You are on page 1of 7

Best Practice & Research Clinical Gastroenterology 54-55 (2021) 101757

Contents lists available at ScienceDirect

Best Practice & Research Clinical Gastroenterology


journal homepage: www.elsevier.com/locate/bpg

Immunosuppressive drugs modes of action


Maria Meneghini a, Oriol Bestard a, Josep Maria Grinyo b, *
a
Nephrology and Kidney Transplantation Unit. Vall D’Hebron University Hospital, Barcelona. Spain
b
University of Barcelona. Department of Clinical Sciences. Barcelona. Spain

A R T I C L E I N F O A B S T R A C T

Keywords: The innate and adaptive immune systems work as a complex interplay between different cell types, involving
Immunosuppression cytokines and chemokines mediating extracellular and paracrine effects. At the intracellular level, the inflam­
Allotransplantation matory cascade is mediated by multifaceted processes that have been better described in the last 10 years.
Autoimmunity
Immunosuppressive agents available in clinical practice act at different points of those cascades at the intra­
cellular or extracellular level. Those drugs can mediate their effects on one or more cell types finally limiting
inflammation and immune responses to antigens. Every immunosuppressive agent is characterized by intrinsic
toxicity and side effects that may be due to the same therapeutic pathways or to off-target secondary effect of
each molecule. We will here review the mechanisms of action of the most widely used immunosuppressive agents
in the field of solid organ transplantation and autoimmune disorders, describing the mechanisms underlying both
the therapeutic and secondary effects.

Introduction When used at low dose (<30 mg of prednisone) the activity is mediated
by interaction with the intracellular cytoplasm glucocorticoid receptor
The introduction of drugs able to modulate the innate and adaptive (GR) a member of the nuclear receptor family of ligand-activated tran­
harms of the immune system has allowed significant advances in the scription factors. After binding of glucocorticoids to GR, the GR un­
outcomes of non- HLA identical solid organ transplantation as well as in dergoes hyperphosphorylation, dissociates from the multiprotein
the control of autoimmune diseases. In fact, both alloimmunity and complex usually associated and can translocate into the nucleus, where
autoimmunity share common biological mechanisms. The most widely it is involved in gene expression regulation (genomic action). The action
used immunosuppressants act at different levels of the immunological can be mediated by direct activation and repression of specific genes but
response, both at the T cell and B-cell level. is mainly due to an interaction with transcription factors. One of the
We will here review the mechanisms of action of the immunosup­ most important is the NF-kb and AP-1 complexes, cytoplasmatic regu­
pressive agents used in clinical practice (Table 1). lators of several genes controlling the transcription and synthesis of
adhesion molecules and cytokines. NF-Kb activation is inhibited by the
Immunosuppressive agents presence of the complex corticosteroids-cytoplasmatic receptor through
the synthesis of the inhibiting factor IkB. This effect results in the inhi­
Small molecules bition of proinflammatory cytokines (IL-1, IL-6, IFN, nitric oxide) in
different cell types [2,3]. Most of the effects are specific for each cell
Corticosteroids type with different genes being targeted according to the GR binding site
Corticosteroids are the steroid hormones of adrenal cortex produc­ and possible recruitment of coactivators.
tion, further classified in glucocorticoids and mineralocorticoids. In At high doses (>30 mg of Prednisone) the effect is not mediated by
clinical practice, glucocorticoids and corticosteroids can be used as genetic transcription but through direct impact on membrane fluidity
synonyms. Corticosteroids are historically the cornerstone of most because of the lipophilic structure of corticosteroids as well as by
immunosuppressive approaches, having pleiotropic effects (and side interaction with cytosolic GR or membrane-bound GR (non-genomic
effects). action). Since no protein synthesis is necessary, the action is faster,
Those molecules act at a different level according to their dosing [1]. within minutes. High doses steroids result in altering of physicochemical

* Corresponding author.
E-mail addresses: mmeneghini@vhebron.net (M. Meneghini), obestard@vhebron.net (O. Bestard), jgrinyo@ub.edu (J.M. Grinyo).

https://doi.org/10.1016/j.bpg.2021.101757
Received 5 June 2021; Accepted 11 June 2021
Available online 16 June 2021
1521-6918/© 2021 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
M. Meneghini et al. Best Practice & Research Clinical Gastroenterology 54-55 (2021) 101757

