapeutic
identical genetic backgrounds and housed under sim- target 24. Although the field of therapeutic intervention
ilar conditions. Identical mouse strains obtained from through targeting the microbiota is still primitive, some
different vendors may also have differences in micro- approaches are already possible. For example, the valida-
biota composition that profoundly affect their physi­ tion of the microbiota as a therapeutic target is provided
ology 152,168. The phenotype of genetically modified by studies showing that patients can be recolonized
mice is not always due to a direct effect of the mutated with a resilient and stable modified microbiota to fight
gene but in some cases is mediated by the presence in the antibiotic-resistant pathogens181. The ultimate goal is to
mutated mouse strains of an altered micro­biota com- discover a bacterial species or a combination of species
position; the altered microbiota is responsible for the that both reduces systemic toxicity and promotes anti-
observed phenotype and can be transmitted by cohousing cancer therapy. Thus, targeting the microbiota in cancer
or faecal transplant to wild-type mice, which acquire the and other diseases is likely to become one of the next
same phenotype as the genetically modified mice169,170. frontiers for precision and personalized medicine.

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

1. Costello, E. K., Stagaman, K., Dethlefsen, L., 24. Faith, J. J. et al. The long-term stability of the human 50. Yip, L. Y. & Chan, E. C. Investigation of host-gut
Bohannan, B. J. & Relman, D. A. The application of gut microbiota. Science 341, 1237439 (2013). microbiota modulation of therapeutic outcome. Drug
ecological theory toward an understanding of the 25. Oh, J., Byrd, A. L., Park, M., Kong, H. H. & Segre, J. A. Metab. Dispos. 43, 1619–1631 (2015).
human microbiome. Science 336, 1255–1262 Temporal stability of the human skin microbiome. Cell 51. Fujita, K. & Sparreboom, A. Pharmacogenetics of
(2012). 165, 854–866 (2016). irinotecan disposition and toxicity: a review. Curr. Clin.
2. Bosch, T. C. & McFall-Ngai, M. J. Metaorganisms as 26. Goodrich, J. K. et al. Human genetics shape the gut Pharmacol. 5, 209–217 (2010).
the new frontier. Zoology (Jena) 114, 185–190 microbiome. Cell 159, 789–799 (2014). 52. Stringer, A. M. et al. Faecal microflora and beta-
(2011). 27. Turnbaugh, P. J. et al. A core gut microbiome in obese glucuronidase expression are altered in an irinotecan-
3. Dzutsev, A., Goldszmid, R. S., Viaud, S., Zitvogel, L. & and lean twins. Nature 457, 480–484 (2009). induced diarrhea model in rats. Cancer Biol. Ther. 7,
Trinchieri, G. The role of the microbiota in 28. David, L. A. et al. Host lifestyle affects human 1919–1925 (2008).
inflammation, carcinogenesis, and cancer therapy. Eur. microbiota on daily timescales. Genome Biol. 15, R89 53. Lin, X. B. et al. Irinotecan (CPT‑11) chemotherapy
J. Immunol. 45, 17–31 (2015). (2014). alters intestinal microbiota in tumour bearing rats.
4. Smith, K., McCoy, K. D. & Macpherson, A. J. Use of 29. David, L. A. et al. Diet rapidly and reproducibly alters PLoS ONE 7, e39764 (2012).
axenic animals in studying the adaptation of mammals the human gut microbiome. Nature 505, 559–563 54. Dabek, M., McCrae, S. I., Stevens, V. J., Duncan, S. H.
to their commensal intestinal microbiota. Semin. (2014). & Louis, P. Distribution of beta-glucosidase and beta-
Immunol. 19, 59–69 (2007). 30. Garrett, W. S. et al. Colitis-associated colorectal cancer glucuronidase activity and of beta-glucuronidase gene
5. Gustafsson, B. E., Daft, F. S., McDaniel, E. G., driven by T‑bet deficiency in dendritic cells. Cancer Cell gus in human colonic bacteria. FEMS Microbiol. Ecol.
Smith, J. C. & Fitzgerald, R. J. Effects of 16, 208–219 (2009). 66, 487–495 (2008).
vitamin K‑active compounds and intestinal 31. Couturier-Maillard, A. et al. NOD2‑mediated dysbiosis 55. McIntosh, F. M. et al. Phylogenetic distribution of
microorganisms in vitamin K‑deficient germfree rats. predisposes mice to transmissible colitis and genes encoding beta-glucuronidase activity in human
J. Nutr. 78, 461–468 (1962). colorectal cancer. J. Clin. Invest. 123, 700–711 colonic bacteria and the impact of diet on faecal
6. Gordon, H. A., Bruckner-Kardoss, E. & (2013). glycosidase activities. Environ. Microbiol. 14,
Wostmann, B. S. Aging in germ-free mice: life tables 32. Hu, B. et al. Microbiota-induced activation of epithelial 1876–1887 (2012).
and lesions observed at natural death. J. Gerontol. 21, IL‑6 signaling links inflammasome-driven inflammation 56. Takasuna, K. et al. Involvement of beta-glucuronidase
380–387 (1966). with transmissible cancer. Proc. Natl Acad. Sci. USA in intestinal microflora in the intestinal toxicity of the
7. De Santis, S., Cavalcanti, E., Mastronardi, M., 110, 9862–9867 (2013). antitumor camptothecin derivative irinotecan
Jirillo, E. & Chieppa, M. Nutritional keys for intestinal References 31 and 32 demonstrated that the hydrochloride (CPT‑11) in rats. Cancer Res. 56,
barrier modulation. Front. Immunol. 6, 612 (2015). carcinogenic phenotypes associated with dysbiosis of 3752–3757 (1996).
