Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2020 Mar 12;177(10):2199–2214. doi: 10.1111/bph.15011

Old dog, new trick: Trivalent arsenic as an immunomodulatory drug

Yishan Ye 1,2, Béatrice Gaugler 1,3, Mohamad Mohty 1,3, Florent Malard 1,3,
PMCID: PMC7174890  PMID: 32022256

Abstract

Trivalent arsenic (As(III)) is recently found to be an immunomodulatory agent. As(III) has therapeutic potential in several autoimmune and inflammatory diseases in vivo. In vitro, it selectively induces apoptosis of immune cells due to different sensitivity. At a non‐toxic level, As(III) shows its multifaceted nature by inducing either pro‐ or anti‐inflammatory functions of immune subsets. These effects are exerted by either As(III)–protein interactions or as a consequence of As(III)‐induced homeostasis imbalance. The immunomodulatory properties also show synergistic effects of As(III) with cancer immunotherapy. In this review, we summarize the immunomodulatory effects of As(III), focusing on the effects of As(III) on immune subsets in vitro, on mouse models of immune‐related diseases, and the role of As(III) in cancer immunotherapy. Updates of the mechanisms of action, the pioneer clinical trials, dosing, and adverse events of therapeutic As(III) are also provided.


Abbreviations

APL

acute promyelocytic leukaemia

As(III)

trivalent arsenic

ATL

adult T‐cell leukaemia/lymphoma

ATRA

all‐trans retinoic acid autoimmune disease

cDC

conventional dendritic cells

CIK

cytokine‐induced killer

iAs

inorganic arsenic compounds

LAK

lymphokine‐activated killer

MDDCs

monocyte‐derived dendritic cells

Nrf2

nuclear factor erythroid 2‐related factor 2

pDCs

plasmacytoid dendritic cells

PML/RARα

promyelocytic leukaemia‐retinoic acid receptor‐α

SLE

systemic lupus erythematosus

1. INTRODUCTION

Inorganic arsenic compounds (iAs) have been used in traditional Chinese and Western medicine for over 2400 years (Emadi & Gore, 2010). Arsenic trioxide (As2O3) was rediscovered in the 1970s in the treatment of acute promyelocytic leukaemia (APL) with striking efficacy and good safety profile (Wang & Chen, 2008 ). So far, As2O3, together with all‐trans retinoic acid (ATRA), has revolutionized the treatment of APL: The outcome changed from 35% to 45% long‐term overall survival by chemotherapy to a complete remission rate of over 95% and a long‐term survival rate up to 90% with a chemotherapy free approach (Cicconi & Lo‐Coco, 2016; Lo‐Coco et al., 2013). Furthermore, As2O3 has also shown promising results in many other malignancies such as adult T‐cell leukaemia/lymphoma (ATL), and NPM1 mutant acute myeloid leukaemia (El Hajj et al., 2015; Kchour et al., 2009).

Modulation of immunity by iAs has been studied and put in clinical use with trivalent arsenicals (As(III)) including As2O3 and sodium arsenite (NaAsO2). As(III) has high affinity for sulfhydryl groups and can bind to cysteine residues in peptides and proteins, leading to protein conformational change and malfunction (Shen, Li, Cullen, Weinfeld, & Le, 2013). As(III) is also a potent ROS inducer (Chou et al., 2004). Moreover, many key proteins are indirectly regulated by As(III) through post‐translational regulations such as sumoylation (Lallemand‐Breitenbach et al., 2001; mechanisms reviewed by Shen et al., 2013).

The iAs are regarded as having general toxic effects on the immune system (Dangleben, Skibola, & Smith, 2013). However, in vitro studies have provided a more detailed picture, revealing the multifaceted effects of As(III) on different immune subsets (Baysan, Yel, Gollapudi, Su, & Gupta, 2007; Gupta et al., 2003). Recent reports on animal models have shown the efficacy of As(III) on several autoimmune and inflammatory diseases (Bobe, Bonardelle, Benihoud, Opolon, & Chelbi‐Alix, 2006; Kavian, Marut, Servettaz, Laude, et al., 2012). In addition, in the era of cancer immunotherapy, emerging evidence indicates As(III) as a promising immunoadjuvant for haematological malignancies and solid tumours (Deaglio et al., 2001; Thomas‐Schoemann et al., 2012). However, during As2O3 treatment, several typical adverse events including cardiac and liver toxicities have been reported (Sanz et al., 2019). These adverse events, even though manageable, must be taken into account by medical practitioners.