Table 1 both T and B lymphocytes are dependent on the de novo pathway of


Description and mode of action of most used immunosuppressants. purine synthesis, those cells are the main target of MPA. Moreover, MPA
Description Mechanisms of action also blocks adhesion molecules by interacting with transfer of fucose and
mannose residues of glycoproteins. In particular, it inhibits the integrin
SMALL MOLECULES
Corticosteroids Steroid hormones Pleiotropic effects. VLA-4 synthesis reducing leukocyte adhesion to endothelial cells [7].
Inhibition of The MPA Cmin plasma has a discrete correlation with AUC (r = 0.64).
proinflammatory In practice it is recommended to maintain MPA Cmin values between 1
genes’transcription. and 3 mg/mL or AUC of MPA between 30 and 55 mcg/ml/h to prevent
Antimetabolite Competitive (Azathioprine, Interference with DNA
agents Methotrexate) or Non- synthesis and cell
acute rejection in kidney transplantation [8].
competitive (MPA) proliferation. Mycophenolate mofetil (MMF) has an oral bioavailability greater
inhibitors of nucleic acid than 90%, with high inter-individual pharmacokinetic variability. After
synthesis absorption, the ester is hydrolyzed releasing the MPA (having a 1st peak
Calcineurin CsA: lipophilic cyclic Binding to Calcineurin
between 0.6 and 1 h). MPA is metabolized to inactive glucuronide
inhibitors endecapeptide; TAC:
macrolide antibiotic (MPAG) with renal elimination. In cases of renal impairment, MPAG in
mTOR inhibitors SRL: macrolide antibiotic; Formation of a complex with excess can compete with MPA for protein binding resulting in an
EVL: SRL analogue the FKBP12, binding to the increased free fraction. MPAG also has biliary excretion and is decon­
mTOR kinase jugated by the intestinal flora, resulting in an enterohepatic circulation
POLYCLONAL BIOLOGIC PREPARATIONS
Polyclonal Polyclonal Depletion of Lymphocytes
which accounts for the second peak of MPA between 6 and 12 h after
antilymphocyte immunoglobulins from and other inhibitory oral administration of MMF [9]. Importantly, this entero-hepatic cir­
sera rabbits immunized with functions on T cells culation, is inhibited by the calcineurin-inhibitor Cyclosporin A, causing
human thymocytes or lower bioavailability of MPA when these two agents are used in
Jurkat cells
combination.
Intravenous Plasma-derived human Pleiotropic
immunoglobulins immunoglobulins immunomodulatory effects. Despite equivalent immunosuppressive effect, some differences exist
BIOLOGIC AGENTS between the two available formulations of MPA. The more recent
MONOCLONAL OR formulation of MPA is mycophenolate sodium enteric-coated (MPS),
FUSION specifically designed to attenuate gastrointestinal effects of MPA drugs.
PROTEINS
The main difference with MMF is that, due to the enteric coating, MPS is
Costimulation Chimeric CTLA-4 proteins Downregulation of CD28-B7
blockade costimulatory signal dissolved in the bowel and not in the gastric acid possibly limiting some
Anti -IL2 R Chimeric monoclonal Blocking and internalization of the gastrointestinal side effects [10].
antibody of IL2-R reducing IL-2 Methotrexate is also an anti-metabolite inhibiting DNA/RNA syn­
dependent T cell activation-
thesis by interference with purine metabolism. It is a folic acid antago­
proliferation
Anti-CD52 Humanized monoclonal Depletion of CD52 positive nist and by inhibiting the enzyme dihydrofolate reductase deprives of
monocolonal antibody cells RNA and DNA all those cells with rapid differentiating cell cycle. It also
antibody has an effect of downregulation of adhesion molecules, and interference
Anti- CD20 Chimeric monoclonal Depletion of CD20 positive at the IL-1β - IL-1R interaction. After rapid entrance into the different
monocolonal antibody cells
cell types, methotrexate is converted in methotrexate polyglutamates,
antibody
poorly moving through cellular membranes. This leads to cellular
Abbreviations: MPA: Mycophenolic acid; CsA: Cyclosporine A; TAC: Tacrolimus; entrapment, causing retention into liver and kidney and possible toxicity
SRL: Sirolimus; EVL: Everolimus; FKBP-12, FK binding protein; IL-2R Interleukin to those cells. The MTX polyglutamates are thought to be responsible for
2 receptor.
most of the anti-inflammatory effects.
Nowadays, it has a limited use as compared with Azathioprine and
properties of cell membrane and modification of intracellular Ca2+ MPA in the setting of antirejection treatments as well as autoimmune
concentrations and interaction with signaling cascades such as MAPK diseases [11].
signaling pathways [4]. In thymocytes, those agents can mediate regu­
lated apoptosis [5]. Calcineurin inhibitors (CNI)
These agents are of great importance in the prevention of solid organ
Antimetabolite agents transplant rejection and in the maintenance treatment of T-cell mediated
These drugs act by interfering with DNA synthesis and cell autoimmune diseases.
proliferation. Cyclosporin A (CsA) and Tacrolimus (TAC) have different structures
Azathioprine is a purine analogue that mediates its effect by pre­ but both act by binding to cytoplasmic proteins belonging to the family
venting the synthesis of nucleic acids. It is metabolized in the liver into of immunophilins and finally interacting with the action of calcineurin
thioinosinic acid and 6-methyl mercaptopurine. The active compound is (also known as Protein-phosphatase 2B) [12].
thioinosinic acid, a purine analogue of guanine interfering with RNA Cyclosporine (CsA) is a highly lipophilic cyclic endecapeptide iso­
and DNA synthesis and resulting in a cytotoxic effect on leukocytes. lated from the fungus Tolypocladium inflatum that binds to cyclophilin.
Degradation of azathioprine metabolites is mediated by xanthine oxi­ The CsA-cyclophilin complex inhibits the action of calcineurin. Tacro­
dase enzyme [6]. limus (TAC) is a macrolide isolated from Streptomyces tsukubaensis.
Mycophenolic acid (MPA) is the active ingredient of two drugs: The cytosolic immunophilin blocked by TAC is called FKBP-12 (FK
Mycophenolate mofetil (MMF) and enteric-coated mycophenolate so­ binding protein). The TAC-FKBP complex also inhibits the enzyme
dium (MPS). calcineurin.
MPA is a reversible and non-competitive inhibitor of the inosine By inhibiting calcineurin, both drugs block the transcription of nu­
monophosphate dehydrogenase (IMPDH) enzyme. This enzyme acts clear factors of activated T cells (NFAT). The result is that T cells are
catalyzing the conversion of inosine monophosphate into guanine. maintained in a resting state because T-cell stimulation signals do not
Therefore, the inhibition results in impaired de novo synthesis of guanine result in the transition of G0 to G1 of cell cycle. Also, calcineurin inhi­
and DNA replication. Both isoforms of IMPDH, the constitutive and bition prevents the synthesis of various cytokines such as IL-2, IL-4 and
inducible are targeted by MPA but the action is significantly stronger on IFN-γ, and IL-2 receptor. At the same time, CNIs act on regulatory
the inducible isoform, which is expressed during cell activation. Since molecules of T-cell activation such as NF-kB, JNK (N-terminal kinase c-