8. Vaishnava, S., Behrendt, C. L., Ismail, A. S., the microbiota in genetically mutated mice can be 57. Wallace, B. D. et al. Alleviating cancer drug toxicity by
Eckmann, L. & Hooper, L. V. Paneth cells directly sense transmitted to wild-type mice by microbiota transfer. inhibiting a bacterial enzyme. Science 330, 831–835
gut commensals and maintain homeostasis at the 33. Vetizou, M. et al. Anticancer immunotherapy by (2010).
intestinal host–microbial interface. Proc. Natl Acad. CTLA‑4 blockade relies on the gut microbiota. Science 58. Mego, M. et al. Prevention of irinotecan induced
Sci. USA 105, 20858 (2008). 350, 1079–1084 (2015). diarrhea by probiotics: a randomized double blind,
This paper identified the role of microbiota This paper presented the first demonstration of placebo controlled pilot study. Complement. Ther.
signalling in maintaining the host–commensal the role of the microbiota in modulating Med. 23, 356–362 (2015).
homeostasis through MYD88‑coupled receptors in responsiveness to anti‑CTLA4 therapy. 59. Wallace, B. D. et al. Structure and inhibition of
epithelial cells. 34. DeVita, V. T. Jr & Chu, E. A history of cancer microbiome beta-glucuronidases essential to the
9. Peterson, L. W. & Artis, D. Intestinal epithelial cells: chemotherapy. Cancer Res. 68, 8643–8653 alleviation of cancer drug toxicity. Chem. Biol. 22,
regulators of barrier function and immune homeostasis. (2008). 1238–1249 (2015).
Nat. Rev. Immunol. 14, 141–153 (2014). 35. Sancho-Martinez, S. M., Prieto-Garcia, L., Prieto, M., 60. Lehouritis, P. et al. Local bacteria affect the efficacy
10. Belkaid, Y. & Naik, S. Compartmentalized and Lopez-Novoa, J. M. & Lopez-Hernandez, F. J. of chemotherapeutic drugs. Sci. Rep. 5, 14554
systemic control of tissue immunity by commensals. Subcellular targets of cisplatin cytotoxicity: an (2015).
Nat. Immunol. 14, 646–653 (2013). integrated view. Pharmacol. Ther. 136, 35–55 61. Viaud, S. et al. The intestinal microbiota modulates
This review discusses the role of the microbiota at (2012). the anticancer immune effects of cyclophosphamide.
different epithelial barriers in regulating immunity 36. Mitchell, E. P. Gastrointestinal toxicity of Science 342, 971–976 (2013).
both locally and systemically. chemotherapeutic agents. Semin. Oncol. 33, This paper describes the role of the microbiota in
11. Belkaid, Y. & Hand, T. W. Role of the microbiota in 106–120 (2006). modulating the anticancer effect of CTX.
immunity and inflammation. Cell 157, 121–141 (2014). 37. Spanogiannopoulos, P., Bess, E. N., Carmody, R. N. & 62. Iida, N. et al. Commensal bacteria control cancer
12. Sender, R., Fuchs, S. & Milo, R. Revised estimates for Turnbaugh, P. J. The microbial pharmacists within us: response to therapy by modulating the tumor
the number of human and bacteria cells in the body. a metagenomic view of xenobiotic metabolism. Nat. microenvironment. Science 342, 967–970 (2013).
PLoS Biol. 14, e1002533 (2016). Rev. Microbiol. 14, 273–287 (2016). This paper describes the role of the microbiota in
13. Chow, J., Tang, H. & Mazmanian, S. K. Pathobionts of 38. Li, H. & Jia, W. Cometabolism of microbes and host: modulating the efficacy of cancer therapy with
the gastrointestinal microbiota and inflammatory implications for drug metabolism and drug-induced CpG-ODNs and platinum drugs.
disease. Curr. Opin. Immunol. 23, 473–480 (2011). toxicity. Clin. Pharmacol. Ther. 94, 574–581 (2013). 63. Galluzzi, L. et al. Systems biology of cisplatin
14. Erny, D. et al. Host microbiota constantly control 39. Feng, R. et al. Transforming berberine into its resistance: past, present and future. Cell Death Dis. 5,
maturation and function of microglia in the CNS. Nat. intestine-absorbable form by the gut microbiota. Sci. e1257 (2014).
Neurosci. 18, 965–977 (2015). Rep. 5, 12155 (2015). 64. Roy, S., Ryals, M. M., Van den Bruele, A. B.,
15. Khosravi, A. et al. Gut microbiota promote 40. Maurice, C. F., Haiser, H. J. & Turnbaugh, P. J. Fitzgerald, T. S. & Cunningham, L. L. Sound
hematopoiesis to control bacterial infection. Cell Host Xenobiotics shape the physiology and gene expression preconditioning therapy inhibits ototoxic hearing loss
Microbe 15, 374–381 (2014). of the active human gut microbiome. Cell 152, 39–50 in mice. J. Clin. Invest. 123, 4945–4949 (2013).
16. Trompette, A. et al. Gut microbiota metabolism of (2013). 65. Abuzeid, W. M. et al. Molecular disruption of RAD50
dietary fiber influences allergic airway disease and 41. Montassier, E. et al. Chemotherapy-driven dysbiosis in sensitizes human tumor cells to cisplatin-based
hematopoiesis. Nat. Med. 20, 159–166 (2014). the intestinal microbiome. Aliment. Pharmacol. Ther. chemotherapy. J. Clin. Invest. 119, 1974–1985
17. Chung, W. S. F. et al. Modulation of the human gut 42, 515–528 (2015). (2009).
microbiota by dietary fibres occurs at the species level. 42. Wilson, I. D. & Nicholson, J. K. Gut microbiome 66. Pabla, N. & Dong, Z. Cisplatin nephrotoxicity:
BMC Biol. 14, 3 (2016). interactions with drug metabolism, efficacy, and mechanisms and renoprotective strategies. Kidney Int.