In this review, in order to promote the better use of this double‐edged sword in immunotherapy, we summarize the mechanisms of action of As(III), the modulation on each immune cell subset, the use of As(III) on preclinical mouse models of immune‐mediated diseases, and the pioneer studies of As(III) as an adjuvant for tumour immunotherapy. We also summarize the standard dosing and therapeutic schedules of As(III) and highlight the possible short‐ and long‐term adverse events that require vigilance by clinicians.

2. PHARMACOLOGICAL MECHANISMS OF ACTION

2.1. As(III) biochemistry

Both As2O3 and NaAsO2 transform to As(OH)3 in aqueous solution. After being taken up by cells through the https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=119#696 transmembrane protein, As(III) exerts its biochemical effects intracellularly (Leung, Pang, Yuen, Kwong, & Tse, 2007). Interaction with the thiol (or sulfhydryl) groups (–SH) of proteins with a high cysteine content constitutes the basic biochemical reaction of As(III) (Emadi & Gore, 2010), which alters protein conformation, resulting in loss of their function, and affects their recruitment and interaction with other proteins and DNA (Shen et al., 2013). Apart from the direct arsenic‐protein binding, recent studies revealed that many key proteins are modulated through more complicated post‐translational stepwise regulations (de Thé, 2018).

2.2. PML and PML nuclear body regulation

It was initially found that the APL promoter, promyelocytic leukaemia‐retinoic acid receptor‐α (PML/https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=590) was especially sensitive to As2O3‐induced degradation, which was the critical step of disease eradication (Chen et al., 1996). The subsequent comprehensive studies by Lallemand‐Breitenbach and de Thé (2018) revealed that As2O3 specifically targets the PML moiety. The PML protein forms nuclear bodies (NBs),which recruit and sumoylate dozens of partner proteins, leading to a variety of biological processes (Lallemand‐Breitenbach et al., 2001; Sahin et al., 2014). Notably, As2O3‐induced oxidative stress could enhance formation of PML NBs, leading to p53 activation in vivo in normal mice (Niwa‐Kawakita et al., 2017).

In APL, through both ROS production and direct binding, As2O3 exerts its dual‐targeting effects (Jeanne et al., 2010). On one hand, As2O3 induces PML/RARα sumoylation, proteasomal degradation, and APL cell differentiation (Fasci, Anania, Lill, & Salvesen, 2015; Lallemand‐Breitenbach & de Thé, 2018). On the other hand, As2O3 targets the wild‐type PML proteins, leading to re‐formation of PML NBs, subsequent p53 activation, and ultimately APL clearance (Ablain et al., 2014; Niwa‐Kawakita et al., 2017). Also, in ATL, As2O3, together with https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=957 induced complete remission of disease, both in mice and in humans, through degradation of the disease driver oncoprotein Tax (El Hajj et al., 2010; El‐Sabban et al., 2000; Kchour et al., 2009). Interestingly, this process was also mediated through a As2O3 enforcement of PML NBs formation, Tax sumoylation, and proteasomal degradation (Dassouki et al., 2015). In addition, the As2O3/ATRA combination significantly decreased NPM1‐mutant acute myeloid leukaemia blasts in patients though oxidative stress generation, p53 activation, and ultimately mutant NPM1 degradation (El Hajj et al., 2015; Martelli et al., 2015).

Apart from tumour suppression, PML and PML NBs were recently found to play key role in mediating innate immune responses (Hsu & Kao, 2018; Lallemand‐Breitenbach et al., 2001; Scherer & Stamminger, 2016). Studies have identified PML as a direct, positive regulator of IFN type I signalling (Kim & Ahn, 2015) and is implicated in the regulation of and extended spectrum of cytokines such as the pro‐inflammatory https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4974 and https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4998 (Lo et al., 2013; Scherer & Stamminger, 2016). Thus, it is important to find out if modulation of PML and PML NBs could explain the effectiveness of As2O3 in autoimmune and inflammatory diseases with a type I IFN signature and abnormal cytokine profile.