2
M. Meneghini et al. Best Practice & Research Clinical Gastroenterology 54-55 (2021) 101757

Jun), Elk-1 and nitric oxide synthase [13]. Aside the action mediated (hydroxyethyl)-rapamycin. The main difference between the two drugs
through calcineurin inhibition, CsA also increased transforming growth is the half-life, being 60 h for SRL and 22 h for EVL. Therefore, SRL is
factor β (TGF-β). IL-2 stimulated T lymphocytes are sensitive to TGF-β administered once a day and EVL every 12 h.
actions that reduces their proliferation, contributing to the immuno­ Both molecules are prodrugs that bind to FKBP12 (cytoplasmic
suppressive effect. However, this effect can also mediate renal intersti­ immunophilin), the same binding protein as for TAC. However, they
tial fibrosis and associated nephrotoxicity. Since both CNIs decrease IL-2 mediate an effect independent of calcineurin inhibition.
synthesis, it must be noted that they have an undesired inhibitory effect The SRL/EVL-FKBP complex binds to the m-TOR protein kinase. It
on the generation of T-reg cells, which are highly dependent on this mTOR is the catalytic subunit of two complexes named mTOR complex
cytokine for their differentiation and expansion [14]. (mTORC) 1 and mTORC2.
Cyclosporine A is soluble in lipids and administered mainly by oral While both are downstream mediators of the PI3K/AKT pathway,
and in limited cases by intravenous route. Oral absorption of CsA suffer mTORC1 is especially involved in regulation of cell growth, prolifera­
of a high degree of intra and inter- individual variability. The tion, and protein synthesis control and mTORC2 has main functions in
bioavailability is around 30%, therefore the intravenous dose recom­ cell survival, polarization and cell structure [24].
mended is 30-30% of oral dose. Classic CsA formulation needed the mTOR inhibitors mediate their action after forming a complex with
presence of bile for absorption. The CsA microemulsion (Neoral ®) im­ the FKBP12, which binds to mTOR with high affinity.
proves the oral bioavailability of the drug and increases drug exposure TORC1 is equally inhibited by SRL and EVL, while SRL has signifi­
by 34% (faster and higher peak, around 67%), decreasing the intra- cantly less effect on mTORC2.
individual variability and improving the correlation between CsA-Cmin The PI3/Akt/mTOR signaling cascade enhances cell proliferation
and actual exposure to the drug [15]. In clinical practice, pharmacoki­ mediated by several cytokines and growth factors: IL-2/IL-2 receptor
netic monitoring is performed by 12-h trough levels (C0) of CsA and TAC complex but also IL-11, G-CSF, insulin growth factor (IGF-1), VEGF,
in whole blood, but controversies exist on the optimal monitoring epidermal growth factor (EGF) and erythropoietin. Therefore, mTOR
strategy [16]. CsA metabolism takes place both in the liver and in in­ inhibitors block cell-cycle progression from G1 to S phase [25]. The lack
testinal epithelial cells through the cytochrome P450 (CYP3A). It is of an inhibition on the axis IL-2/IL-2 receptor might explain a beneficial
therefore highly susceptible to drug-to-drug interactions. effect on the T-reg population, that seem to be increased in mTORi
The most relevant side effect of CNIs is nephrotoxicity, that can be treated patients.
both acute-reversible and chronic associated to structural damage. A Importantly, these drugs are not nephrotoxic but may delay the re­
hemodynamic effect is due to decreased glomerular filtration rate by covery from acute tubular necrosis. Moreover, mTOR inhibitors may
vasoconstriction of both the afferent arteriole and efferent arteriole. This exacerbate CNI-related nephrotoxicity, therefore low doses of CNI seem
effect may depend on the activation of the renin-angiotensin- recommendable when used in conjunction with mTOR inhibitors. Also,
aldosterone system induced by endothelin. In addition, an endothelial regarding their impact on kidney function, a direct effect of mTOR in­
dysfunction associated to CNIs has been described, due to increased hibitors on renal podocytes is responsible for the appearance of pro­
thromboxane production and decreased synthesis of vasodilating agents teinuria for impaired integrity of the glomerular capillary wall.