18. Dinan, T. G. & Cryan, J. F. Regulation of the stress toxicity. Transl Res. 179, 204–222 (2017). 73, 994–1007 (2008).
response by the gut microbiota: implications for 43. Haiser, H. J. & Turnbaugh, P. J. Developing a 67. Wagner, J. M. & Karnitz, L. M. Cisplatin-induced
psychoneuroendocrinology. metagenomic view of xenobiotic metabolism. DNA damage activates replication checkpoint
Psychoneuroendocrinology 37, 1369–1378 (2012). Pharmacol. Res. 69, 21–31 (2013). signaling components that differentially affect tumor
19. Sommer, F. & Bäckhed, F. The gut microbiota engages 44. Carmody, R. N. & Turnbaugh, P. J. Host–microbial cell survival. Mol. Pharmacol. 76, 208–214 (2009).
different signaling pathways to induce Duox2 interactions in the metabolism of therapeutic and diet- 68. Zhu, S., Pabla, N., Tang, C., He, L. & Dong, Z. DNA
expression in the ileum and colon epithelium. Mucosal derived xenobiotics. J. Clin. Invest. 124, 4173–4181 damage response in cisplatin-induced nephrotoxicity.
Immunol. 8, 372–379 (2015). (2014). Arch. Toxicol. 89, 2197–2205 (2015).
20. Wells, J. M., Rossi, O., Meijerink, M. & van Baarlen, P. 45. Bjorkholm, B. et al. Intestinal microbiota regulate 69. Park, S. B. et al. Chemotherapy-induced peripheral
Epithelial crosstalk at the microbiota–mucosal xenobiotic metabolism in the liver. PLoS ONE 4, neurotoxicity: a critical analysis. CA Cancer J. Clin. 63,
interface. Proc. Natl Acad. Sci. USA 108 (Suppl. 1), e6958 (2009). 419–437 (2013).
4607–4614 (2011). 46. Selwyn, F. P., Cheng, S. L., Klaassen, C. D. & Cui, J. Y. 70. Hooper, L. V. & Macpherson, A. J. Immune
21. Tulstrup, M. V.‑L. et al. Antibiotic treatment affects Regulation of hepatic drug-metabolizing enzymes in adaptations that maintain homeostasis with the
intestinal permeability and gut microbial composition germ-free mice by conventionalization and probiotics. intestinal microbiota. Nat. Rev. Immunol. 10,
in Wistar rats dependent on antibiotic class. PLoS Drug Metab. Dispos. 44, 262–274 (2016). 159–169 (2010).
ONE 10, e0144854 (2015). 47. Selwyn, F. P., Cui, J. Y. & Klaassen, C. D. RNA-Seq 71. Sonis, S. T. The pathobiology of mucositis. Nat. Rev.
22. Backhed, F. et al. Dynamics and stabilization of the quantification of hepatic drug processing genes in Cancer 4, 277–284 (2004).
human gut microbiome during the first year of life. Cell germ-free mice. Drug Metab. Dispos. 43, 1572–1580 72. Kim, S., Lee, T. J., Park, J. W. & Kwon, T. K.
Host Microbe 17, 690–703 (2015). (2015). Overexpression of cFLIPs inhibits oxaliplatin-mediated
23. Dominguez-Bello, M. G. et al. Delivery mode shapes 48. Selwyn, F. P. et al. Developmental regulation of drug- apoptosis through enhanced XIAP stability and Akt
the acquisition and structure of the initial microbiota processing genes in livers of germ-free mice. Toxicol. activation in human renal cancer cells. J. Cell.
across multiple body habitats in newborns. Proc. Natl Sci. 147, 84–103 (2015). Biochem. 105, 971–979 (2008).
Acad. Sci. USA 107, 11971–11975 (2010). 49. Kang, M. J. et al. The effect of gut microbiota on drug 73. Laurent, A. et al. Controlling tumor growth by
References 22 and 23 describe the establishment metabolism. Expert Opin. Drug Metab. Toxicol. 9, modulating endogenous production of reactive
of the human microbiota during early life. 1295–1308 (2013). oxygen species. Cancer Res. 65, 948–956 (2005).

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

74. Gui, Q. F., Lu, H. F., Zhang, C. X., Xu, Z. R. & 97. Suárez-Zamorano, N. et al. Microbiota depletion potential clinical applications. Aliment. Pharmacol.
Yang, Y. H. Well-balanced commensal microbiota promotes browning of white adipose tissue and Ther. 40, 409–421 (2014).
contributes to anti-cancer response in a lung cancer reduces obesity. Nat. Med. 21, 1497–1501 (2015). 124. Vanhoecke, B. W. et al. Low-dose irradiation affects the
mouse model. Genet. Mol. Res. 14, 5642–5651 98. de Matos-Neto, E. M. et al. Systemic inflammation in functional behavior of oral microbiota in the context of
(2015). cachexia — is tumor cytokine expression profile the mucositis. Exp. Biol. Med. (Maywood) 241, 60–70
75. Tesniere, A. et al. Immunogenic death of colon cancer culprit? Front. Immunol. 6, 629 (2015). (2016).
cells treated with oxaliplatin. Oncogene 29, 482–491 99. Antoun, S., Baracos, V. E., Birdsell, L., Escudier, B. & 125. Broin, P. Ó. et al. Intestinal microbiota-derived
(2010). Sawyer, M. B. Low body mass index and sarcopenia metabolomic blood plasma markers for prior radiation
76. Michaud, M. et al. Subversion of the chemotherapy- associated with dose-limiting toxicity of sorafenib in injury. Int. J. Radiat. Oncol. Biol. Phys. 91, 360–367
induced anticancer immune response by the ecto- patients with renal cell carcinoma. Ann. Oncol. 21, (2015).
