You are on page 1of 10

RevIeWS

Methotrexate and its mechanisms


of action in inflammatory arthritis
Bruce N. Cronstein   1 ✉ and Thomas M. Aune   2
Abstract | Despite the introduction of numerous biologic agents for the treatment of rheumatoid
arthritis (RA) and other forms of inflammatory arthritis, low-​dose methotrexate therapy remains
the gold standard in RA therapy. Methotrexate is generally the first-​line drug for the treatment
of RA , psoriatic arthritis and other forms of inflammatory arthritis, and it enhances the effect of
most biologic agents in RA. Understanding the mechanism of action of methotrexate could be
instructive in the appropriate use of the drug and in the design of new regimens for the treatment
of RA. Although methotrexate is one of the first examples of intelligent drug design, multiple
mechanisms potentially contribute to the anti-​inflammatory actions of methotrexate, including
the inhibition of purine and pyrimidine synthesis, transmethylation reactions, translocation of
nuclear factor-​κB (NF-​κB) to the nucleus, signalling via the Janus kinase (JAK)–signal transducer
and activator of transcription (STAT) pathway and nitric oxide production, as well as the promotion
of adenosine release and expression of certain long non-​coding RNAs.

Since its introduction and widespread adoption in its well-​known toxic effects. A better understanding of
the 1980s for the treatment of rheumatoid arthritis the mechanisms of action of methotrexate could lead to
(RA), low-​dose methotrexate therapy has considerably more appropriate treatment regimens. The mechanisms
changed the approach to therapy as well as the outcome discussed in this Review are general and, although these
of therapy for RA1. On seeing a rheumatologist, most mechanisms are best documented in RA, they could
patients with RA are initially given low-​dose metho­ apply to other forms of inflammatory arthritis.
trexate therapy, and many of the patients require no
additional therapies2. Even for patients who have had Pharmacology of methotrexate
an inadequate response to methotrexate and are treated Whether administered orally or parenterally at a low
with a biologic agent, methotrexate therapy is generally dose, methotrexate has a relatively short half-​life of
continued because of the seemingly additive effects of approximately 6 hours and is undetectable in serum
biologic drugs and methotrexate and because metho- after 18 hours6. The bioavailability of orally adminis-
trexate diminishes resistance to many biologic drugs1. tered methotrexate is highly variable owing to the lim-
Moreover, in patients with RA, low-​dose methotrexate ited capacity of the gut to absorb methotrexate, and the
therapy, with or without concomitant biologic therapy, maximal absorption of a single oral dose is <25 mg (ref.7).
is associated with a lower rate of large joint replacement Methotrexate is excreted both as methotrexate and as
than therapy with biologic agents alone, supporting 7-hydroxymethotrexate (the major metabolite of metho­
the hypothesis that low-​dose methotrexate therapy is trexate) in the urine. In the 1980s, methotrexate was first
chondroprotective3–5. reported as a ‘pro-drug’ that accumulates in tissue in a
The use and effectiveness of methotrexate in the polyglutamated form8,9. It is now known that metho­
treat­ment of rheumatic disease will probably be further trexate monoglutamate (the native form of the drug)
enhanced by a better understanding of the pharmacol- undergoes serial polyglutamation within cells, with vary­
1
The New York University ogy and anti-​inflammatory mechanisms of action of this ing numbers of glutamic acid additions, and metho-
School of Medicine, New York, drug. Indeed, emerging data suggest that appropriate trexate polyglutamates can be detected in the tissues for
NY, USA.
biomarkers might be identified that can determine who weeks8 (Fig. 1). Polyglutamated methotrexate is the active
2
Vanderbilt University will benefit most from methotrexate treatment. Some of form of the drug, and the potency of poly­glutamated
Medical Center, Nashville,
TN, USA.
these biomarkers might reflect the mechanism of action methotrexate as an inhibitor of a number of enzymes
✉e-​mail: bruce.cronstein@ of methotrexate. In this Review, we discuss the pharma- differs from the potency of the native compound. The
nyumc.org cology and metabolic and cellular mechanisms by which enzyme inhibited most potently by methotrexate poly­
https://doi.org/10.1038/ methotrexate suppresses inflammation. We also examine glutamates is 5-aminoimidazole-4-carboxamide ribo­
s41584-020-0373-9 the mechanisms by which methotrexate induces some of nucleotide (AICAR) transformylase (ATIC), which

Nature Reviews | Rheumatology


Reviews

antiproliferative drug for the treatment of cancer10. By


Key points
contrast, reductions in peripheral leukocyte counts are
• Methotrexate polyglutamates inhibit aminoimidazole-4-carboxamide ribonucleotide considered a toxic reaction to methotrexate in the treat-
(AICAR) transformylase (ATIC), leading to intracellular accumulation of AICAR ment of inflammatory disease, for which methotrexate is
and increased adenosine release; adenosine binds to cell surface receptors and administered at doses 100-fold to 1,000-fold lower than
suppresses many inflammatory and immune reactions.
the dose administered for malignancies. Administration
• Methotrexate inhibits dihydrofolate reductase, preventing the reduction of of folic acid or even folinic acid (except on the day that
dihydrobiopterin (BH2) to tetrahydrobiopterin (BH4), leading to nitric oxide synthase
methotrexate is administered) can prevent the toxic
uncoupling and increased sensitivity of T cells to apoptosis, thereby diminishing
effects of methotrexate in patients with rheumatic dis-
immune responses.
eases11–14. Although folinic acid supplementation has
• Methotrexate inhibits activation of nuclear factor-​κB (NF-​κB) by increasing both
been reported to lead to flare of RA in patients taking
adenosine release and activation of adenosine receptor A2a and by inhibiting the
reduction of BH2 to BH4. methotrexate15,16, a meta-​analysis does not support this
association in the treatment of RA17. Given that metho­
• Methotrexate increases the expression of long intergenic non-​coding RNA p21
(lincRNA-​p21), which is a multifunction long non-​coding RNA that regulates, both trexate is effective even when concurrent folic acid or
directly and indirectly, a variety of critical immune and inflammatory processes. folinic acid supplementation prevents reductions in
• By modulating cell-​specific signalling pathways, methotrexate inhibits important peripheral leukocyte counts, the anti-​inflammatory
pro-inflammatory properties of major cell lineages involved in rheumatoid arthritis effects of methotrexate are unlikely to require inhibition
pathogenesis, including T cells, macrophages, endothelial cells and fibroblast-like of cellular proliferation.
synoviocytes.
Inhibition of transmethylation reactions. Polyamines,
such as spermine and spermidine, accumulate in syn-
catalyses the last committed step in de novo purine ovial tissue and fluid18, mononuclear cells19 and urine
biosynthesis; methotrexate polyglutamate is >2,000 of patients with RA20. Monocytes can hydrolyse these
times more potent as an inhibitor of ATIC than metho- polyamines to ammonia and H2O2, which function as
trexate monoglutamate9. Methotrexate polyglutamates cytotoxins that can injure the cells and tissues of the
also inhibit other folate-​dependent enzymes involved in joint19,21,22. Inhibition of dihydrofolate reductase (DHFR;
de novo pyrimidine and purine synthesis (Fig. 2a) as well an enzyme that catalyses the reduction of dihydrofolate
as the enzymes involved in transmethylation reactions to tetrahydrofolate) by methotrexate diminishes the
and polyamine synthesis (Fig. 2c). formation of the methyl donors tetrahydrofolate and
5-methyltetrahydrofolate and reduces the synthesis of
Postulated mechanisms of action polyamines (Fig. 2c). Thus, one hypothesis is that inhibi-
Methotrexate was initially developed to inhibit the tion of transmethylation reactions by methotrexate and
de novo synthesis of purines and pyrimidines that is subsequent reduction in polyamine production results
required for DNA and RNA synthesis and the prolifer- in diminished downstream production of ammonia and
ation of many different types of malignant cells, as well H2O2, thereby diminishing synovial injury. This hypoth-
as non-​malignant cells (Box 1). Although this mech- esis was the basis for clinical testing of a direct trans-
anism might contribute to the mechanism by which methylation inhibitor, 3-deazaadenosine, as a therapy
methotrexate suppresses inflammation (Fig. 2a), other for RA; however, although this drug effectively inhib-
mechanisms of action have been postulated, includ- ited transmethylation reactions in the cells of patients
ing enhanced adenosine release (Fig. 2b), inhibition of taking the drug, it had no effect on the course of RA
transmethylation reactions that are required for some (reviewed elsewhere23). This finding suggests that inhib­
cellular functions (Fig. 2c), diminished accumulation of ition of transmethylation reactions has a small part in
polyamines (Fig. 2c) and nitric oxide synthase uncoupling the overall anti-​inflammatory actions of methotrexate
(Fig. 2d). Methotrexate regulates, both directly and indi- therapy in inflammatory disease.
rectly, the function of nearly every cell type involved in
inflammation, including neutrophils, monocytes, T cells, Adenosine release. As noted already, ATIC is potently
B cells, endothelial cells and fibroblast-like synovio- inhibited by methotrexate polyglutamates 9,24, and
cytes (FLSs). In this section, we discuss the previously AICAR accumulates intracellularly in the tissues of
described molecular mechanisms of action of metho- methotrexate-​t reated mice25. AICAR inhibits AMP
trexate and, where relevant, discuss the effects of these deaminase and adenosine deaminase, leading to the
molecular mechanisms on the functions of various cell release of adenine nucleotides into the extracellular
types involved in inflammation. space (Fig. 2b); adenine nucleotides are converted into
adenosine by the action of the cell surface enzymes
Metabolic mechanisms ectonucleoside triphosphate dephosphorylase 1 (also
Inhibition of purine and pyrimidine synthesis. known as CD39) and ecto-5′-nucleotidase (also known
Methotrexate targets critical folate-​dependent enzy- as CD73). Adenosine is a potent stimulus for a family
matic steps in the de novo synthesis of purines and of receptors (adenosine receptors A1a, A2a, A2b and A3),
pyrimidines, the building blocks for RNA and DNA. all of which have potent inhibitory effects on nearly all
Reductions in the number of circulating leukocytes inflammatory cell types (reviewed elsewhere26 and sum-
(neutrophils and lymphocytes), cells that require rapid marized in Box 2). The hypothesis that adenosine, via
proliferation of precursor cells in the bone marrow, these receptors, mediates the anti-​inflammatory effects
typically occur in patients taking methotrexate as an of methotrexate was supported by studies in mice in