2.3. Apoptosis induction

As(III) can induce immune cell apoptosis through both the mitochondrial‐mediated and the receptor‐mediated pathways (Gupta et al., 2003; Yu, Chen, Liao, Chang, & Yu, 2002). The pro‐apoptotic mechanisms include generation of oxidative stress, caspase activation, alteration of the https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=2844 family proteins, and up‐regulation/down‐regulation of several survival‐related signalling pathways such as NF‐κB, https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=289/https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=519, and https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=1870 apoptotic signalling pathways (Gupta et al., 2003; Lemarie, Morzadec, Merino et al., 2006; Lemarie, Morzadec, Bourdonnay, Fardel, & Vernhet, 2006; Yu et al., 2002). Within its therapeutic range, As(III) is able to induce apoptosis of specific types of sensitive immune cells (Bobe et al., 2006; Thomas‐Schoemann et al., 2012). In a mouse model of colon cancer, regulatory T cells are preferentially depleted, leading to immunomodulatory effects (Thomas‐Schoemann et al., 2012). The in vitro EC50 of As(III) on different immune subsets and the possible mechanisms of apoptosis induction are summarized in Table 1.

TABLE 1.

Trivalent arsenic induces immune cell apoptosis: sensitivity and mechanisms of action

Cell subtype Species Type of As(III) EC50 (culture time) Possible mechanism References
PBMC Human NaAsO2 5 μmol·L−1 (48 and 72 hr) N/A (Yu et al., 2002)
Neutrophil Human As2O3 <5 μmol·L−1 (22 hr)
  1. H2O2 generation

  2. Activation of caspases

  3. De novo protein synthesis

  4. Syk kinase activation

(Antoine et al., 2010; Binet et al., 2006)
Monocyte
Monocyte (during macrophagic differentiation with GM‐CSF or M‐CSF) Human As2O3 ~1 μmol·L−1 (6 days)
  1. Inhibition of NF‐κB‐related survival pathways

(Lemarie, Morzadec, Merino, et al., 2006)
Promonocytic U937 cells Human As2O3 >4 μmol·L−1 (4 days)
Dendritic cell (DC)
Monocyte‐derived DCs Human NaAsO2 >2 μmol·L−1 (6 days) N/A (Macoch et al., 2013)
T cell
Primary CD4+ T cell Human As2O3 >5 μmol·L−1 (48 hr)
  1. ROS generation

(Gupta et al., 2003)
Primary CD8+ T cell Human As2O3 >5 μmol·L−1 (48 hr) 2. Regulation of the Bcl‐2 family proteins
CD4+ T cell within PBMCs Human NaAsO2 N/A
  1. TNF‐R1 apoptotic signalling

(Yu et al., 2002)
CD4+CD25+ Treg Human As2O3 ~1 μmol·L−1 (38 h)
  1. ROS and RNS accumulation

  2. High sensitivity to oxydative stress

(Thomas‐Schoemann et al., 2012)
CD4+CD25 effector T cell Human As2O3 ~2.5 μmol·L−1 (38 hr) N/A (Thomas‐Schoemann et al., 2012)
B cell
Ramos cell line Human As2O3 >10 μmol·L−1 (24 hr)
  1. ROS generation

  2. Up‐regulation of Bax and Bim expression

(Baysan et al., 2007)

Abbreviations: As2O3, arsenic trioxide; As(III), trivalent arsenic; EC50, half maximal effective concentration; H2O2, hydrogen peroxide; M‐CSF, macrophage colony‐stimulating factor; NaAsO2, sodium arsenite; PBMC, peripheral blood mononuclear cell; RNS, reactive nitrogen species.

2.4. ROS

ROS accumulation leads to oxidative stress, causing damage to nucleic acids, proteins, and lipids, which can lead to changes in cell signalling, and, ultimately, apoptosis (Schieber & Chandel, 2014). As(III) induces immune cell intracellular ROS accumulation via inhibition of antioxidants such as https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=6737 GSH peroxidase, and GSH reductase (Gupta et al., 2003; Singh, Kumar, Lal, Raisuddin, & Sahu, 2010) and activation of enzymes generating ROS such as https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=993 (Lemarie, Bourdonnay, Morzadec, Fardel, & Vernhet, 2008). As a consequence, expressions of redox‐sensitive genes such as HMOX1, NQO1, and GCLM are up‐regulated (Bourdonnay, Morzadec, Fardel, & Vernhet, 2009). Especially, nuclear factor erythroid 2‐related factor 2 (Nrf2), a stress‐activated transcription factor responsible for inducing a battery of cytoprotective genes, is activated by As(III) within mouse splenocytes, human T cells, human primary macrophages, and human monocyte‐derived dendritic cells (MDDCs; Bourdonnay, Morzadec, Fardel, & Vernhet, 2009; Duan et al., 2017; Macoch et al., 2015; Morzadec et al., 2014). The Nrf2 modulation is probably due to As(III)‐induced Nrf2 sumoylation, recruitment to the PML NBs, and degradation by RNF4‐mediated proteolysis (Malloy et al., 2013).