ATPase CD39. Oncoimmunology 1, 393–395 (2012). 1594–1598 (2010). 126. Wang, A. et al. Gut microbial dysbiosis may predict
77. Ghiringhelli, F. et al. Activation of the NLRP3 100. Toledo, M. et al. A multifactorial anti-cachectic diarrhea and fatigue in patients undergoing pelvic
inflammasome in dendritic cells induces approach for cancer cachexia in a rat model cancer radiotherapy: a pilot study. PLoS ONE 10,
IL‑1beta‑dependent adaptive immunity against undergoing chemotherapy. J. Cachexia Sarcopenia e0126312 (2015).
tumors. Nat. Med. 15, 1170–1178 (2009). Muscle 7, 48–59 (2016). 127. Takemura, N. et al. Blockade of TLR3 protects mice
78. Pateras, I. S. et al. The DNA damage response and 101. Conte, E. et al. Cisplatin-induced cachexia in rats from lethal radiation-induced gastrointestinal
immune signaling alliance: is it good or bad? Nature causes alterations in skeletal muscle calcium syndrome. Nat. Commun. 5, 3492 (2014).
decides when and where. Pharmacol. Ther. 154, homeostasis. Biophys. J. 108 (Suppl. 1), 108a (2015). 128. Vacchelli, E. et al. Trial Watch: Toll-like receptor
36–56 (2015). 102. Garcia, J. M., Cata, J. P., Dougherty, P. M. & agonists for cancer therapy. Oncoimmunology 2,
79. Vacchelli, E. et al. Chemotherapy-induced antitumor Smith, R. G. Ghrelin prevents cisplatin-induced e25238 (2013).
immunity requires formyl peptide receptor 1. Science mechanical hyperalgesia and cachexia. Endocrinology 129. Hu, B. et al. The DNA-sensing AIM2 inflammasome
350, 972–978 (2015). 149, 455–460 (2008). controls radiation-induced cell death and tissue injury.
80. Kroemer, G., Galluzzi, L., Kepp, O. & Zitvogel, L. 103. Bruggeman, A. R. et al. Cancer cachexia: beyond Science 354, 765–768 (2016).
Immunogenic cell death in cancer therapy. Annu. Rev. weight loss. J. Oncol. Pract. 12, 1163–1171 (2016). 130. Ciorba, M. A. et al. Lactobacillus probiotic protects
Immunol. 31, 51–72 (2013). 104. Cvan Trobec, K. et al. Influence of cancer cachexia on intestinal epithelium from radiation injury in a TLR‑2/
This paper reviewed the mechanisms underlying drug liver metabolism and renal elimination in rats. cyclo-oxygenase‑2‑dependent manner. Gut 61,
the phenomenon of immunogenic cell death in J. Cachexia Sarcopenia Muscle 6, 45–52 (2015). 829–838 (2012).
cancer therapy. 105. Bindels, L. B. & Delzenne, N. M. Muscle wasting: the 131. Jones, R. M. et al. Lactobacilli modulate epithelial
81. Zwielehner, J. et al. Changes in human fecal gut microbiota as a new therapeutic target? Int. cytoprotection through the Nrf2 pathway. Cell Rep.
microbiota due to chemotherapy analyzed by TaqMan- J. Biochem. Cell Biol. 45, 2186–2190 (2013). 12, 1217–1225 (2015).
PCR, 454 sequencing and PCR-DGGE fingerprinting. 106. Klein, G. L., Petschow, B. W., Shaw, A. L. & Weaver, E. 132. Jones, R. M. et al. Symbiotic lactobacilli stimulate gut
PLoS ONE 6, e28654 (2011). Gut barrier dysfunction and microbial translocation in epithelial proliferation via Nox-mediated generation of
82. Daillere, R. et al. Enterococcus hirae and Barnesiella cancer cachexia: a new therapeutic target. Curr. Opin. reactive oxygen species. EMBO J. 32, 3017–3028
intestinihominis facilitate cyclophosphamide-induced Support. Palliat. Care 7, 361–367 (2013). (2013).
therapeutic immunomodulatory effects. Immunity 45, 107. Yeh, K. Y. et al. Omega‑3 fatty acid-, micronutrient-, 133. Delia, P. et al. Use of probiotics for prevention of
931–943 (2016). and probiotic-enriched nutrition helps body weight radiation-induced diarrhea. World J. Gastroenterol.
This paper characterized the role of different stabilization in head and neck cancer cachexia. Oral 13, 912–915 (2007).
bacterial species in regulating antitumour T cell Surg. Oral Med. Oral Pathol. Oral Radiol. 116, 41–48 134. Sharma, A. et al. Lactobacillus brevis CD2 lozenges
responses induced by CTX. (2013). reduce radiation- and chemotherapy-induced
83. Ghoreschi, K. et al. Generation of pathogenic TH17 108. Varian, B. J. et al. Beneficial bacteria inhibit cachexia. mucositis in patients with head and neck cancer: a
cells in the absence of TGF-beta signalling. Nature Oncotarget 7, 11803–11816 (2016). randomized double-blind placebo-controlled study.