www.nature.com/nrrheum
Reviews

a O COOH
O COOH
NH2 N
H
O N N
H N N COOH
N
HN N COOH CH3
H H2N N N
H2N N N
Folic acid Methotrexate
O COOH

NH2 N
H
N
N N COOH
H
H2N N N
Aminopterin

b Glutamyl
residues
O COOH FPGS O COOH

NH2 N NH2 N
H GGH H
N N OH
N N COOH N N
CH3 CH3 O
H2N N N H2N N N

Methotrexate (monoglutamate) Methotrexate polyglutamate

Fig. 1 | molecular structures of folic acid, aminopterin and methotrexate. a | Aminopterin and methotrexate (formerly
known as amethopterin) are structurally highly similar to folic acid. b | Within cells, methotrexate monoglutamate
undergoes serial polyglutamation to form methotrexate polyglutumates (methotrexateGlu(1–7)), the active form of
methotrexate. In this process, additional glutamate residues are added to the terminal glutamate moiety of methotrexate
by the action of the enzyme folylpolyglutamate synthase (FPGS). This reaction is reversible, and the peptidase γ-glutamyl
hydrolase (GGH) mediates the opposite reaction.

which methotrexate increased adenosine release into a non-​selective adenosine receptor antagonist, reduced
inflamed tissue, and the anti-​inflammatory effects of the therapeutic response to methotrexate in patients
methotrexate were reversed by selective adenosine recep­ with RA32. By contrast, in a large retrospective study
tor A2a antagonists25. In a subsequent study in rats with of patients with RA receiving long-​term methotrexate
adjuvant arthritis, non-​selective adenosine receptor therapy, caffeine consumption was not associated with
antagonists (theophylline and caffeine)27 reversed the poor responses to methotrexate; however, the patients
anti-​inflammatory effects of methotrexate. Methotrexate with a poor response to methotrexate early in the course
does not inhibit inflammation in A2a-deficient mice, of disease were probably undercounted in this study33.
A3-deficient mice or CD73-deficient mice, further con- More recently, emerging evidence suggests that adeno-
firming the hypothesis that adenosine release mediates sine production and release is one mechanism by which
the anti-​inflammatory effects of methotrexate in animal regulatory T (Treg) cells diminish cellular immunity and
models28–30. inflammation26 (leading to a growing number of stud-
Demonstrating that adenosine has a role in medi- ies of adenosine receptor antagonists in the treatment
ating the anti-​inflammatory effects of methotrexate in of cancer34). Adenosine is produced by Treg cells via
patients with inflammatory diseases has been more CD39-mediated and CD73-mediated dephosphoryla-
difficult. Measurement of adenosine concentrations tion of ATP, and some data indicate that low levels of
in biological fluids requires special care owing to the CD39 on Treg cells are associated with poor responses to
rapid uptake of adenosine by cells and metabolism of methotrexate in patients with RA35. In a parallel fashion,
adenosine to inosine by adenosine deaminase present methotrexate-​mediated increases in adenosine release
in plasma. One study31 reported that methotrexate treat- by B cells lead to diminished immunization against thera­
ment of patients with RA increases adenosine-​mediated peutic monoclonal antibodies (such as anti-​TNF anti-
and dipyridamole-​mediated vasodilation (adenosine is bodies) in a B-​cell activating factor (BAFF)-dependent
a potent vasodilator and dipyridamole functions as a fashion and have been postulated to provide the basis
vasodilator by blocking adenosine uptake and thereby for methotrexate-​mediated suppression of antidrug anti-
increasing extracellular adenosine levels). This find- bodies in patients receiving combined methotrexate and
ing confirms that methotrexate therapy does indeed anti-​TNF therapy36.
increase adenosine release in humans as the higher
levels of adenosine released in patients treated with Nitric oxide synthase uncoupling. In addition to catalys-
methotrexate would lead to increased adenosine levels ing the reduction of dihydrofolate to tetrahydrofolate,
and adenosine-​mediated increases in vascular flow. In a DHFR also catalyses the reduction of dihydrobiopterin
randomized prospective study, ingestion of caffeine, (BH2) to tetrahydrobiopterin (BH4); methotrexate

Nature Reviews | Rheumatology


Reviews

a Folinic
b
Methotrexate Folic acid Adenosine
acid release
RFC1 FRβ or PCFT Extracellular

Intracellular ATP ADP AMP Adenosine

Methotrexate Folic acid Folinic acid


CD39 CD73
FPGS GGH ATP ADP AMP Adenosine
DHF THF
MethotrexateGlu(1–7) DHFR
AMPDA ADA

5-CH3-THF 5,10-CH2-THF
MTHFR IMP Inosine
AICAR
MethotrexateGlu(1–7)
dTMP
ATIC
TYMS
dUMP
FAICAR ↑ AICAR

ATIC
Decreased purine
and pyrimidine
production FAICAR

d
MethotrexateGlu(1–7) Methotrexate/
methotrexateGlu(1–7)