2.5. Signalling pathway regulation

Due to the different affinity of different proteins to As(III), and the potential post‐translational modulation by other structures such as PML NBs, As(III) has the potential to selectively target specific signalling pathways. In macrophages, for example, non‐toxic concentrations of As2O3 induce up‐regulation of 32 and depression of 91 genes respectively, indicating a global multi‐directional change of the cell‐signalling (Bourdonnay et al., 2009). Moreover, As(III) may inactivate up to 200 enzymes, and many are crucial regulators of important signalling pathways (Shen et al., 2013). As(III) inhibits IĸB kinase, whose integrity is key to the activation of the NF‐κB pathway (Lemarie, Morzadec, Merino, et al., 2006). Other pathways affected include the Rho‐kinase/p38‐kinase pathway and https://www.guidetopharmacology.org/GRAC/FamilyDisplayForward?familyId=518 (Binet & Girard, 2008; Lemarie et al., 2008) which will be described in detail in the next section.

3. THE MULTIFACETED EFFECTS OF TRIVALENT ARSENIC ON IMMUNE CELLS

3.1. Systemic immunomodulation

Chronic arsenic exposure leads to systemic immunosuppression (Dangleben et al., 2013). However, clinical use of As2O3 has shown a good safety profile, with limited side effects (Shen et al., 1997). No long‐term, treatment‐related, immune‐mediated disease or tumourigenesis is reported. During the As2O3 single‐agent treatment for APL, there are inhibitory effects on haematopoietic progenitor cells, and it takes 145 and 265 days for circulating T and B cells and 655 days for NK cells to achieve the median normal levels (Alex et al., 2018). The detailed effects of As(III) on each immune subset are described in the following sections.

3.2. Trivalent arsenic and granulocytes

The multifaceted effects of As(III) on different subsets of immnuocytes are summarized in Figure 1. High concentrations of As2O3 induce apoptosis of human neutrophils via generation of H2O2, de novo protein synthesis, caspase activation, and https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=2230 activation (Antoine, Ennaciri, & Girard, 2010; Binet, Cavalli, Moisan, & Girard, 2006). The As2O3‐induced de novo synthesized proteins include annexin‐1 and heat shock proteins (Binet, Chiasson, & Girard, 2008). Moreover, As2O3 induces endoplasmic reticulum stress within human neutrophils, which elicits either self‐protective mechanisms via the activation of unfolded protein response (UPR) or cell apoptosis, independent of caspase‐4 activation (Binet, Chiasson, & Girard, 2010a; Binet, Chiasson, & Girard, 2010b). Importantly, As2O3 recruits the MAPKs, activates p38 and JNK, and ultimately enhances the major neutrophil functions including adhesion, migration, degranulation, and phagocytosis of opsonized sheep red blood cells (Binet & Girard, 2008). Syk activation also helps in this agonistic process (Antoine et al., 2010). In addition, NaAsO2 promotes the formation of neutrophil extracellular traps, which play an important role in defence against pathogen infection (Wei et al., 2018). Moreover, in an acute As2O3‐exposed mouse model the neutrophil cell numbers were increased in the broncho‐alveolar lavage fluid (BALF; Li et al., 2017). Collectively, neutrophils are recruited and functionally enhanced during As(III) treatment, leading to a pro‐inflammatory response.

FIGURE 1.