such as nitric oxide and prostaglandin E2 [17]. Moreover, direct cyto­ In the field of solid organ transplantation, a possible anti-viral effect
toxicity on renal tubular cells can be exerted especially by the intrave­ of mTOR inhibitor has emerged, in fact in different cohorts of transplant
nous formulations due to the vehicle, polyoxyethylated castor oil, the recipients it has been observed a reduced incidence of Cytomegalovirus
Cremaphor, which also produces a decrease in renal plasma flow. Direct and Polyoma Virus infections in patients receiving those agents. It is not
CsA tubular toxicity has been shown in in vitro models [18]. fully explained the mechanism behind those observations. The most
The chronic non-reversible nephrotoxic effects are histologically interesting hypotheses are the increased CD8 memory T-cells response
represented by arteriolar hyalinosis, interstitial fibrosis, and tubular and functionality, the interference with the mTOR (PI3K-AkT) cascade,
atrophy that can be described as “striped” [19]. The underlying mech­ being this an anti-apoptotic signal necessary for viral survival and
anisms are the prolonged hemodynamic-ischemic effects, the increased transmission, and finally a direct antiviral effect by reduction of viral
levels of TGF-β1 and angiotensin II, inhibition of P-glycoprotein and protein synthesis [26].
induction of apoptosis in renal tubular epithelial cells [20,21]. Another important observation from clinical studies, is that patients
TAC differentiates from CsA in the pathway it interferes with but has under mTOR inhibitor treatment display lower incidence of neoplasms
the same final target. It has been shown to be superior to Cyclosporine A after solid organ transplantation. This effect possibly relies on the un­
in the prevention of kidney and liver allograft rejection and has a slightly controlled activation of the mTOR (AkT/PI3K) pathway in a large pro­
different spectrum of side effects (discussed in detail elsewhere). Among portion of malignancies. Additionally, mTOR inhibitors block
these, it must be noted that tacrolimus leads to increased glucose translation of some procarcinogenic mRNAs including VEGF (angio­
intolerance as compared to CsA [22]. genesis), cyclin D1 (cell cycle), and IL-10 (decreased Jak/STAT activity)
Apart from the classical formulations, extended-release preparations [27].
exist, allowing once-daily administration, possibly facilitating adher­ mTOR inhibitors are substrates of cytochrome P-450 (CYP3A4) and
ence. A recent MELTDose technology formulation has also been intro­ P-glycoprotein, therefore suffer from the same drug interference phe­
duced. This solid solution galenic formulation has a different nomena as CNIs.
pharmacokinetic profile and improved absorption, reducing the dose
from 1 to 0.7 in conversion studies. This formulation results in lower and Polyclonal biologic preparations
delayed Cmax, which results in a more sustained inhibition of calcineurin
activity [23]. Polyclonal ANTILYMPHOCYTE sera
The most widely used polyclonal anti-lymphocyte preparations are
Mammalian target of rapamycin (mTOR) inhibitors anti-thymocyte globulins (ATG). ATG are obtained from rabbits immu­
The inhibitors of mTOR approved as antirejection treatments in solid nized with human thymocytes (Thymoglobulin) or Jurkat cells (ATG-
organ transplantation are Rapamycin (Sirolimus) and Everolimus (EVL). Fresenius) and therefore contain a mix of antibodies directed against
Sirolimus (SRL) is a macrolide antibiotic produced by Streptomyces several lymphocyte surface molecules. Those sera have a depleting effect
hygroscopicus, an actinomycete originally isolated in Rapa Nui Island. on lymphocytes, mainly mediated by T-cell apoptosis in peripheral
This molecule has been first used as an antifungal, but after the obser­ lymphoid tissues by means of caspase-3 activation [28]. In addition,
vation of immunosuppressive effect it is now included in antirejection ATG down-modulates surface CD3, CD2, CD4 and CD8 molecules on the
maintenance schemes. EVL is an analogue of rapamycin, 40-O- remaining lymphocytes impairing T-cell functional responses. When