DHFR
DHF THF

DHFR

Homocysteine BH2 BH4


5-CH3-THF 5,10-CH2-THF
NO NO
MTHFR
synthase synthase

SAH Methionine ROS production NO production

SAM 10-CH3-THF JNK activation

Polyamine synthesis Methyl donor AP1 activity

Fig. 2 | methotrexate regulates essential biochemical reactions. polyamines (owing to diminished concentrations of the methyl donor
a | Methotrexate is taken up by cells via reduced folate carrier 1 (RFC1), 5-methyltetra­hydrofolate (5-CH3-THF)). d | Methotrexate inhibition of
where it is polyglutamated by folylpolyglutamate synthase (FPGS). DHFR inhibits tetrahydrobiopterin (BH4) production, thereby diminish-
Methotrexate polyglutamates accumulate intracellularly and can ing nitric oxide (NO) production and increasing the production of
inhibit a number of enzymatic reactions, including those mediated by reactive oxygen species (ROS) in a process known as ‘NO synthase
dihydro­folate reductase (DHFR), methylenetetrahydrofolate reductase uncoupling’. Increased JUN N-​terminal kinase (JNK) activation promotes
(MTHFR), thymidylate synthase (TYMS) and 5-aminoimidazole-4- activator protein 1 (AP1) activity and inhibits nuclear factor-κB activa-
carboxamide ribonucleotide (AICAR) transformylase (ATIC), leading to tion (not shown). BH2, dihydrobiopterin; DHF, dihydrofolate; dTMP,
diminished production of purines and pyrimidines. b | Methotrexate-​ deoxythymidine monophosphate; dUMP, deoxyuridine monophosphate;
mediated inhibition of ATIC leads to accumulation of intracellular AICAR , FAICAR, formyl AICAR; FRβ, folate receptor-​β; GGH, γ-glutamyl hydro-
which ulti­mately leads to an increase in extracellular adenosine levels lase; IMP, inosine monophosphate; PCFT, proton-​coupled folate trans-
owing to reduced activity of the enzymes AMP deaminase (AMPDA) and porter; SAH, S-adenosylhomocysteine; SAM, S-adenosylmethionine;
adenine deaminase (ADA). c | Methotrexate inhibits the synthesis of THF, tetrahydrofolate.

www.nature.com/nrrheum
Reviews

Long non-​coding RNAs


inhibits the reduction of both dihydrofolate and BH2 at a gene adjacent to CDKN1A (encoding p21), hence its
(lncRNAs). A newly discovered similar concentrations (Fig. 2d). BH4 is a necessary cofac- name; lincRNA-​p21 is induced by p53 as part of the
class of RNAs that are tor for all nitric oxide synthases (a family of enzymes that DNA damage response. LincRNA-​p21 orchestrates
defined as RNAs of more than catalyse the production of nitric oxide from l-​arginine). the p53-mediated apoptotic response by repressing
200 base pairs in length, are
transcribed from genes but
In the absence of BH4, nitric oxide synthases produce transcription of numerous genes encoding proteins that
not translated into proteins reactive oxygen species, such as hydrogen peroxide, inhibit apoptosis. As such, lincRNA-​p21 is required for
due to the presence of multiple rather than nitric oxide, a process termed ‘nitric oxide proper induction of apoptosis but does not seem to affect
translational stop codons synthase uncoupling’37–39. Increased concentrations of regulation of the cell cycle, two of the central pathways
and, as RNAs, regulate the
reactive oxygen species activate JUN N-​terminal kinases regulated by p53 in response to DNA damage or other
expression of target genes and
proteins to carry out a vast
(JNKs), leading to increased phosphorylation of the cellular stress45. LincRNA-​p21 is also induced by hypoxia
array of biologic functions. transcription factor JUN and increased transcriptional and promotes the expression of the transcription factor
activity of the JUN–FOS heterodimer activator pro- HIF1α, an important mediator of the cellular response
Warburg effect tein 1 (AP1). This transcription factor is a key regu­lator to hypoxia that promotes the Warburg effect46. In the
A shift from oxidative
phosphorylation to glycolysis
of apoptosis as well as many other cellular processes. cytoplasm, an additional function of lincRNA-​p21 is to
during cell activation. AP1 activation results in the induction of genes encod- bind to select target mRNAs to inhibit their recruitment
ing proteins that induce cell cycle arrest and promote to the ribosome and translation into proteins47. Thus,
sensitivity to apoptosis, including TP53, CDKN1A, lincRNA-​p21 is a multifunctional lncRNA regulating a
CDKN1B, CHEK2, BCL3 and HRK. HRK encodes a variety of critical biologic processes.
protein, activator of apoptosis harakiri, that promotes Circulating levels of both p53 and lincRNA-​p21 are
apoptosis by interacting with the apoptotic inhibitors decreased in RA and are restored to levels in healthy
BCL-2 and BCL2L1, thus explaining the increase in sen- individuals during methotrexate therapy48. In T cells,
sitivity of T cells to apoptosis in response to methotrexate methotrexate induces the expression of lincRNA-​p21
stimulation40,41. In T cells, methotrexate also inhibits (Fig. 3a). Induction of lincRNA-​p21 by metho­trexate
TNF-stimulated increases in nuclear factor-κB (NF-κB) in T cells is not a result of nitric oxide synthase uncou-
transcriptional activity, a major pro-​inflammatory signal­ pling or adenosine release and adenosine receptor
ling pathway. Methotrexate-​mediated inhibition of NF-​κB activation, but rather depends on activation of the
activation occurs through JNK-​dependent upregulation DNA damage sentinel DNA-​dependent protein kinase
of p53 via the pathway described above42. (DNA-​P K). Exactly how methotrexate activates
DNA-​PK is not understood, but inhibition of DNA-​PK
LincRNA-​p21 expression. Emerging data indicate that clearly abrogates methotrexate-​mediated induction
methotrexate regulates the expression of some long non-​ of lincRNA-​p21 (ref.48). In T cells, lincRNA-​p21 inhib-
coding RNAs (lncRNAs; a general review of lncRNAs is its NF-​κB activity but does not seem to target the
provided in ref.43). One such example, long intergenic genes RELA and NFKB1, encoding key components of
non-​coding RNA p21 (lincRNA-​p21)44, is encoded by NF-​κB, or disrupt intracellular signalling pathways that

Box 1 | Development of methotrexate


Methotrexate (formerly known as amethopterin) and its close analogue aminopterin were among the first products of
intelligent drug design. These drugs were developed by Yellapragada Subbarow, a biochemist at Harvard University,
for the treatment of cancers on the basis of their structural similarity to folic acid (Fig. 1) and their capacity to inhibit
folate-dependent enzymes. These drugs were first used, in high doses, to treat leukaemias and other forms of malignancy
in 1948, and aminopterin, in chemotherapeutic doses, was first used for the treatment of rheumatoid arthritis (RA) in 1951
(ref.100). Aminopterin was ultimately taken off of the market owing to manufacturing difficulties, leaving only methotrexate.
During the 1960s and 1970s, low-​dose methotrexate therapy was increasingly used to treat severe psoriasis, and its use was
extended to RA. In the mid-1980s, methotrexate was successfully tested in patients with RA in rigorous studies101–103 and
subsequently licensed by the FDA for the treatment of patients with RA in 1988 (ref.45). Since its initial approval, the use
of methotrexate has increased, as has the willingness of physicians to use higher, and more effective, doses of the drug.
More importantly, the recognition that oral bioavailability of the drug is quite limited led to the development and approval,
in 2013 and 2014, of two different parenteral (subcutaneous) formulations of the drug for use in the treatment of rheumatic
disease104,105.

Aminopterin and First report of Systematic reports Two different preparations of


amethopterin aminopterin use for of use of low-dose methotrexate for subcutaneous
(methotrexate) the treatment of methotrexate to use in the treatment of RA
synthesized psoriasis and RA treat RA licensed by the FDA

1947 1948 1951 1964 1985 1988 2013 2014

Aminopterin Aminopterin use Methotrexate


introduced for discontinued licensed for the
the treatment because of treatment of RA
of cancer manufacturing by the FDA
difficulties