467, 967–971 (2010). 109. Schieber, A. M. P. et al. Disease tolerance mediated by Eur. J. Cancer 48, 875–881 (2012).
84. Chitapanarux, I. et al. Randomized controlled trial of microbiome E. coli involves inflammasome and IGF‑1 135. Crawford, P. A. & Gordon, J. I. Microbial regulation of
live Lactobacillus acidophilus plus Bifidobacterium signaling. Science 350, 558–563 (2015). intestinal radiosensitivity. Proc. Natl Acad. Sci. USA
bifidum in prophylaxis of diarrhea during radiotherapy 110. Mavragani, I. V. et al. Key mechanisms involved in 102, 13254–13259 (2005).
in cervical cancer patients. Radiat. Oncol. 5, 31 ionizing radiation-induced systemic effects. A current This paper describes the radioresistance of
(2010). review. Toxicol. Res. 5, 12–33 (2016). germ-free mice and characterized the underlying
85. Wang, Y. et al. Pharmacological inhibition of NADPH 111. Azzam, E. I. & Little, J. B. The radiation-induced molecular mechanisms.
oxidase protects against cisplatin induced bystander effect: evidence and significance. Hum. Exp. 136. Santulli, G. Angiopoietin-like proteins: a
nephrotoxicity in mice by two step mechanism. Food Toxicol. 23, 61–65 (2004). comprehensive look. Front. Endocrinol. 5, 4
Chem. Toxicol. 83, 251–260 (2015). 112. Vacchelli, E. et al. Trial Watch: anticancer (2014).
86. Cario, E. Toll-like receptors in the pathogenesis of radioimmunotherapy. Oncoimmunology 2, e25595 137. Grootaert, C. et al. Bacterial monocultures,
chemotherapy-induced gastrointestinal toxicity. Curr. (2013). propionate, butyrate and H2O2 modulate the
Opin. Support. Palliat. Care 10, 157–164 (2016). 113. Apetoh, L. et al. Toll-like receptor 4‑dependent expression, secretion and structure of the fasting-
87. Frank, M. et al. TLR signaling modulates side effects of contribution of the immune system to anticancer induced adipose factor in gut epithelial cell lines.
anticancer therapy in the small intestine. J. Immunol. chemotherapy and radiotherapy. Nat. Med. 13, Environ. Microbiol. 13, 1778–1789 (2011).
194, 1983–1995 (2015). 1050–1059 (2007). 138. Jacouton, E. et al. Lactobacillus rhamnosus
88. Mercado-Lubo, R. & McCormick, B. A. The interaction 114. Nikitaki, Z. et al. Systemic mechanisms and effects of CNCMI‑4317 modulates Fiaf/Angptl4 in intestinal
of gut microbes with host ABC transporters. Gut ionizing radiation: a new ‘old’ paradigm of how the epithelial cells and circulating level in mice. PLoS ONE
Microbes 1, 301–306 (2010). bystanders and distant can become the players. 10, e0138880 (2015).
89. Napenas, J. J. et al. Molecular methodology to assess Semin. Cancer Biol. 37–38, 77–95 (2016). 139. Korecka, A. et al. ANGPTL4 expression induced by
the impact of cancer chemotherapy on the oral 115. Ermolaeva, M. A. et al. DNA damage in germ cells butyrate and rosiglitazone in human intestinal
bacterial flora: a pilot study. Oral Surg. Oral Med. induces an innate immune response that triggers epithelial cells utilizes independent pathways. Am.
Oral Pathol. Oral Radiol. Endod. 109, 554–560 systemic stress resistance. Nature 501, 416–420 J. Physiol. Gastrointest. Liver Physiol. 304,
(2010). (2013). G1025–G1037 (2013).
90. Niu, Q. Y., Li, Z. Y., Du, G. H. & Qin, X. M. 1H NMR 116. Al‑Mayah, A. et al. The non-targeted effects of 140. Duncan, A. M., Ronen, A. & Blakey, D. H. Diurnal
based metabolomic profiling revealed doxorubicin- radiation are perpetuated by exosomes. Mutat. Res. variation in the response of gamma-ray-induced
induced systematic alterations in a rat model. 772, 38–45 (2015). apoptosis in the mouse intestinal epithelium. Cancer
J. Pharm. Biomed. Anal. 118, 338–348 (2016). 117. Demaria, S. & Formenti, S. C. Radiation as an Lett. 21, 163–166 (1983).
91. Rigby, R. J. et al. Intestinal bacteria are necessary for immunological adjuvant: current evidence on dose and 141. Ishihara, H. et al. Circadian transitions in radiation
doxorubicin-induced intestinal damage but not for fractionation. Front. Oncol. 2, 153 (2012). dose-dependent augmentation of mRNA levels for
doxorubicin-induced apoptosis. Gut Microbes 7, 118. Demaria, S. et al. Ionizing radiation inhibition of DNA damage-induced genes elicited by accurate real-
414–423 (2016). distant untreated tumors (abscopal effect) is immune time RT‑PCR quantification. J. Radiat. Res. 51,
92. Nigro, G., Rossi, R., Commere, P. H., Jay, P. & mediated. Int. J. Radiat. Oncol. Biol. Phys. 58, 265–275 (2010).
Sansonetti, P. J. The cytosolic bacterial peptidoglycan 862–870 (2004). 142. Ruifrok, A. C., Weil, M. M., Thames, H. D. &
sensor Nod2 affords stem cell protection and links 119. Zitvogel, L., Ayyoub, M., Routy, B. & Kroemer, G. Mason, K. A. Diurnal variations in the expression of
microbes to gut epithelial regeneration. Cell Host Microbiome and anticancer immunosurveillance. Cell radiation-induced apoptosis. Radiat. Res. 149,
Microbe 15, 792–798 (2014). 165, 276–287 (2016). 360–365 (1998).