3
M. Meneghini et al. Best Practice & Research Clinical Gastroenterology 54-55 (2021) 101757

analyzing the composition of the polyclonal sera it has been possible to ANTI-IL-2R monoclonal antibodies
identify antibodies directed against adhesion molecules on lymphocytes, IL-2 receptor (IL-2R) is constituted of three sub-units not covalently
monocytes and neutrophils, and chemokine receptors on lymphocytes bound: IL-2Rα (CD25, 55 kDa), IL2Rβ (CD122, 70/75 kDa) and IL-2Rγ
and monocytes. All those mechanisms may have a role in reducing the (CD132, 64 kDa). While β and γ chains are constitutively expressed, the
ability of T cells to infiltrate graft tissues during ischemia reperfusion α chain is expressed only on activated T lymphocytes, conferring a high
injury and acute allograft rejection [29]. affinity for IL-2 and consequent T-cell clonal expansion driven by the
The result of ATG treatment is a profound CD4+ T-cell depletion, cytokine.
possibly in the long term. After depletion, the repopulation process takes Anti IL-2 receptor monoclonal antibodies (IL-2Ra) are directed
place, involving both naïve and memory T cells together with thymo­ against this α chain (CD25) of the IL2-R. The binding of IL2Ra to CD25
poiesis. The administration of ATG induces peripheral expansion and leads to internalization of IL2-R blocking clonal T cell expansion
new thymic emigration of T cells with a T regulatory (Treg) phenotype dependent of IL-2. A possible escaping pathway is activation because of
[30]. Additionally, functional studies indicate that ATG interfere with IL-7 or IL-15 pathways.
the functionality of dendritic cells (DC) reducing the capacity of mature Two forms of IL2-R antagonists exist: a first humanized form,
DC to stimulate allogenic and autologous T cells [31]. daclizumab is now out of use while the chimeric version, Basiliximab, is
Even if ATG administration often provokes the generation of anti­ currently used for induction treatment in solid organ transplantation.
bodies directed against rabbit immunoglobulins it has been reported Basiliximab has a high avidity for its target, the half-life is 8 days and
that a second course of rabbit ATG (rATG) results in similar lymphocyte standard dose is an administration of 20 mg on days 0 and 4 after
depletion and is as well tolerated as a first course [32]. transplantation. The discontinuation of production of daclizumab in
ATG are used both for induction in allotransplantation and in 2009 because of commercial reasons resulted in Basiliximab as the main
treatment of severe T-cell mediated rejection. IL-2Ra available in clinical practice. In the few studies comparing both
IL2R antagonists in the prevention of acute rejection, there were no
Intravenous immunoglobulins significant differences between them [36].
Intravenous immunoglobulins (IVIG) are a compound of plasma-
derived immunoglobulins from several human donors, conferring a Anti CD52 monocolonal antibody
wide immunological repertoire. IVIG have an immunomodulatory effect Alemtuzumab (Campath-1H) is a humanized monoclonal antibody
whose mechanisms of action are not completely known. They modulate directed against the pan-lymphocyte CD52 antigen present on human
expression and function of Fc receptors (common fragment of immu­ lymphoid and myeloid cells. It is a depleting antibody inducing a pro­
noglobulins), interfere with complement activity, interfere with cyto­ found and durable lymphopenia through a Fc-mediated mechanism,
kines action, inhibit T and B-cell activation and differentiation. In vitro, mediating antibody-dependent cell-mediated cytotoxicity of target cells.
it has been shown that IVIG administration in mixed lymphocyte reac­ Most B and T lymphocytes display high CD52 expression, but also the
tion results in reduced expression of CD40, CD86, Class II MHC, CD19 majority of NK cells, monocytes and macrophages, and a small per­
and adhesion molecules [33]. centage of granulocytes. It is not expressed on erythrocytes or hemato­
This compound can be used in solid organ transplantation for poietic stem cells [37].
treatment of antibody-mediated rejection as well as in autoimmune It is used as a depleting therapy in the induction treatment of solid
diseases. organ allotransplantation [38], as well as in autoimmune diseases.

Biologic agents, monoclonal antibodies or Fusion proteins ANTI-CD20 monoclonal antibody


The CD20 receptor is a transmembrane protein expressed on mature
Costimulation blockade B lymphocytes. Rituximab is a chimeric monoclonal antibody directed
After T cell receptor (TCR) interaction with MHC and antigenic against CD20, of the IgG1k subtype. It has a high affinity for CD20 and
peptide by antigen presenting cells (APC), the activation of cos­ mediates B-cell depletion through complement mediated toxicity as well
timulation pathways is the essential second step of T-cell activation. as antibody-mediated cytotoxicity by effector immune cells. The main
The only therapeutic agents currently available in clinical practice effect is observed in circulating B cells in peripheral blood, and to a
acting as costimulatory blockers are Abatacept and Belatacept. lesser extent in secondary lymphoid organs [39]. The consequent B cell
Both drugs are fusion proteins designed to block the CD28 ligands on depletion is fast (72 h) with long-lasting effect up to 12 months. It is
antigen presenting cells, CD80 and CD86 (B7). B7 molecules are hyper- usually not associated to hypogammaglobulinemia because plasma cells
expressed after APC activation and the interaction with CD28 results in are not targeted, since they do not express CD20. Importantly, since also
T-cell activation and proliferation, while its binding with the receptor regulatory B cells express CD20, Rituximab can impair the regulatory
CTLA-4, expressed by already activated T cells, leads to hypo-activation and anti-inflammatory functions of those cells producing IL-10. This
and reduced polyclonal expansion as negative regulation. might explain the disappointing results of Rituximab alone as induction
Abatacept and Belatacept are chimeric proteins CTLA-4 Ig with high therapy in allotransplantation or inflammatory bowel diseases, even
affinity for B7-1 (CD80) on the antigen presenting cells, resulting in a T- leading to exacerbations [40].
cell hyporesponsiveness against specific antigens. Moreover, some in It is used for desensitization for AB0 incompatible liver trans­
vitro studies demonstrated an additional direct effect on B cells (inde­ plantation, for the treatment of antibody mediated rejection after allo­
pendent from the inhibition of follicular T helper cells) impairing acti­ transplantation and to target B cells in antibody mediated autoimmune
vation and differentiation to antibody-secreting plasmablasts [34]. diseases such as autoimmune hepatitis, IgG4 mediated diseases, and
The difference between Abatacept and Belatacept consists of a primary biliary cholangitis [41].
change in 2 amino acids in the B7 binding region, resulting in 4-times Ofatumumab, a completely humanized second generation anti-CD20
higher affinity of Belatacept for CD80 and two times higher for CD86. monoclonal antibody binds to a different CD20 epitope than Rituximab
In vivo it leads to higher inhibition of T-cell activation up to 10 times, [42] and may limit adverse reactions and constitute a possible alterna­
therefore it is the only costimulation inhibitor drug approved for the tive in cases of intolerance to Rituximab.
prevention of kidney transplant rejection. Some experiences in liver
transplantation are available, being an appealing option to avoid CNI Other biologic agents
associated nephrotoxicity [35].
In recent years, a main advance in the field of immunosuppression
has been the generation of monoclonal antibodies targeting different