Nature Reviews | Rheumatology


Reviews

Box 2 | effects of adenosine on inflammatory cells26 treated with methotrexate, suggesting that the effects of
methotrexate on JAK–STAT signalling might dampen
Neutrophils but not completely inhibit this inflammatory signalling
• Inhibits oxidant generation pathway in patients.
• Inhibits adhesion and recruitment
• Inhibits neutrophil extracellular trap formation Inhibition of NF-​κB signalling. Activation and nuclear
translocation of NF-​κB has a central function in inflam-
macrophages
mation and inflammatory changes in many tissues and
• Increases M1 to M2 transformation cell types. As noted already, adenosine has been postu-
• Inhibits cytokine expression lated to mediate many of the anti-​inflammatory actions
• Inhibits osteoclast differentiation of methotrexate (Fig. 2b), including direct inhibition of
T cells
NF-​κB activation in a variety of cell types and tissues, such
as monocytes, macrophages and endothelial cells52–61.
• Inhibits T cell receptor-​triggered activation
In addition, as described earlier, methotrexate can also
• Inhibits activation-​induced cell death
inhibit NF-​κB activation by diminishing the expres­sion
• Inhibits FAS–FASL-mediated cell death of RELA (via the promotion of lincRNA-p21 expression)
• Increases regulatory T (Treg) cell differentiation in T cells and by promoting BH4 depletion and JNK
• Mediates Treg cell-​mediated suppression of T cell activation (Fig. 3a). A whole-​genome association study
proliferation identified a single-​nucleotide polymorphism in TNFAIP3
endothelial cells (encoding A20, a critical suppressor of NF-​κB activation)
• Increases barrier integrity
that was associated with a favourable response to metho­
trexate in patients with inflammatory arthritis62. More
• Inhibits oedema formation
recently, researchers found that macrophages stimulated
Fibroblast-​like synoviocytes with only granulocyte–macrophage colony-​stimulating
• Inhibits metalloproteinase production factor (GM-​CSF), but not other stimuli, responded
directly to methotrexate treatment through a thymidylate
synthase-​mediated and p53-mediated mechanism63,
lead to NF-​κB activation. Rather, lincRNA-p21 binds to and that GM-​CSF-stimulated macrophages upregu-
RELA mRNA to inhibit its translation, thus reducing the lated A20 in response to methotrexate64, rendering them
amount of NF-​κB available to mediate transcriptional more ‘tolerant’. Thus, there are multiple mechanisms by
activation in response to external inflammatory stimuli, which methotrexate therapy suppresses the activation
such as TNF. Consistent with this notion, patients with of NF-​κB and downstream expression of inflammatory
RA receiving methotrexate therapy have substantially mechanisms. Some of these mechanisms seem to be
reduced levels of NF-​κB subunit p65 (encoded by RELA) limited to specific cell types (for example, lincRNA-​p21
compared with patients with RA not receiving metho- and A20 induction in T cells and GM-CSF-stimulated
trexate therapy. In this way, induction of lincRNA-​p21 macrophages, respectively), whereas adenosine affects
might also contribute to the efficacy of methotrexate activation of NF-​κB in multiple cell types.
therapy in RA.
Cellular mechanisms
Inhibition of JAK–STAT signalling. IL-6 and other Effects on T cells. T cells from patients with RA are resis­
stimuli activate a critical signalling system that leads tant to cell cycle checkpoint signals and apoptosis31,32.
to phosphorylation of signal transducer and activator The expression and function of genes that encode pro-
of transcription (STAT) proteins by receptor-​associated teins critical for cell cycle checkpoint programmes and
Janus kinases (JAKs), which stimulates production apoptosis are reduced in RA. The expression levels of
of a variety of inflammatory signals49. On the basis of these genes are restored to normal or healthy control
high-​throughput screening data, methotrexate and its levels in patients with RA receiving methotrexate ther-
analogue, aminopterin, were shown to inhibit signalling apy40,41. Examples of such genes include TP53, CDKN1A,
via the JAK1–STAT3 and JAK2–STAT5 transcriptional CDKN1B and CHEK2. In T cells, these genes are directly
pathways in Drosophila cells and, subsequently, in human induced by methotrexate at submicromolar concentra-
macrophage cell lines50,51. The results suggested that the tions, similar to the concentrations shown to achieve
inhibitory effects were folate independent as folinic a therapeutic benefit for patients with RA, thus supporting
acid did not reverse methotrexate-​mediated inhibition the notion that checkpoint reprogramming by metho­
of STAT5 phosphorylation, and knockdown of various trexate might contribute to its therapeutic benefits.
folate-​dependent enzymes had no effect on the capacity Under these conditions, methotrexate does not directly
of methotrexate to inhibit STAT5 phosphorylation50,51. induce cell cycle arrest or apoptosis in T cells. Rather,
It is unclear whether methotrexate treatment in these methotrexate markedly increases the sensitivity of T cells
cells directly inhibits JAK-​mediated phosphorylation of to apoptosis (Fig. 3a). Thus, the known resistance of RA
STAT3 and STAT5 or whether other intracellular fac- T cells to apoptosis might be reversed by methotrexate
tors are involved as the effect of methotrexate was tested therapy.
only in whole cells. The authors of this report noted that In addition, the pro-​inflammatory transcription
STAT phosphorylation was only moderately inhibited factor NF-​κB is constitutively active in T cells from
in peripheral blood CD4+ T cells, B cells and monocytes patients with RA, and this chronic activity is corrected

www.nature.com/nrrheum
Reviews

in patients with RA receiving methotrexate therapy65 of differing levels of nitric oxide synthases, with T cells
(as discussed earlier) (Fig. 3a). Methotrexate-​mediated having high levels of nitric oxide synthase activity, thus
inhibition of chronically active NF-​κB in RA T cells might enabling the nitric oxide synthase uncoupling pathway to
result from the ability of methotrexate to induce expres- predominate, and FLSs having low levels of nitric oxide
sion of p53, a transcription factor with anti-​inflammatory synthase activity, enabling the adenosine–adenosine
properties66–79, or induction of lincRNA-​p21, both of receptor pathway to predominate. Thus, methotrex-
which reduce or inhibit NF-​κB activity. ate also inhi­bits NF-​κB activity in FLSs, which might
contribute to its therapeutic efficacy in RA.
Effects on fibroblast-​like synoviocytes. FLSs also partici­
pate in RA pathogenesis, and NF-​κB is highly acti­vated Effects on monocytes. The effects of methotrexate have
in these cells in RA74,80. High NF-​κB activity in FLSs is also been examined in monocyte cell lines81. In con-
thought to contribute to their proliferation, angio­genesis trast to T cells and FLSs, monocytes undergo apopto-
and additional inflammatory properties80. Methotrexate sis in response to methotrexate. Further, methotrexate
inhi­bits NF-κB activity in FLSs. However, metho­trexate induces a dose-​dependent increase in expression of
does not inhibit NF-κB activity in FLSs through BH4 pro-​inflammatory cytokines, including IL-1, TNF and
depletion and nitric oxide synthase uncoupl­ing. Rather, IL-6, in monocytic cell lines81. Induction of these pro-​
methotrexate-stimulated release of adenosine and sub- inflammatory cytokines by methotrexate seems to result
sequent activation of adenosine recep­tors seems to from inhibition of dihydrofolate reduction to tetra­
inhibit the normally high basal levels of NF-​κB activity hydrofolate and not inhibition of BH2 reduction to BH4
in FLSs (Fig. 3b). The differences in responses to metho­ and nitric oxide synthase ‘uncoupling’ or promotion of
trexate between T cells and FLSs seem to be because adenosine receptor signalling (Fig. 3c). This effect might

a T cells b Fibroblast-like synoviocytes c Monocytes


Methotrexate Methotrexate Methotrexate

DHFR Unknown ATIC DHFR


BH2 BH4 mechanism AICAR FAICAR DHF TTF

DNA-PK
NOS uncoupling Adenosine release Unknown
NF-κB–dependent
mechanism

ROS production LincRNA-p21 Adenosine receptor binding

JNK NF-κB activity NF-κB activity Apoptosis IL-1 production IL-6 production

Apoptosis Increase in Anti-inflammatory Anti-inflammatory Pro-inflammatory


sensitivity expression of cell
cycle checkpoints

Anti-inflammatory

Fig. 3 | Cell-specific mechanisms of methotrexate in rheumatoid REL A mRNA , thus reducing levels of the pro-inflammatory transcrip-
arthritis. The actions of methotrexate in distinct cell lineages differ tion factor nuclear factor-κB (NF-κB), inhi­biting inflammation. b | In
depending on the metabolism of each particular cell type. a | In T cells, fibroblast-like synoviocytes, methotrexate also inhibits NF-κB activity
methotrexate inhibits dihydrofolate reductase (DHFR)-mediated reduc- but this effect occurs via inhibition of 5-aminoimidazole-4-carboxamide
tion of dihydrobiopterin (BH2) to tetrahydrobiopterin (BH4), leading to ribonucleotide (AICAR) transformylase (ATIC), increased adenosine
nitric oxide synthase (NOS) uncoupling and increased reactive oxygen release and activation of adenosine receptors on the same cell, lead-
species (ROS) production. ROS activate JUN N-​terminal kinase (JNK), ing to inhibition of NF-​κB with subsequent anti-​inflammatory effects.
and activated JNK induces genes encoding proteins that regulate sen- c | In monocytes, methotrexate can promote apoptosis and increase the
sitivity to apoptosis and cell cycle progression. In T cells, methotrexate expression of pro-inflammatory cytokines via an unknown NF-κB-
also activates DNA-​dependent protein kinase (DNA-​PK) by unknown dependent mechanism that might occur via inhibition of DHFR-mediated
mechanisms, which leads to the induction of long intergenic non-​ reduction of dihydrofolate (DHF) to tetrahydrofolate (TTF). FAICAR ,
coding RNA p21 (lincRNA-​p21). LincRNA-p21 inhibits the translation of formyl AICAR.