93. Jiang, C. et al. Intestine-selective farnesoid X receptor 120. Deng, L. et al. Irradiation and anti‑PD‑L1 treatment 143. Leone, V. et al. Effects of diurnal variation of gut
inhibition improves obesity-related metabolic synergistically promote antitumor immunity in mice. microbes and high-fat feeding on host circadian clock
dysfunction. Nat. Commun. 6, 10166 (2015). J. Clin. Invest. 124, 687–695 (2014). function and metabolism. Cell Host Microbe 17,
94. Parseus, A. et al. Microbiota-induced obesity requires 121. Baird, J. R. et al. Radiotherapy combined with novel 681–689 (2015).
farnesoid X receptor. Gut 66, 429–437 (2016). STING-targeting oligonucleotides results in regression 144. Liang, X., Bushman, F. D. & FitzGerald, G. A.
95. Das, S. K. et al. Adipose triglyceride lipase contributes of established tumors. Cancer Res. 76, 50–61 (2016). Rhythmicity of the intestinal microbiota is regulated by
to cancer-associated cachexia. Science 333, 122. Barker, H. E., Paget, J. T. E., Khan, A. A. & gender and the host circadian clock. Proc. Natl Acad.
233–238 (2011). Harrington, K. J. The tumour microenvironment after Sci. USA 112, 10479–10484 (2015).
96. Ruud, J. et al. Inflammation- and tumor-induced radiotherapy: mechanisms of resistance and 145. Mukherji, A., Kobiita, A., Ye, T. & Chambon, P.
anorexia and weight loss require MyD88 in recurrence. Nat. Rev. Cancer 15, 409–425 (2015). Homeostasis in intestinal epithelium is
hematopoietic/myeloid cells but not in brain 123. Touchefeu, Y. et al. Systematic review: the role of the orchestrated by the circadian clock and microbiota
endothelial or neural cells. FASEB J. 27, 1973–1980 gut microbiota in chemotherapy- or radiation-induced cues transduced by TLRs. Cell 153, 812–827
(2013). gastrointestinal mucositis — current evidence and (2013).

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

146. Nguyen, K. D. et al. Circadian gene Bmal1 regulates 171. Wheeler, M. L. et al. Immunological consequences of 197. Westbrook, A. M. et al. The role of tumour necrosis
diurnal oscillations of Ly6Chi inflammatory monocytes. intestinal fungal dysbiosis. Cell Host Microbe 19, factor-alpha and tumour necrosis factor receptor
Science 341, 1483–1488 (2013). 865–873 (2016). signalling in inflammation-associated systemic
147. Thaiss, C. A. et al. Transkingdom control of microbiota 172. Howitt, M. R. et al. Tuft cells, taste-chemosensory genotoxicity. Mutagenesis 27, 77–86 (2012).
diurnal oscillations promotes metabolic homeostasis. cells, orchestrate parasite type 2 immunity in the gut. 198. Gyurkocza, B., Rezvani, A. & Storb, R. F. Allogeneic
Cell 159, 514–529 (2014). Science 351, 1329–1333 (2016). hematopoietic cell transplantation: the state of the
148. Maier, I., Berry, D. M. & Schiestl, R. H. Intestinal 173. Kernbauer, E., Ding, Y. & Cadwell, K. An enteric virus art. Expert Rev. Hematol. 3, 285–299 (2010).
microbiota reduces genotoxic endpoints induced by can replace the beneficial function of commensal 199. Taur, Y., Jenq, R. R., Ubeda, C., van den Brink, M. &
high-energy protons. Radiat. Res. 181, 45–53 bacteria. Nature 516, 94–98 (2014). Pamer, E. G. Role of intestinal microbiota in
(2014). 174. Cadwell, K. The virome in host health and disease. transplantation outcomes. Best Pract. Res. Clin.
149. Holohan, C., Van Schaeybroeck, S., Longley, D. B. & Immunity 42, 805–813 (2015). Haematol. 28, 155–161 (2015).
Johnston, P. G. Cancer drug resistance: an evolving 175. Ramanan, D. et al. Helminth infection promotes 200. Taur, Y. et al. The effects of intestinal tract bacterial
paradigm. Nat. Rev. Cancer 13, 714–726 (2013). colonization resistance via type 2 immunity. Science diversity on mortality following allogeneic
150. Couzin-Frankel, J. Breakthrough of the year 2013. 352, 608–612 (2016). hematopoietic stem cell transplantation. Blood 124,
Cancer immunotherapy. Science 342, 1432–1433 176. Young, G. R. et al. Resurrection of endogenous 1174–1182 (2014).
(2013). retroviruses in antibody-deficient mice. Nature 491, 201. Jenq, R. R. et al. Regulation of intestinal
151. Mellman, I., Coukos, G. & Dranoff, G. Cancer 774–778 (2012). inflammation by microbiota following allogeneic bone
immunotherapy comes of age. Nature 480, 480–489 177. Turnbaugh, P. J. et al. The effect of diet on the human marrow transplantation. J. Exp. Med. 209, 903–911
(2011). gut microbiome: a metagenomic analysis in (2012).