4
M. Meneghini et al. Best Practice & Research Clinical Gastroenterology 54-55 (2021) 101757

mediators of the inflammatory cascade. Those agents have been already Anti interleukin IL-12-23 (ustekinumab) monoclonal antibodies
incorporated as first-line treatment of different rheumatological diseases target two important cytokines involved in lymphocyte function. IL-12 is
and have potential use in many autoimmune mediated disorders, alone produced by antigen-presenting cells and is necessary for the develop­
or in combination with other immunosuppressive agents. ment of T cells with Th1 profile, secreting interferon-gamma. IL-23 is
Currently available biologic agents more widely used in clinical produced by antigen-presenting cells, monocytes, and macrophages and
practice are anti-tumor necrosis factor-alpha (TNF-α), anti-integrin and triggers the differentiation and activation of Th17 cells. Ustekinumab
anti-interleukin IL-12-23. targets a subunit (p40) common to the IL-12 and IL-23 receptors
Anti-Tumor necrosis factor α (TNF-α) agents (Infliximab, adalimu­ expressed on T cells and NK cells. It is a human monoclonal IgG1 anti­
mab, certolizumab, and golimumab) are monoclonal antibodies inhib­ body, of subcutaneous or intravenous administration [46].
iting interaction of the pleomorphic TNF-α cytokine with both soluble Tofacitinib belongs to the family of Janus kinase (JAK) inhibitors,
(sTNFR) and membrane bound (TNFR1 and TNFR2) receptors, therefore and initially considered as specific JAK-3 inhibitor. JAK are enzymes
blocking the inflammatory cascade including NK-kB. In addition, it has belonging to the Janus proteins family [47]. They belong to the group of
been shown that TNF-α inhibitors can induce apoptosis of TNF- tyrosin-kinase, involved in the proximal steps of signal transduction
α–producing immune cells, reducing further production and signaling from positive cytokine signals (IL-9, IL-12, IL-23 and IFN-γ) toward cell
amplification. Infliximab is a chimeric engineered IgG1 monoclonal nucleus. While Tofacitinib is non-selective, targeting the different JAK
antibody, Adalimumab and Golimumab are fully humanized IgG1 while isoforms has been explored, in particular JAK-3 transmits activation
Certolizumab is a PEGylated Fab fragment of humanized IgG1 mono­ signals mediated by the shared γ chain of IL-2, IL-4, IL-7, IL-9, IL-15 e
clonal antibody. Pegylation enhances antibody stability, half-life, and IL-21 receptors on NK cells, thymocytes and activated B and T cells,
bioavailability [43,44]. possibly limiting the effect to activated cells and not those in quiescent
The anti-integrin drugs (vedolizumab, natalizumab) act by blocking state.
the interaction between endothelial cells and adhesion molecules, a Fig. 1 shows the targets of the main immunosuppressants on T cells
limiting step for immune cells migration and tissue inflammation. and antigen presenting cells and their interactions.
Natalizumab blocks α4 integrin, necessary for white cells movement
toward target organs and migration of leukocytes to inflamed sites. Summary
Vedolizumab is a humanized anti-α4β7-integrin IgG1 monoclonal anti­
body. It acts by blocking α4β7-integrin, a gut-specific homing receptor In this review, the mechanisms of action of the main immunosup­
necessary for adhesion of leukocytes to the gut selectively [45]. pressants have been discussed. It should be noted that more than one

Fig. 1. Cell targets on T cells and antigen presenting cells and their interactions and immunosuppressive agents. Adapted from the book: Hernando Nefrología
Clinica, 5th edition, Panamericana.