Nature Reviews | Rheumatology


Reviews

Box 3 | Predictors of poor response to methotrexate methotrexate therapy without affecting therapeutic effi-
cacy has led to most clinicians prescribing folic acid with
• High DAS28 (refs93,95) or low DAS28 (ref.94) methotrexate and has resulted in a reduced prevalence
(conflicting results) of methotrexate-​related adverse effects12,13,88,89.
• High Health Assessment Questionnaire (HAQ) score93,94 Currently nearly all patients with inflammatory arth­
• High BMI93 ritis are given methotrexate first, in the absence of a con-
• High score on Hospital Anxiety and Depression Scale94 traindication, and if patients do not respond, then they
• High tender joint count94 are prescribed another therapy, usually in combination
• Low erythrocyte folate level93 with methotrexate2. As not all patients respond to metho­
• Single-​nucleotide polymorphism in ABCB1 and trexate, being able to predict which patients are most
ABCC3 (ref.93) likely to respond would be useful to more effectively pre-
• Rheumatoid factor negative94 scribe the right treatment to patients to diminish disease
activity and to reduce unnecessary exposure to poten-
• Current smoker95
tially toxic drugs. Great hope was placed in identifying
• No alcohol consumption95
genetic markers of metho­trexate response, including
some discussed in this Review (TNFAIP3), but, to date,
have clinical implications and might help explain some no factors or biomarkers have been clearly validated as
of the adverse effects of methotrexate, including mucosi- predictive. Both candidate gene studies and genome-​
tis and pneumonitis, which could be mediated by these wide association studies have identified a variety of
pro-​inflammatory cytokines. potential genetic variants associated with a favourable
Adenosine also regulates monocyte function via response to methotrexate, although these associations do
stimu­lation of adenosine receptors, and hence metho- not seem to be reproducible when tested in other popu-
trexate might affect monocytes indirectly by promoting lations90,91. More recently, a machine learning analysis of
adenosine release by other cells. Binding of adenosine transcriptomic data of whole blood from patients with
to adenosine receptor A1 on human peripheral blood RA showed the potential predictive value for metho-
monocytes stimulates the formation of multinucleated trexate response of the expression of genes involved in a
giant cells82. In addition, the binding of adenosine to number of inflammatory pathways (most notably, genes
adenosine receptors A2a and A3 on monocytes inhib- involved in the response to type I interferon); however,
its the synthesis and release of TNF and IL-6 and pro- the inclusion of all of the measured gene transcripts
motes the transition of inflammatory M1 monocytes to in the model gave a better predictive value than the inclu-
anti-inflammatory M2 monocytes (reviewed elsewhere26). sion of gene transcripts of the interferon pathway alone92.
Other studies have attempted to define clinical and labo-
Toxic effects of methotrexate ratory markers that are useful in predicting methotrexate
Clearly many of the toxic effects of long-​term low-​dose response (Box 3), although the markers identified do not
methotrexate therapy result from the antifolate proper- appear to be constant across these studies93–95, perhaps
ties of the drug. These toxic effects include diminished because of the use of different end points.
peripheral blood white cell counts, stomatitis and hair
loss and probably result from methotrexate-​mediated Conclusions
inhibition of cellular proliferation. Many of these toxic Examination of the molecular basis for the therapeutic
effects can be prevented by concomitant administration activity of methotrexate shows that multiple pathways
of folic acid or folinic acid (except on the day of metho- are activated by methotrexate, all of which contribute to
trexate administration) as methotrexate and folinic acid the suppression of inflammation in RA. The underlying
compete for the same transporter for cellular uptake. biochemical mechanisms include inhibition of ATIC
By contrast, a number of the toxic effects of methotrex- (leading to increased release of adenosine, which sup-
ate probably result from methotrexate-​mediated adeno- presses inflammatory and immune functions via inter­
sine release. Among these adenosine-​related toxic effects action with cell surface receptors); inhibition of DHFR and
are the feeling of tiredness many patients experience on diminished reduction of BH2 to BH4 (leading to uncou-
the day of methotrexate administration (adenosine is pling of nitric oxide synthases and increased production
released in the brain and binds to adenosine receptors of reactive oxygen species) and JNK activation; increased
in the central nervous system, promoting sleep83–85), expression of lincRNA-​p21, a lncRNA that diminishes
rheumatoid nodulosis82 and hepatic fibrosis86,87. a variety of immune and inflammatory reactions; and
direct and indirect (adenosine-​mediated) inhibition
Therapeutic implications of signalling via NF-​κB and the JAK–STAT pathway,
Methotrexate remains the most commonly used drug critical promoters of inflammatory and immune reac-
in the treatment of RA and other rheumatic diseases. tions. Many of the effects of methotrexate that have
Because the bioavailability of orally administered metho­ been described seem to be cell-​type specific. In T cells,
trexate is limited, many patients can probably achieve dominant pathways include increased sensitivity to
better responses to the drug by parenteral adminis­tration apoptosis and inhibition of NF-​κB activity. Moreover,
of methotrexate or by dividing the dose of the drug over the suppression of T cell activity by Treg cells via the
the course of a day as opposed to taking a single daily secretion of adenosine is promoted by methotrexate, and
oral dose. The demonstration that folic acid administra- adenosine itself promotes the differentiation of Treg cells
tion can prevent many of the toxic effects associated with that can suppress inflammation. Many of these in vitro

www.nature.com/nrrheum
Reviews

effects also occur in patients with RA who are receiving these repair mechanisms. Therefore, we speculate that
methotrexate therapy. the danger signal that initiates the innate immune
One hypothesis that arises from the various mecha­ response in RA could also be an internal signal produced
nisms of action of methotrexate in RA is that metho- by this defective repair machinery rather than an exter-
trexate regulates responses to ‘danger signals’. RA is nal signal and might ultimately result in a continuous
considered to arise from an adaptive immune response cycle of chronic inflammation. Methotrexate directly tar-
to self-​antigens, and in the search for such antigens, gets this proposed internal danger signal, which might
citrullinated peptides, the levels of which are increased contribute to its therapeutic effects in RA in addition
in RA, have been implicated96,97. An adaptive immune to the suppression of an overly exuberant immune and
response to a foreign antigen is generally thought inflammatory response.
to require both a danger signal, to activate the innate Clearly, all of the mechanisms described to date will
immune response, and a foreign antigen, to stimulate contribute to the therapeutic efficacy of methotrexate
the adaptive immune response98,99. By altering the con- therapy in RA. Insights into non-​folate-dependent anti-​
centrations of adenosine, methotrexate alters responses inflammatory mechanisms of methotrexate have led to
to these antigenic danger signals. Moreover, the cell cycle a marked reduction in clinical toxicity of the drug and
checkpoints and DNA repair machinery responsible for thereby increased its use in the treatment of RA.
maintaining appropriate cellular function also inhibit
pro-​inflammatory pathways, and methotrexate regulates Published online xx xx xxxx