152. Sivan, A. et al. Commensal Bifidobacterium promotes humanized gnotobiotic mice. Sci. Transl Med. 1, 202. Holler, E. et al. Metagenomic analysis of the stool
antitumor immunity and facilitates anti‑PD‑L1 efficacy. 6ra14 (2009). microbiome in patients receiving allogeneic stem
Science 350, 1084–1089 (2015). 178. Baxter, N. T., Zackular, J. P., Chen, G. Y. & cell transplantation: loss of diversity is associated
This paper demonstrated that the presence of Schloss, P. D. Structure of the gut microbiome with use of systemic antibiotics and more
Bifidobacterium spp. in the gut microbiota following colonization with human feces determines pronounced in gastrointestinal graft-versus-host
promotes antitumour immunity in mice that is colonic tumor burden. Microbiome 2, 20 (2014). disease. Biol. Blood Marrow Transplant. 20,
amplified by anti‑PDL1 therapy. 179. Plantinga, T. S. et al. Differential Toll-like receptor 640–645 (2014).
153. Paulos, C. M. et al. Microbial translocation augments recognition and induction of cytokine profile by 203. Taur, Y. et al. Intestinal domination and the risk of
the function of adoptively transferred self/tumor- Bifidobacterium breve and Lactobacillus strains of bacteremia in patients undergoing allogeneic
specific CD8+ T cells via TLR4 signaling. J. Clin. Invest. probiotics. Clin. Vaccine Immunol. 18, 621–628 hematopoietic stem cell transplantation. Clin. Infect.
117, 2197–2204 (2007). (2011). Dis. 55, 905–914 (2012).
This paper used a therapy model of adoptive T cell 180. Kadowaki, N. et al. Subsets of human dendritic cell 204. Jenq, R. R. et al. Intestinal blautia is associated with
transfer preceded by TBI in mice, to demonstrate precursors express different toll-like receptors and reduced death from graft-versus-host disease. Biol.
for the first time that the microbiota modulates respond to different microbial antigens. J. Exp. Med. Blood Marrow Transplant. 21, 1373–1383 (2015).
anticancer therapy. 194, 863–869 (2001). 205. Ho, J. T. K., Chan, G. C. F. & Li, J. C. B. Systemic
154. Dudley, M. E. et al. Adoptive cell therapy for patients 181. Pamer, E. G. Resurrecting the intestinal microbiota to effects of gut microbiota and its relationship with
with metastatic melanoma: evaluation of intensive combat antibiotic-resistant pathogens. Science 352, disease and modulation. BMC Immunol. 16, 21
myeloablative chemoradiation preparative regimens. 535–538 (2016). (2015).
J. Clin. Oncol. 26, 5233–5239 (2008). 182. Goldszmid, R. S. & Trinchieri, G. The price of immunity. 206. Boleij, A. et al. The Bacteroides fragilis toxin gene is
155. Guiducci, C., Vicari, A. P., Sangaletti, S., Trinchieri, G. Nat. Immunol. 13, 932–938 (2012). prevalent in the colon mucosa of colorectal cancer
& Colombo, M. P. Redirecting in vivo elicited tumor 183. Jobin, C. Colorectal cancer: CRC — all about microbial patients. Clin. Infect. Dis. 60, 208–215 (2015).
infiltrating macrophages and dendritic cells towards products and barrier function? Nat. Rev. 207. Koshiol, J. et al. Salmonella enterica serovar Typhi and
tumor rejection. Cancer Res. 65, 3437–3446 (2005). Gastroenterol. Hepatol. 9, 694–696 (2012). gallbladder cancer: a case-control study and meta-
156. Vicari, A. P. et al. Reversal of tumor-induced dendritic 184. Rao, V. P. et al. Innate immune inflammatory response analysis. Cancer Med. 5, 3310–3235 (2016).
cell paralysis by CpG immunostimulatory against enteric bacteria Helicobacter hepaticus 208. Lecuit, M. et al. Immunoproliferative small intestinal
oligonucleotide and anti-interleukin 10 receptor induces mammary adenocarcinoma in mice. Cancer disease associated with Campylobacter jejuni. N. Engl.
antibody. J. Exp. Med. 196, 541–549 (2002). Res. 66, 7395–7400 (2006). J. Med. 350, 239–248 (2004).
157. Stewart, C. A. et al. Interferon-dependent IL‑10 185. Salcedo, R. et al. MyD88‑mediated signaling prevents 209. Senff, N. J. et al. European Organization for Research
production by Tregs limits tumor Th17 inflammation. development of adenocarcinomas of the colon: role of and Treatment of Cancer and International Society for
J. Clin. Invest. 123, 4859–4874 (2013). interleukin 18. J. Exp. Med. 207, 1625–1636 Cutaneous Lymphoma consensus recommendations
158. Netea, M. G. et al. Trained immunity: a program of (2010). for the management of cutaneous B‑cell lymphomas.
innate immune memory in health and disease. Science 186. Allen, I. C. et al. The NLRP3 inflammasome functions Blood 112, 1600–1609 (2008).