FIGURE 1

Diverse functions of As(III) on immune cells. Compounds comprising trivalent arsenic (As(III)) can drive both promoting (top of figure in green) and suppressive (bottom of figure in blue) immune responses in different immune cell subsets

For eosinophils, in a mouse model of asthma, eosinophil recruitment and a reduction of chemotaxis level in BALF were detected after As2O3 treatment (Chu et al., 2010; Zhou et al., 2006). Therefore, eosinophil functions may be impaired during As(III) exposure. For basophils, the direct effects of As(III) have not yet been investigated. However, activation of basophils may be impaired in vivo during As(III) administration, because levels of IgE, an important driver of basophil activation, were decreased in the BALF of As2O3‐treated mice (Chu et al., 2010; Zhou et al., 2006). For mast cells, it was reported that NaAsO2 inhibits anti‐IgE stimulated degranulation via suppression of early tyrosine phosphorylation (Hutchinson et al., 2011). Therefore, As(III) may impair mast cell activation, especially that during the process of allergy (Shim et al., 2016).

3.3. Trivalent arsenic and monocytes/macrophages

Recent studies have shown that monocytes are not simply macrophage precursors but can acquire inflammatory effector functions, distinct from those of macrophages. (Guilliams, Mildner, & Yona, 2018). As2O3 induces human monocyte apoptosis during macrophage differentiation through down‐regulation of the NF‐κB‐related pathway (Lemarie, Morzadec, Merino, et al., 2006). Moreover, As2O3 increases LPS‐dependent expression of the inflammatory IL‐8 gene, by stimulating a redox‐sensitive pathway that strengthens p38‐kinase activation (Bourdonnay, Morzadec, Fardel, & Vernhet, 2011).

For macrophages, high concentration of As2O3 induces cell apoptosis through a mitochondrial‐dependent pathway (Sengupta & Bishayi, 2002; Srivastava et al., 2016). Also, it changes human monocyte‐derived macrophage morphology, reduces their adhesive capacity, decreases macrophagic surface marker expressions and impairs phagocytosis of E. coli in vitro (Lemarie, Morzadec, Bourdonnay, et al., 2006). Interestingly, As2O3 potentiates macrophage secretion of inflammatory https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=5074, IL‐1α, and https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4382 and the induction of allogeneic or autologous T‐cell responses (Lemarie et al., 2008; Sakurai, Ohta, & Fujiwara, 2005). These in vitro observations were confirmed by ex vivo data (Banerjee et al., 2009; Bishayi & Sengupta, 2003). The functional changes are probably due to superoxide generation, activation of the Rho‐kinase/p38‐kinase pathway (Lemarie, Morzadec, Merino, et al., 2006), and modulation of the UPR signalling (Srivastava et al., 2013). Especially, activating transcription factor 4 protein, a UPR transcription factor, plays a key role in As2O3‐mediated regulation of macrophage functions (Srivastava et al., 2016). In addition, As2O3 also globally regulates redox‐sensitive gene expression in human macrophages (Bourdonnay, Morzadec, Fardel, & Vernhet, 2009; Bourdonnay, Morzadec, Sparfel, et al., 2009). Therefore, As(III) exerts dual effects on macrophages by impairing their clearance capacity, while enhancing their pro‐inflammatory functions.

3.4. Trivalent arsenic and dendritic cells

Two types of dendritic cells (DCs), namely, conventional dendritic cells (cDC) and plasmacytoid dendritic cells (pDCs), can be distinguished in vivo, even though MDDCs are widely used for research in vitro, because they are more readily available. NaAsO2 inhibits dendritic differentiation of monocytes in vitro (Bahari & Salmani, 2017). It also decreases MDDCs viability, maturation, phagocytic capacity, and their ability to secrete the pro‐inflammatory cytokines https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4977 and https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4978 and to stimulate T helper (Th) cells to secrete https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4968 (Macoch, Morzadec, Fardel, & Vernhet, 2013). The inhibition of IL‐12 is mediated by the induced expression of Nrf2 (Malloy et al., 2013). Moreover, As2O3 inhibits IFN‐α secretion by murine pDCs (Kavian, Marut, Servettaz, Laude, et al., 2012).

3.5. Trivalent arsenic and T cells

T cells play a central role in https://en.wikipedia.org/wiki/Cell-mediated_immunity. High concentrations of As(III) induce apoptosis of T helper cells (CD4+ T cell), cytotoxic T cells (CD8+ T cell), and regulatory T cells (Treg; Gupta et al., 2003; Tenorio & Saavedra, 2005; Thomas‐Schoemann et al., 2012). Both mitochondrial‐mediated and the receptor‐mediated pathways are involved in T‐cell apoptosis. Gupta et al. (2003) revealed that As2O3 induces apoptosis of CD4+ and CD8+ T cells via the mitochondrial pathway by enhancing the generation of oxidative stress and by regulating the expression of Bcl‐2 family proteins. When CD4+ T cells were investigated within human peripheral blood mononuclear cells, TNF‐α released from other mononuclear cells after NaAsO2 exposure induced CD4+ T‐cell apoptosis through TNFR1 apoptotic signalling pathway (Yu et al., 2002).