1. Weinblatt, M. E. Methotrexate: who would have 16. Joyce, D. A., Will, R. K., Hoffman, D. M., Laing, B. 30. Montesinos, M. C. et al. Adenosine A2A or A3
predicted its importance in rheumatoid arthritis? & Blackbourn, S. J. Exacerbation of rheumatoid receptors are required for inhibition of inflammation
Arthritis Res. Ther. 20, 103 (2018). arthritis in patients treated with methotrexate after by methotrexate and its analog MX-68. Arthritis
2. Singh, J. A. et al. 2015 American College of administration of folinic acid. Ann. Rheum. Dis. 50, Rheum. 48, 240–247 (2003).
Rheumatology guideline for the treatment 913–914 (1991). 31. Riksen, N. P. et al. Methotrexate modulates the kinetics
of rheumatoid arthritis. Arthritis Care Res. 68, 1–25 17. Tishler, M., Caspi, D., Fishel, B. & Yaron, M. The effects of adenosine in humans in vivo. Ann. Rheum. Dis. 65,
(2016). of leucovorin (folinic acid) on methotrexate therapy in 465–470 (2006).
3. Aaltonen, K. J. et al. Do biologic drugs affect the need rheumatoid arthritis patients. Arthritis Rheum. 31, 32. Nesher, G., Mates, M. & Zevin, S. Effect of caffeine
for and outcome of joint replacements in patients 906–908 (1988). consumption on efficacy of methotrexate in rheumatoid
with rheumatoid arthritis? A register-​based study. 18. Yukioka, K. et al. Polyamine levels in synovial tissues arthritis. Arthritis Rheum. 48, 571–572 (2003).
Semin. Arthritis Rheum. 43, 55–62 (2013). and synovial fluids of patients with rheumatoid 33. Benito-​Garcia, E. et al. Dietary caffeine intake does not
4. Asai, S. et al. Effects of concomitant methotrexate on arthritis. J. Rheumatol. 19, 689–692 (1992). affect methotrexate efficacy in patients with rheumatoid
large joint replacement in patients with rheumatoid 19. Nesher, G. & Moore, T. L. The in vitro effects of arthritis. J. Rheumatol. 33, 1275–1281 (2006).
arthritis treated with tumor necrosis factor inhibitors: methotrexate on peripheral blood mononuclear cells. 34. Allard, D., Turcotte, M. & Stagg, J. Targeting A2
a multicenter retrospective cohort study in Japan. Modulation by methyl donors and spermidine. adenosine receptors in cancer. Immunol. Cell Biol. 95,
Arthritis Care Res. 67, 1363–1370 (2015). Arthritis Rheum. 33, 954–959 (1990). 333–339 (2017).
5. Asai, S. et al. Concomitant methotrexate protects 20. Furumitsu, Y. et al. Levels of urinary polyamines in 35. Peres, R. S. et al. Low expression of CD39 on
against total knee arthroplasty in patients with patients with rheumatoid arthritis. J. Rheumatol. 20, regulatory T cells as a biomarker for resistance to
rheumatoid arthritis treated with tumor necrosis factor 1661–1665 (1993). methotrexate therapy in rheumatoid arthritis.
inhibitors. J. Rheumatol. 42, 2255–2260 (2015). 21. Nesher, G., Osborn, T. G. & Moore, T. L. In vitro effects of Proc. Natl Acad. Sci. USA 112, 2509–2514 (2015).
6. Herman, R. A., Veng-​Pedersen, P., Hoffman, J., methotrexate on polyamine levels in lymphocytes from 36. Bitoun, S. et al. Methotrexate and BAFF interaction
Koehnke, R. & Furst, D. E. Pharmacokinetics of rheumatoid arthritis patients. Clin. Exp. Rheumatol. 14, prevents immunization against TNF inhibitors.
low-dose methotrexate in rheumatoid arthritis 395–399 (1996). Ann. Rheum. Dis. 77, 1463–1470 (2018).
patients. J. Pharm. Sci. 78, 165–171 (1989). 22. Nesher, G., Moore, T. L. & Dorner, R. W. In vitro effects 37. Chalupsky, K. & Cai, H. Endothelial dihydrofolate
7. Schiff, M. H. & Sadowski, P. Oral to subcutaneous of methotrexate on peripheral blood monocytes: reductase: critical for nitric oxide bioavailability and
methotrexate dose-​conversion strategy in the modulation by folinic acid and S-adenosylmethionine. role in angiotensin II uncoupling of endothelial nitric
treatment of rheumatoid arthritis. Rheumatol. Int. 37, Ann. Rheum. Dis. 50, 637–641 (1991). oxide synthase. Proc. Natl Acad. Sci. USA 102,
213–218 (2017). 23. Chan, E. S. & Cronstein, B. N. Methotrexate–how 9056–9061 (2005).
8. Kremer, J. M., Galivan, J., Streckfuss, A. & Kamen, B. does it really work? Nat. Rev. Rheumatol. 6, 175–178 38. Crabtree, M. J., Tatham, A. L., Hale, A. B., Alp, N. J.
Methotrexate metabolism analysis in blood and liver of (2010). & Channon, K. M. Critical role for tetrahydrobiopterin
rheumatoid arthritis patients. Association with hepatic 24. Allegra, C. J., Drake, J. C., Jolivet, J. & Chabner, B. A. recycling by dihydrofolate reductase in regulation of
folate deficiency and formation of polyglutamates. Inhibition of phosphoribosylaminoimidazolecarboxamide endothelial nitric-​oxide synthase coupling – relative
Arthritis Rheum. 29, 832–835 (1986). transformylase by methotrexate and dihydrofolic acid importance of the de novo biopterin synthesis versus
9. Chabner, B. A. et al. Polyglutamation of methotrexate. polyglutamates. Proc. Natl Acad. Sci. USA 82, salvage pathways. J. Biol. Chem. 284, 28128–28136
Is methotrexate a prodrug? J. Clin. Invest. 76, 4881–4885 (1985). (2009).
907–912 (1985). 25. Cronstein, B. N., Naime, D. & Ostad, E. 39. Sugiyama, T., Levy, B. D. & Michel, T. Tetrahydrobiopterin
10. Steffen, J. A. & Stolzmann, W. M. Studies on in vitro The antiinflammatory mechanism of methotrexate: recycling, a key determinant of endothelial nitric-oxide
lymphocyte proliferation in cultures synchronized by increased adenosine release at inflamed sites synthase-​dependent signaling pathways in cultured
the inhibition of DNA synthesis. I. Variability of S plus diminishes leukocyte accumulation in an in vivo vascular endothelial cells. J. Biol. Chem. 284,
G2 periods of first generation cells. Exp. Cell Res. 56, model of inflammation. J. Clin. Invest. 92, 12691–12700 (2009).
453–460 (1969). 2675–2682 (1993). 40. Spurlock, C. F. et al. Increased sensitivity to apoptosis
11. Morgan, S. L., Baggott, J. E., Lee, J. Y. & Alarcon, G. S. 26. Cronstein, B. N. & Sitkovsky, M. Adenosine and induced by methotrexate is mediated by JNK. Arthritis
Folic acid supplementation prevents deficient blood adenosine receptors in the pathogenesis and treatment Rheum. 63, 2606–2616 (2011).
folate levels and hyperhomocysteinemia during of rheumatic diseases. Nat. Rev. Rheumatol. 13, 41. Spurlock, C. F., Tossberg, J. T., Fuchs, H. A., Olsen, N. J.
longterm, low dose methotrexate therapy for 41–51 (2017). & Aune, T. M. Methotrexate increases expression of cell
rheumatoid arthritis: implications for cardiovascular 27. Montesinos, M. C. et al. Reversal of the cycle checkpoint genes via JNK activation. Arthritis
disease prevention. J. Rheumatol. 25, 441–446 (1998). antiinflammatory effects of methotrexate by Rheum. 64, 1780–1789 (2012).
12. Morgan, S. L. et al. Supplementation with folic acid the nonselective adenosine receptor antagonists 42. Spurlock, C. F. et al. Methotrexate-mediated inhibition
during methotrexate therapy for rheumatoid arthritis. theophylline and caffeine: evidence that the of nuclear factor κB activation by distinct pathways in
A double-​blind, placebo-​controlled trial. Ann. Int. Med. antiinflammatory effects of methotrexate are mediated T cells and fibroblast-like synoviocytes. Rheumatology
121, 833–841 (1994). via multiple adenosine receptors in rat adjuvant 54, 178–187 (2015).
13. Morgan, S. L. et al. The effect of folic acid arthritis. Arthritis Rheum. 43, 656–663 (2000). 43. Rinn, J. L. & Chang, H. Y. Genome regulation by long
supplementation on the toxicity of low-​dose 28. Montesinos, M. C. et al. The antiinflammatory noncoding RNAs. Annu. Rev. Biochem. 81, 145–166
methotrexate in patients with rheumatoid arthritis. mechanism of methotrexate depends on extracellular (2012).
Arthritis Rheum. 33, 9–18 (1990). conversion of adenine nucleotides to adenosine by 44. Huarte, M. et al. A large intergenic noncoding RNA
14. Shiroky, J. B. et al. Low-​dose methotrexate with ecto-5’-nucleotidase: findings in a study of ecto-5’- induced by p53 mediates global gene repression in
leucovorin (folinic acid) in the management of nucleotidase gene-deficient mice. Arthritis Rheum. the p53 response. Cell 142, 409–419 (2010).
rheumatoid arthritis. Results of a multicenter 56, 1440–1445 (2007). 45. Weinblatt, M. E. Methotrexate in rheumatoid arthritis:
randomized, double-​blind, placebo-​controlled trial. 29. Montesinos, M. C., Desai, A. & Cronstein, B. N. a quarter century of development. Trans. Am. Clin.
Arthritis Rheum. 36, 795–803 (1993). Suppression of inflammation by low-​dose methotrexate Climatol. Assoc. 124, 16–25 (2013).
15. Shea, B. Folic acid or folinic acid for reducing side is mediated by adenosine A2A receptor but not A3 46. Yang, F., Zhang, H. F., Mei, Y. D. & Wu, M. Reciprocal
effects of methotrexate for people with rheumatoid receptor activation in thioglycollate-​induced peritonitis. regulation of HIF-1α and lincRNA-​p21 modulates the
arthritis. J. Evid. Based Med. 6, 202–203 (2013). Arthritis Res. Ther. 8, R53 (2006). Warburg effect. Mol. Cell 53, 88–100 (2014).