352, aaf1098 (2016). as a negative regulator of tumorigenesis during colitis- 210. Ferreri, A. J. et al. Chlamydophila psittaci eradication
159. Rosenberg, S. A., Yang, J. C. & Restifo, N. P. Cancer associated cancer. J. Exp. Med. 207, 1045–1056 with doxycycline as first-line targeted therapy for
immunotherapy: moving beyond current vaccines. (2010). ocular adnexae lymphoma: final results of an
Nat. Med. 10, 909–915 (2004). 187. Sears, C. L. & Garrett, W. S. Microbes, microbiota, and international phase II trial. J. Clin. Oncol. 30,
160. Eggermont, A. M. Therapeutic vaccines in solid colon cancer. Cell Host Microbe 15, 317–328 (2014). 2988–2994 (2012).
tumours: can they be harmful? Eur. J. Cancer 45, 188. Kostic, A. D. et al. Fusobacterium nucleatum 211. Lakritz, J. R. et al. Gut bacteria require neutrophils to
2087–2090 (2009). potentiates intestinal tumorigenesis and modulates promote mammary tumorigenesis. Oncotarget 6,
161. Page, D. B., Postow, M. A., Callahan, M. K., the tumor-immune microenvironment. Cell Host 9387–9396 (2015).
Allison, J. P. & Wolchok, J. D. Immune modulation in Microbe 14, 207–215 (2013). 212. Rutkowski, M. R. et al. Microbially driven
cancer with antibodies. Annu. Rev. Med. 65, 189. Rubinstein, M. R. et al. Fusobacterium nucleatum TLR5‑dependent signaling governs distal malignant
185–202 (2014). promotes colorectal carcinogenesis by modulating progression through tumor-promoting inflammation.
162. Sharma, P. & Allison, J. P. The future of immune E‑cadherin/beta-catenin signaling via its FadA adhesin. Cancer Cell 27, 27–40 (2015).
checkpoint therapy. Science 348, 56–61 (2015). Cell Host Microbe 14, 195–206 (2013). 213. Chu, H. & Mazmanian, S. K. Innate immune
163. Teply, B. A. & Lipson, E. J. Identification and 190. Gur, C. et al. Binding of the Fap2 protein of recognition of the microbiota promotes host–
management of toxicities from immune checkpoint- Fusobacterium nucleatum to human inhibitory microbial symbiosis. Nat. Immunol. 14, 668–675
blocking drugs. Oncology (Williston Park) 28 (Suppl. receptor TIGIT protects tumors from immune cell (2013).
3), 30–38 (2014). attack. Immunity 42, 344–355 (2015). 214. McFall-Ngai, M. et al. Animals in a bacterial world, a
164. Dubin, K. et al. Intestinal microbiome analyses identify 191. IARC Working Group on the Evaluation of Carcinogenic new imperative for the life sciences. Proc. Natl Acad.
melanoma patients at risk for checkpoint-blockade- Risks to Humans. Schistosomes, liver flukes and Sci. USA 110, 3229–3236 (2013).
induced colitis. Nat. Commun. 7, 10391 (2016). Helicobacter pylori. Lyon, 7–14 June 1994. IARC 215. Kremer, N. et al. Initial symbiont contact orchestrates
165. Hand, T. W. et al. Acute gastrointestinal infection Monogr. Eval. Carcinog. Risks Hum. 61, 1–241 host-organ-wide transcriptional changes that prime
induces long-lived microbiota-specific T cell responses. (1994). tissue colonization. Cell Host Microbe 14, 183–194
Science 337, 1553–1556 (2012). 192. Poutahidis, T. et al. Pathogenic intestinal bacteria (2013).
166. Yang, X. et al. Targeting the tumor microenvironment enhance prostate cancer development via systemic 216. Hansen, C. H. et al. Patterns of early gut colonization
with interferon‑β bridges innate and adaptive immune activation of immune cells in mice. PLoS ONE 8, shape future immune responses of the host. PLoS
responses. Cancer Cell 25, 37–48 (2014). e73933 (2013). ONE 7, e34043 (2012).
167. Zitvogel, L., Pitt, J. M., Daillere, R., Smyth, M. J. & 193. Fox, J. G. et al. Gut microbes define liver cancer risk in 217. Stappenbeck, T. S., Hooper, L. V. & Gordon, J. I.
Kroemer, G. Mouse models in oncoimmunology. Nat. mice exposed to chemical and viral transgenic Developmental regulation of intestinal angiogenesis
Rev. Cancer 16, 759–773 (2016). hepatocarcinogens. Gut 59, 88–97 (2010). by indigenous microbes via Paneth cells. Proc. Natl
168. Ivanov, I. I., Frutos Rde, L., Manel, N., Yoshinaga, K. & 194. Yamamoto, M. L. et al. Intestinal bacteria modify Acad. Sci. USA 99, 15451–15455 (2002).
Rifkin, D. B. Specific microbiota direct the lymphoma incidence and latency by affecting systemic 218. Le, Y. et al. Biologically active peptides interacting with
differentiation of IL‑17‑producing T‑helper cells in the inflammatory state, oxidative stress, and leukocyte the G protein-coupled formylpeptide receptor. Protein
mucosa of the small intestine. Cell Host Microbe 4, genotoxicity. Cancer Res. 73, 4222–4232 (2013). Pept. Lett. 14, 846–853 (2007).
337 (2008). 195. Farrell, J. J. et al. Variations of oral microbiota are 219. Chen, K. et al. Formylpeptide receptor‑2 contributes
169. Nowarski, R. et al. Epithelial IL‑18 equilibrium controls associated with pancreatic diseases including to colonic epithelial homeostasis, inflammation, and
barrier function in colitis. Cell 163, 1444–1456 pancreatic cancer. Gut 61, 582–588 (2012). tumorigenesis. J. Clin. Invest. 123, 1694–1704
(2015). 196. Fan, X. et al. Human oral microbiome and prospective (2013).
170. Elinav, E. et al. NLRP6 inflammasome regulates risk for pancreatic cancer: a population-based nested
colonic microbial ecology and risk for colitis. Cell 145, case-control study. Gut http://dx.doi.org/10.1136/ Competing interests statement
745–757 (2011). gutjnl-2016-312580 (2016). The authors declare no competing interests.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 15


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like