5
M. Meneghini et al. Best Practice & Research Clinical Gastroenterology 54-55 (2021) 101757

agent can target the same molecular pathway at different levels and [15] Kovarik JM, Mueller EA, Niese D. Clinical development of a cyclosporine
microemulsion in transplantation. In: Therapeutic drug monitoring. Ther drug
some drugs can display significant interactions between them. It is
monit; 1996. p. 429–34.
important to know all those mechanisms when using and combining the [16] Schiff J, Cole E, Cantarovich M. Therapeutic monitoring of calcineurin inhibitors
available drugs in the clinical practice. Also, the mechanisms underlying for the nephrologist. Clin J Am Soc Nephrol Clin J Am Soc Nephrol 2007;2:374–84.
most relevant side effects should be considered when choosing treat­ [17] Lanese DM, Conger JD. Effects of endothelin receptor antagonist on cyclosporine-
induced vasoconstriction in isolated rat renal arterioles. J Clin Invest 1993;91(5):
ment schemes as well as for the design of more modern and specific 2144–9.
molecules. [18] Homan KA, Kolesky DB, Skylar-Scott MA, Herrmann J, Obuobi H, Moisan A, et al.
Bioprinting of 3D convoluted renal proximal tubules on perfusable chips. Sci Rep
2016 Oct 11;6(1):1–13.
Practice points [19] Ojo AO, Held PJ, Port FK, Wolfe RA, Leichtman AB, Young EW, et al. Chronic renal
failure after transplantation of a nonrenal organ. N Engl J Med 2003 Sep 4;349(10):
931–40.
- The development of effective immunosuppressive agents has [20] Han SW, Li C, Ahn KO, Lim SW, Song HG, Jang YS, et al. Prolonged endoplasmic
improved the control of immune-mediate injury in allo­ reticulum stress induces apoptotic cell death in an experimental model of chronic
transplantation and autoimmunity cyclosporine nephropathy. Am J Nephrol 2008 Sep;28(5):707–14.
[21] Farouk SS, Rein JL. The many faces of calcineurin inhibitor toxicity—what the FK?
- Intrinsic toxicity and side effects limit the use of immunosuppressant Adv Chron Kidney Dis 2020 Jan 1;27(1):56–66.
agents especially in chronic regimens [22] Chakkera HA, Kudva Y, Kaplan B. Calcineurin inhibitors: pharmacologic
- Comprehension of biological mechanisms underlying alloimmunity mechanisms impacting both insulin resistance and insulin secretion leading to
glucose dysregulation and diabetes mellitus. Clinical Pharmacology and
and alloimmune processes can identify new molecular targets or
Therapeutics. Nature Publishing Group 2017;101:114–20.
uncover the potential use of other agents developed in oncology or [23] Fontova P, Colom H, Rigo-Bonnin R, Bestard O, Vidal-Alabró A, van
rheumatology. Merendonk LN, et al. Sustained inhibition of calcineurin activity with a melt-dose
once-daily tacrolimus formulation in renal transplant recipients. Clin Pharmacol
Ther 2021.
Research agenda [24] Fantus D, Rogers NM, Grahammer F, Huber TB, Thomson AW. Roles of mTOR
complexes in the kidney: implications for renal disease and transplantation. Nature
Reviews Nephrology. Nature Publishing Group 2016;12:587–609.
- Modern technologies permit a deeper knowledge of the mechanisms [25] Ponticelli C. Can mTOR inhibitors reduce the risk of late kidney allograft failure?
underlying therapeutic and toxic effects of immunosuppressive Transpl Int 2008;21:2–10.
agents and can be used to develop new molecules with more specific [26] Bak S, Tischer S, Dragon A, Ravens S, Pape L, Koenecke C, et al. Selective effects of
mTOR inhibitor Sirolimus on naïve and CMV-specific T cells extending its
molecular targets. applicable range beyond immunosuppression. Front Immunol 2018;9:2953.
- The development of new molecules with more specific targets to [27] Bhat M, Watt KD. Mammalian target of rapamycin inhibition after solid organ
control immune responses without redundant effects is highly transplantation: can it, and does it, reduce cancer risk? Clin Transplant 2015 Jul 1;
29(7):654–63.
recommendable. [28] Ashok kumar C, Sun Q, Ningappa M, Higgs BW, Mazariegos G, Zeevi A, et al.
Antithymocyte globulin facilitates alloreactive T-cell apoptosis by means of
caspase-3: potential implications for monitoring rejection-free outcomes.
Transplantation 2015 Jan 15;99(1):164–70.
Declaration of competing interest [29] Préville X, Flacher M, LeMauff B, Beauchard S, Davelu P, Tiollier J, et al.
Mechanisms involved in antithymocyte globulin immunosuppressive activity in a
None. nonhuman primate model. Transplantation 2001 Feb 15;71(3):460–8.
[30] Bouvy AP, Klepper M, Kho MML, Boer K, Betjes MGH, Weimar W, et al. The impact
of induction therapy on the homeostasis and function of regulatory T cells in
References kidney transplant patients. Nephrol Dial Transplant 2014;29(8):1587–97.
[31] Naujokat C, Berges C, Fuchs D, Sadeghi M, Opelz G, Daniel V. Antithymocyte
globulins suppress dendritic cell function by multiple mechanisms. Transplantation
[1] Buttgereit F, Da Silva JPA, Boers M, Burmester GR, Cutolo M, Jacobs J, et al.
2007 Feb;83(4):485–97.
Standardised nomenclature for glucocorticoid dosages and glucocorticoid
[32] Rodríguez-Reimundes E, Buron F, Chauvet C, Daoud S, Thaunat O, Brunet M, et al.
treatment regimens: current questions and tentative answers in rheumatology. Ann
Retreatment by antithymocyte globulin for second kidney transplantation:
Rheum Dis 2002;61(8):718–22.