Nature Reviews | Rheumatology


Reviews

47. Yoon, J. H. et al. LincRNA-​p21 suppresses target 69. Menendez, D., Shatz, M. & Resnick, M. A. Interactions 92. Plant, D. et al. Profiling of gene expression biomarkers
mRNA translation. Mol. Cell 47, 648–655 (2012). between the tumor suppressor p53 and immune as a classifier of methotrexate nonresponse in patients
48. Spurlock, C. F., Tossberg, J. T., Matlock, B. K., Olsen, N. J. responses. Curr. Opin. Oncol. 25, 85–92 (2013). with rheumatoid arthritis. Arthritis Rheumatol. 71,
& Aune, T. M. Methotrexate inhibits NF-κB activity 70. Cooks, T., Harris, C. C. & Oren, M. Caught in the 678–684 (2019).
via long intergenic (noncoding) RNA-p21 induction. cross fire: p53 in inflammation. Carcinogenesis 35, 93. de Rotte, M. et al. Development and validation
Arthritis Rheumatol. 66, 2947–2957 (2014). 1680–1690 (2014). of a prognostic multivariable model to predict
49. Malemud, C. J. The role of the JAK/STAT signal 71. Takatori, H., Kawashima, H., Suzuki, K. & Nakajima, H. insufficient clinical response to methotrexate in
pathway in rheumatoid arthritis. Ther. Adv. Role of p53 in systemic autoimmune diseases. rheumatoid arthritis. PLoS One 13, e0208534
Musculoskelet. Dis. 10, 117–127 (2018). Crit. Rev. Immunol. 34, 509–516 (2014). (2018).
50. Gremese, E., Alivernini, S., Tolusso, B., Zeidler, M. P. 72. Rackov, G. et al. p21 mediates macrophage 94. Sergeant, J. C. et al. Prediction of primary
& Ferraccioli, G. JAK inhibition by methotrexate reprogramming through regulation of p50-p50 NF-κB non-response to methotrexate therapy using
(and csDMARDs) may explain clinical efficacy as and IFN-β. J. Clin. Invest. 126, 3089–3103 (2016). demographic, clinical and psychosocial variables:
monotherapy and combination therapy. J. Leukoc. Biol. 73. Wu, Z. H., Shi, Y. L., Tibbetts, R. S. & Miyamoto, S. results from the UK Rheumatoid Arthritis Medication
106, 1063–1068 (2019). Molecular linkage between the kinase ATM and NF-κB Study (RAMS). Arthritis Res. Ther. 20, 147 (2018).
51. Thomas, S. et al. Methotrexate Is a JAK/STAT pathway signaling in response to genotoxic stimuli. Science 95. Teitsma, X. M. et al. Inadequate response to treat-​
inhibitor. PLoS One 10, e0130078 (2015). 311, 1141–1146 (2006). to-target methotrexate therapy in patients with
52. Hassanian, S. M., Dinarvand, P. & Rezaie, A. R. 74. Zhang, T. et al. p53 predominantly regulates IL-6 new-onset rheumatoid arthritis: development and
Adenosine regulates the proinflammatory signaling production and suppresses synovial inflammation in validation of clinical predictors. Ann. Rheum. Dis. 77,
function of thrombin in endothelial cells. J. Cell Physiol. fibroblast-​like synoviocytes and adjuvant-​induced 1261–1267 (2018).
229, 1292–1300 (2014). arthritis. Arthritis Res. Ther. 18, 271 (2016). 96. Szodoray, P. et al. Anti-​citrullinated protein/peptide
53. Yang, J. et al. Adenosine increases LPS-​induced 75. McInnes, I. B. & Schett, G. Cytokines in the pathogenesis autoantibodies in association with genetic and
nuclear factor kappa B activation in smooth muscle of rheumatoid arthritis. Nat. Rev. Immunol. 7, 429–442 environmental factors as indicators of disease
cells via an intracellular mechanism and modulates it (2007). outcome in rheumatoid arthritis. Autoimmun. Rev. 9,
via actions on adenosine receptors. Acta Physiol. 210, 76. Zheng, S. J., Lamhamedi-​Cherradi, S. E., Wang, P., 140–143 (2010).
590–599 (2014). Xu, L. Y. & Chen, Y. H. Tumor suppressor p53 inhibits 97. Luban, S. & Li, Z. G. Citrullinated peptide and its
54. Mediero, A., Perez-​Aso, M. & Cronstein, B. N. autoimmune inflammation and macrophage function. relevance to rheumatoid arthritis: an update. Int. J.
Activation of adenosine A2A receptor reduces Diabetes 54, 1423–1428 (2005). Rheum. Dis. 13, 284–287 (2010).
osteoclast formation via PKA- and ERK1/2-mediated 77. Okuda, Y., Okuda, M. & Bernard, C. C. A. 98. Gallucci, S. & Matzinger, P. Danger signals: SOS to the
suppression of NFκB nuclear translocation. Br. J. Regulatory role of p53 in experimental autoimmune immune system. Curr. Opin. Immunol. 13, 114–119
Pharmacol. 169, 1372–1388 (2013). encephalomyelitis. J. Neuroimmunol. 135, 29–37 (2001).
55. Tang, L. M. et al. Activation of adenosine A2A receptor (2003). 99. Matzinger, P. Friendly and dangerous signals: is the
attenuates inflammatory response in a rat model of 78. Kawashima, H. et al. Tumor suppressor p53 inhibits tissue in control? Nat. Immunol. 8, 11–13 (2007).
small-​for-size liver transplantation. Transpl. Proc. 42, systemic autoimmune diseases by inducing regulatory 100. Gubner, R., August, S. & Ginsberg, V. Therapeutic
1915–1920 (2010). T cells. J. Immunol. 191, 3614–3623 (2013). suppression of tissue reactivity. II. Effect of aminopterin
56. Di Paola, R. et al. Adenosine A2A receptor-​selective 79. Munoz-​Fontela, C., Mandinova, A., Aaronson, S. A. in rheumatoid arthritis and psoriasis. Am. J. Med. Sci.
stimulation reduces signaling pathways involved in the & Lee, S. W. Emerging roles of p53 and other tumour-​ 221, 176–182 (1951).
development of intestine ischemia and reperfusion suppressor genes in immune regulation. Nat. Rev. 101. Weinblatt, M. E. et al. Efficacy of low-​dose methotrexate
injury. Shock 33, 541–551 (2010). Immunol. 16, 741–750 (2016). in rheumatoid arthritis. N. Engl. J. Med. 312, 818–822
57. Ramanathan, M., Pinhal-Enfield, G., Hao, I. & 80. Bartok, B. & Firestein, G. S. Fibroblast-​like synoviocytes: (1985).
Leibovich, S. J. Synergistic up-regulation of vascular key effector cells in rheumatoid arthritis. Immunol. Rev. 102. Williams, H. J. et al. Comparison of low-​dose oral
endothelial growth factor (VEGF) expression in 233, 233–255 (2010). pulse methotrexate and placebo in the treatment of
macrophages by adenosine A2A receptor agonists and 81. Olsen, N. J., Spurlock, C. F. & Aune, T. M. Methotrexate rheumatoid arthritis: a controlled clinical trial.