There is different sensitivity to As(III) toxicity among the T‐cell subsets. CD4+ are more sensitive than CD8+ T cells to the pro‐apoptotic toxicity of NaAsO2, as demonstrated both in vitro and in vivo in human and mouse model (Duan et al., 2017; Vega, Montes de Oca, Saavedra, & Ostrosky‐Wegman, 2004; Yu et al., 2002). This difference leads to a reduced CD4+/CD8+ ratio, which is associated with immune dysfunction (Lu et al., 2015). Tregs are important CD4+ T cells mediating immune tolerance. in vitro tests show that As2O3, at the same concentration, preferentially induces apoptosis of human purified CD4+CD25+ Tregs than CD4+CD25 effector T cells and decreases the Treg frequency in APL patients' peripheral blood (Xu et al., 2018). As2O3‐mediated selective Treg depletion was also reported in vivo in a mouse model (Thomas‐Schoemann et al., 2012). As2O3 induces Treg apoptosis through ROS and reactive nitrogen species generation, and the differential effect of As2O3 on Treg versus other CD4+ cells may be related to differences in the cells' redox status (Thomas‐Schoemann et al., 2012). However, some other studies reported opposite results (Tohyama, Tanaka, Onda, Sugiyama, & Hirano, 2013; Zhao, Yang, et al., 2018). After exposure to NaAsO2, the mRNA level of the Treg specific transcription factor Foxp3 is up‐regulated in the spleen and thymus of the treated mice (Duan et al., 2017). Interestingly, Tohyama et al. (2013) reported that in mitogen‐activated human peripheral blood mononuclear cell, 5‐μM As2O3 decreases the Treg frequency after treatment for 48 hr and, conversely, increases its frequency after 96 hr of culture. These results indicate that short‐term exposure to As(III) may deplete Treg, in contrast to long‐term exposure leading to Treg increase.

Upon activation, naïve CD4+ T cells proliferate and differentiate into fully functional effector T cells, which lead to either inflammatory responses or immune suppression (Wan & Flavell, 2009). It is reported that As(III) exposure inhibits the proliferation of both human‐ and murine‐activated T cells, by affecting the initial activation step of T‐cell receptor signalling, and by inhibiting IL‐2 expression, at both the protein and mRNA levels (Conde, Acosta‐Saavedra, Goytia‐Acevedo, & Calderon‐Aranda, 2007; Morzadec, Bouezzedine, Macoch, Fardel, & Vernhet, 2012; Soto‐Pena & Vega, 2008; Vega et al., 1999). In addition, NaAsO2 blocks T cells in the G1 phase and, thus, delays the entry into cell cycle (Galicia, Leyva, Tenorio, Ostrosky‐Wegman, & Saavedra, 2003). More importantly, As(III) significantly alters Th cell differentiation. As2O3 inhibits IFN‐γ expression, the characteristic Th1 cytokine, at both mRNA and protein levels during the activation with anti‐CD3/anti‐CD28 of both human and murine T cells (Duan et al., 2017; VanDenBerg et al., 2017). However, As(III) does not change and sometimes increases the secretion of https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4996 and https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4980, the characteristic Th2 cytokines, from activated T cells (Galicia et al., 2003; Soto‐Pena & Vega, 2008). These observations indicate that As(III) probably alters the Th1/Th2 balance towards a Th2 response. However, in ATL patients treated with arsenic/IFN/https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4825, this combination therapy returns the Treg/Th2 cytokine profile at diagnosis towards a Th1 profile, with a diminution of IL‐10 (Kchour et al., 2013). This effect is probably due to the depletion of ATL leukaemia cells that disrupt the immune system initially.