Efficacy, tolerance and safety. Transpl Immunol 2013 Jan;28(1):6–8.
[2] De Bosscher K, Vanden Berghe W, Haegeman G. The interplay between the
[33] Jordan SC, Toyoda M, Kahwaji J, Vo AA. Clinical aspects of intravenous
glucocorticoid receptor and nuclear factor-κB or activator protein-1: molecular
immunoglobulin use in solid organ transplant recipients. American Journal of
mechanisms for gene repression. Endocr Rev Endocr Rev 2003;24:488–522.
Transplantation. Am J Transplant; 2011;11:196–202.
[3] Necela BM, Cidlowski JA. Mechanisms of glucocorticoid receptor action in
[34] Leibler C, Thiolat A, Hénique C, Samson C, Pilon C, Tamagne M, et al. Control of
noninflammatory and inflammatory cells. Proc Am Thorac Soc Proc Am Thorac Soc
humoral response in renal transplantation by Belatacept depends on a direct effect
2004;1:239–46.
on B cells and impaired T follicular helper-B cell crosstalk. J Am Soc Nephrol 2018
[4] Ramamoorthy S, Cidlowski JA. Corticosteroids. Mechanisms of action in health and
Mar 10;29(3):1049–62.
disease. Rheumatic Disease Clinics of North America. W.B. Saunders 2016;42:
[35] Perez CP, Patel N, Mardis CR, Meadows HB, Taber DJ, Pilch NA. Belatacept in solid
15–31.
organ transplant: review of current literature across transplant types.
[5] Boldizsar F, Talaber G, Szabo M, Bartis D, Palinkas L, Nemeth P, et al. Emerging
Transplantation. Lippincott Williams and Wilkins 2018;102:1440–52.
pathways of non-genomic glucocorticoid (GC) signalling in T cells.
[36] Kandus A, Arnol M, Omahen K, Oblak M, Vidan-Jeras B, Kmetec A, et al.
Immunobiology. Urban & Fischer 2010;215:521–6.
Basiliximab versus daclizumab combined with triple immunosuppression in
[6] Anstey A, Lear JT. Azathioprine: clinical pharmacology and current indications in
deceased donor renal transplantation: a prospective, randomized study.
autoimmune disorders. BioDrugs BioDrugs 1998;9:33–47.
Transplantation 2010 Apr;89(8):1022–7.
[7] Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action.
[37] Waldmann H. A personal history of the CAMPATH-1H antibody. SUPPL Med Oncol
Immunopharmacology 2000;47(2–3):85–118.
2002;19.
[8] Villarroel MC, Hidalgo M, Jimeno A. Mycophenolate mofetil: an update. Drugs
[38] Dhesi S, Boland B, Colquhoun S. Alemtuzumab and liver transplantation: a review.
Today (Barc) Drugs Today 2009;45:521–32.
Curr Opin Organ Transplant Curr Opin Organ Transplant 2009;14:245–9.
[9] Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacodynamics of
[39] Abulayha A, Bredan A, El Enshasy H, Daniels I. Rituximab: modes of action,
mycophenolate in solid organ transplant recipients. Clin Pharmacokinet Clin
remaining dispute and future perspective. Future Medicine Ltd. Future Oncol 2014;
Pharmacokinet 2007;46:13–58.
10:2481–92.
[10] Sanford M, Keating GM. Enteric-coated mycophenolate sodium: a review of its use
[40] Shahmohammadi S, Sahraian MA, Shahmohammadi A, Doosti R, Zare-Mirzaie A,
in the prevention of renal transplant rejection. Drugs Drugs 2008;68:2505–33.
Naser Moghadasi A. A presentation of ulcerative colitis after rituximab therapy in a
[11] Mantzaris GJ. Thiopurines and methotrexate use in IBD patients in a biologic Era.
patient with multiple sclerosis and literature review. Elsevier B.V. Multiple
Curr Treat Options Gastroenterol 2017 Mar;15(1):84–104.
Sclerosis and Related Disorders 2018;22:22–6.
[12] Azzi JR, Sayegh MH, Mallat SG. Calcineurin inhibitors: 40 Years Later, can’t live
[41] Cargill T, Culver EL. The role of B cells and B cell therapies in immune-mediated
without. J Immunol 2013 Dec 15;191(12):5785–91.
liver diseases. Frontiers Media S.A. Front Immunol 2021;12:1212.
[13] Shi YY, Hesselink DA, van Gelder T. Pharmacokinetics and pharmacodynamics of
[42] Klein C, Lammens A, Schäfer W, Georges G, Schwaiger M, Mössner E, et al. Epitope
immunosuppressive drugs in elderly kidney transplant recipients. Transplantation
interactions of monoclonal antibodies targeting CD20 and their relationship to
Reviews. W.B. Saunders 2015;29:224–30.
functional properties. mAbs 2013 Jan;5(1):22–33.
[14] Miroux C, Moralès O, Carpentier A, Dharancy S, Conti F, Boleslowski E, et al.
[43] Nielsen OH, Ainsworth MA. Tumor necrosis factor inhibitors for inflammatory
Inhibitory effects of cyclosporine on human regulatory T cells in vitro. Transplant
bowel disease. N Engl J Med 2013 Aug 22;369(8):754–62.
Proc 2009;41(8):3371–4.

6
M. Meneghini et al. Best Practice & Research Clinical Gastroenterology 54-55 (2021) 101757

[44] Costantino G, della Torre A, Lo Presti MA, Caruso R, Mazzon E, Fries W. Treatment [46] Jefremow A, Neurath MF. All are equal, some are more equal: targeting IL 12 and
of life-threatening type I refractory coeliac disease with long-term infliximab. Dig 23 in ibd – a clinical perspective. ImmunoTargets Ther 2020 Nov;9:289–97.
Liver Dis 2008 Jan;40(1):74–7. [47] Salas A, Hernandez-Rocha C, Duijvestein M, Faubion W, McGovern D, Vermeire S,
[45] Zundler S, Becker E, Schulze LL, Neurath MF. Immune cell trafficking and retention et al. JAK–STAT pathway targeting for the treatment of inflammatory bowel
in inflammatory bowel disease: mechanistic insights and therapeutic advances. Gut disease. Nature Research Nat Rev Gastroenterol Hepatol 2020;17:323–37.
2019 Sep;68(9):1688–700.

You might also like