endotoxin involves transcriptional regulation via the induces production of IL-1 and IL-6 in the monocytic Arthritis Rheum. 28, 721–730 (1985).
hypoxia response element in the VEGF promoter. cell line U937. Arthritis Res. Ther. 16, R17 (2014). 103. Kremer, J. M. & Lee, J. K. The safety and efficacy of
Mol. Biol. Cell 18, 14–23 (2007). 82. Merrill, J. T. et al. Adenosine A1 receptor promotion the use of methotrexate in long-​term therapy for
58. Zernecke, A. et al. CD73/ecto-5’-nucleotidase protects of multinucleated giant cell formation by human rheumatoid arthritis. Arthritis Rheum. 29, 822–831
against vascular inflammation and neointima formation. monocytes: a mechanism for methotrexate-induced (1986).
Circulation 113, 2120–2127 (2006). nodulosis in rheumatoid arthritis. Arthritis Rheum. 40, 104. US Food and Drug Administration. Drug approval
59. Sands, W. A., Martin, A. F., Strong, E. W. & Palmer, T. M. 1308–1315 (1997). package. accessdata.fda.gov https://www.
Specific inhibition of nuclear factor-κB-dependent 83. Chagoya de Sanchez, V. et al. Day-​night variations of accessdata.fda.gov/drugsatfda_docs/nda/2013/
inflammatory responses by cell type-specific adenosine and its metabolizing enzymes in the brain 204824Orig2s000TOC.cfm (2014).
mechanisms upon A2A adenosine receptor gene cortex of the rat–possible physiological significance 105. US Food and Drug Administration. Rasuvo
transfer. Mol. Pharmacol. 66, 1147–1159 (2004). for the energetic homeostasis and the sleep-wake (methotrexate) injection accessdata.fda.gov.
60. Lukashev, D., Ohta, A., Apasov, S., Chen, J. F. cycle. Brain Res. 612, 115–121 (1993). https://www.accessdata.fda.gov/drugsatfda_docs/
& Sitkovsky, M. Cutting edge: physiologic attenuation 84. Chagoya de Sanchez, V. Circadian variations of nda/2014/205776Orig1s000TOC.cfm (2015).
of proinflammatory transcription by the Gs protein- adenosine and of its metabolism. Could adenosine be
coupled A2A adenosine receptor in vivo. J. Immunol. a molecular oscillator for circadian rhythms? Can. J. Acknowledgements
173, 21–24 (2004). Physiol. Pharmacol. 73, 339–355 (1995). The work of T.M.A. was supported by grants from the US
61. Bshesh, K. et al. The A2A receptor mediates an 85. Chagoya de Sanchez, V. et al. Temporal variations of National Institutes of Health (R21AR063846, R42AI53948,
endogenous regulatory pathway of cytokine expression adenosine metabolism in human blood. Chronobiol. R01AI044924), the American College of Rheumatology
in THP-1 cells. J. Leukoc. Biol. 72, 1027–1036 Int. 13, 163–177 (1996). ‘Within Our Reach’ grant programme (ACR124405), the
(2002). 86. Chan, E. S. et al. Adenosine A2A receptors play a role in US National Center for Advancing Translation Sciences
62. Plant, D. et al. A genetic marker at the OLIG3/ the pathogenesis of hepatic cirrhosis. Br. J. Pharmacol. (UL1TR000445) and the US National Science Foundation
TNFAIP3 locus associates with methotrexate 148, 1144–1155 (2006). Graduate Research Fellowship Program.
continuation in early inflammatory polyarthritis: 87. Che, J., Chan, E. S. & Cronstein, B. N. Adenosine A2A
results from the Norfolk arthritis register. receptor occupancy stimulates collagen expression Author contributions
Pharmacogenomics J. 12, 128–133 (2012). by hepatic stellate cells via pathways involving The authors contributed equally to all aspects of the article.
63. Municio, C. et al. Methotrexate selectively targets protein kinase A, Src, and extracellular signal-regulated
human proinflammatory macrophages through a kinases 1/2 signaling cascade or p38 mitogen-activated Competing interests
thymidylate synthase/p53 axis. Ann. Rheum. Dis. protein kinase signaling pathway. Mol. Pharmacol. 72, B.N.C. declares that he holds equity in Regenosine, a biotech
75, 2157–2165 (2016). 1626–1636 (2007). start-​up developing therapies for osteoarthritis, and CanFite
64. Municio, C. et al. Methotrexate limits inflammation 88. Morgan, S. L., Baggott, J. E., Koopman, W. J., Biopharma, that he has received grant support from AstraZeneca
through an A20-dependent cross-​tolerance mechanism. Krumdieck, C. L. & Alarcon, G. S. Folate and Kairos Inc., and that he has a number of patents involving
Ann. Rheum. Dis. 77, 752–759 (2018). supplementation and methotrexate. Ann. Rheum. Dis. adenosine receptors and hepatic fibrosis, wound healing, bone
65. Spurlock, C. F. III et al. Methotrexate-mediated 52, 315–316 (1993). regeneration and osteoarthritis. T.M.A. declares that he has no
inhibition of nuclear factor κB activation by distinct 89. Alarcon, G. S. & Morgan, S. L. Guidelines for folate competing interests.
pathways in T cells and fibroblast-like synoviocytes. supplementation in rheumatoid arthritis patients
Rheumatology 54, 178–187 (2015). treated with methotrexate: comment on the guidelines
Peer review information
66. Vousden, K. H. & Prives, C. Blinded by the light: for monitoring drug therapy. Arthritis Rheum. 40, 391
Nature Reviews Rheumatology thanks W. Wei and the other,
the growing complexity of p53. Cell 137, 413–431 (1997).
anonymous, reviewer(s) for their contribution to the peer
(2009). 90. Roszkiewicz, J. & Smolewska, E. In the pursuit of
review of this work.
67. Kang, C. et al. The DNA damage response induces methotrexate treatment response biomarker in
inflammation and senescence by inhibiting autophagy Juvenile idiopathic arthritis — are we getting closer
of GATA4. Science 349, aaa5612 (2015). to personalised medicine? Curr. Rheumatol. Rep. 19, Publisher’s note
68. Cooks, T. et al. Mutant p53 prolongs NF-κB activation 19 (2017). Springer Nature remains neutral with regard to jurisdictional
and promotes chronic inflammation and inflammation- 91. Taylor, J. C. et al. Genome-​wide association study of claims in published maps and institutional affiliations.
associated colorectal cancer. Cancer Cell 23, 634–646 response to methotrexate in early rheumatoid arthritis
(2013). patients. Pharmacogenomics J. 18, 528–538 (2018). © Springer Nature Limited 2020

www.nature.com/nrrheum

You might also like