Moreover, As(III) was found to be a potent inhibitor of https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=4982. At a concentration which is not able to affect IFN‐γ secretion by Th1 cells, NaAsO2 almost totally blocks the IL‐17 secretion by human Th17 cells, via mRNA reduction of the retinoic‐related orphan receptor (ROR)C gene which encodes RORγt, the key transcription factor of Th17 (Morzadec et al., 2012). As the Th17/Treg balance is critical in keeping immune homeostasis, As(III) treatment may lead to a regulatory response due to the potent inhibition of Th17. The effects of As2O3 on apoptosis and functions of CD4+ T cells are summarized in Figure 2.

FIGURE 2.

FIGURE 2

Mechanisms of action underlying the effects of As2O3 on CD4+ T cells

3.6. Trivalent arsenic and B cells

Baysan et al. showed that As2O3 induces apoptosis of the Bcl‐2 negative human B‐cell line Ramos via the mitochondrial pathway, by up‐regulating the expression of pro‐apoptotic proteins Bax and Bim (Dangleben et al., 2013). Moreover, it also blocks the mitogen‐mediated B‐cell differentiation towards plasmacytes and their IgM secretion (Rousselot et al., 1999). In addition, As2O3, together with https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=6825, reduces the deposition and expression of IgG and IgM in both the xenograft and recipient sera in a heart xenotransplant model (Jiao et al., 2016). Collectively, As(III) treatment probably leads to a global suppression of the humoral immune system.

3.7. Trivalent arsenic and NK cells

As2O3 reduces NK cells differentiation from haematopoietic stem cell precursors, leading to a delayed NK cell reconstitution in treated APL (Alex et al., 2018). As2O3 also facilitates NK cell‐mediated cytotoxicity towards several cancer cell lines, through modulation of both NK cell receptors and malignant cell ligand profile (Alex et al., 2018; Kim et al., 2008). Collectively, As2O3 may enhance the cytotoxicity of NK cells.

3.8. Trivalent arsenic and inflammasomes

In the human monocyte cell line THP‐1 and mouse bone marrow‐derived macrophages, As2O3 inhibits the https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=1770 inflammasome and subsequent IL‐1β and IL‐18 secretion by targeting PML (Ahn et al., 2018; Lo et al., 2013). Moreover, As2O3 inhibits https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=1768, https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=2793/https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=1782 inflammasomes in the same subset of cells (Maier, Crown, Liu, Leppla, & Moayeri, 2014). However, in the human keratinocyte cell line and mouse skin tissue, NaAsO2 promotes IL‐1β and IL‐18 secretion via https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=2923 inflammasome activation (Zhang et al., 2016). In addition, the inflammasome https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=1769 polymorphism is associated with arsenic‐induced skin lesions in humans (Bhattacharjee et al., 2013). Therefore, As(III) exerts bidirectional regulation on inflammasomes, which is probably tissue dependent.

4. TRIVALENT ARSENIC IN MOUSE MODELS OF IMMUNE‐MEDIATED DISEASES

4.1. Autoimmune diseases and asthma

Recently, As(III) was found to have therapeutic efficacy in several mouse models of autoimmune and inflammatory diseases (summarized in Table 2). As2O3 induces the regression of almost all the lesions in MRL/lpr mice, a mouse model of systemic lupus erythematosus (SLE) and lymphoproliferative syndrome, by elimination of the unusually activated T lymphocytes, and reduction of the related autoantibodies (Bobe et al., 2006). In the 2,4,6‐trinitrobenzene sulfonic acid‐induced murine model of inflammatory bowel disease, As2O3 reduces the induced colitis via NF‐κB down‐regulation and caspase‐3 activation (Singer, Trugnan, & Chelbi‐Alix, 2011). In a murine model of systemic sclerosis constructed by intradermal injections of hypochlorous acid (HOCl), As2O3 improves skin and lung fibrosis through ROS‐mediated killing of activated fibroblasts (Kavian, Marut, Servettaz, Nicco, et al., 2012). Moreover, As2O3 improves the pathological changes in a rat model of rheumatoid arthritis through a pro‐apoptotic effect on rheumatoid arthritis fibroblast‐like synoviocytes (Mei et al., 2011). In addition, NaAsO2 decreases the disease incidence and delays its onset in non‐obese diabetic mouse, a mouse model of autoimmune type 1 diabetes, which is due to the reduced proliferation and activation of T cells (Lee et al., 2015).

TABLE 2.

Use of trivalent arsenic in mouse models of immune‐mediated diseases

Investigated disease Mouse strain Optimal therapeutic regime Clinical efficacy Possible mechanism References
Autoimmune diseases

SLE/lymphoproliferative syndrome

MRL/lpr mice

As2O3 (i.p)

5 μg·g−1·day−1 for 2 months starting from 2 months of age

1. Prolong survival

2. Quasi‐total regression of antibody‐ and cell‐mediated manifestations

1. Anti‐DNA autoantibody↓RF↓IL‐18↓IFN‐γ↓NO metabolite↓TNF‐α↓Fas ligand↓IL‐10↓ in serum

2. Reduction of immune‐complex deposits in glomeruli

(Bobe et al., 2006)
Inflammatory bowel diseases TNBS‐induced colitis murine model (BALB/c)

As2O3 (i.p)

Prevention: 5 μg·g−1·day−1 from 4 days before TNBS administration

Treatment: 5 μg·g−1·day−1 when the disease was noted

1. Prolong survival

2. Reduced the induced colitis as assessed by macroscopic and microscopic scores

1. NF‐κB inhibition

2. TNF‐α↓IL‐1β↓IL‐12↓IL‐17↓IL‐18↓IL‐23↓ expressions in colonic extracts

3. Elimination of inflamed cells through apoptosis

(Singer et al., 2011)
Systemic sclerosis

SSc induced by

intradermal injections of HOCl (BALB/c)

As2O3 (i.p)

5 μg·g−1·day−1 for 6 weeks, simultaneously with HOCl injection

1. Reduction in skin and lung fibrosis

2. prevention of endothelial injuries

1. ROS generation that selectively kills activated fibroblasts

2. Autoantibody↓IL‐4↓ and IL‐13↓ from activated T cells

(Kavian, Marut, Servettaz, Nicco, et al., 2012)

Rheumatoid arthritis

Collagen‐induced arthritis rat model

As2O3 (N/A)

Inhibition of synovial hyperplasia and inflammation 1. Apoptosis induction of RA fibroblast‐like synoviocytes through NF‐κB signalling pathway and caspase cascade (Mei et al., 2011)
Type I diabetes Non‐obese diabetic mice

NaAsO2 (oral)

5 μg·g−1·day−1 for 8 weeks from 8 weeks of age

1. Decrease of incidence and disease onset delay

1. Reduction of infiltration of immunocytes in islets

2. Inhibition of T cell proliferation/activation

(Lee et al., 2015)
Airway chronic inflammatory disorder
Asthma Chicken OVA‐challenged murine model of asthma (BALB/c)

As2O3 (i.p)

4 μg·g−1·day−1 for 7 days, 30 min before each challenge

1. Amelioration of the allergen‐driven airway hyperresponsiveness

1. Attenuation of airway eosinophils chemotaxin and recruitment in bronchoalveolar lavage fluid (BALF)

2. IkBα expression increase and NF‐κB decrease

(Chu et al., 2010; Zhou et al., 2006)
Allograft rejection and graft‐versus‐host disease (GVHD)
Cardiac allograft rejection

Model 1. C3H → C57BL/6

Model 2. BALB/c → C57BL/6

Model 3. BALB/c → C57BL/6 (allo‐primed T cells pre‐transferred)

Model 1. As2O3 (i.p) (1 μg·g−1·day−1, Days 3–10) + CsA

Model 2. As2O3 (i.p) (5 μg·g−1·day−1, Days 0–10) + (anti‐CD154 and anti‐LFA‐1)

Model 3. As2O3 (i.p) (3 μg·g−1·day−1, days 0‐10) + (anti‐CD154 and anti‐LFA‐1)

1. Prolong allograft survival

1. Inhibition of graft lymphocyte infiltration

2. IFN‐γ↓IL‐2↓TGF‐β↑ in both

recipient serum and the graft

3. CD4+ Tm (%)↑ CD8+ Tm (%)↓ but Treg (%)↑ in the spleen of recipients

(Yan et al., 2009; Xu et al., 2010;

Lin et al., 2011)

Cardiac allograft rejection

Model 1. C57BL/6 → nude mouse (CD4+ Tm pre‐transferred)

Model 2. C57BL/6 → nude mouse (allo‐primed CD8+ Tm pre‐transferred)

Models 1 and 2. As2O3 (i.p) (3 μg·g−1·day−1, Days 0–10)

1. Prolong accelerated allograft rejection mediated by CD4 and CD8 memory T cells

1. CD4+ Tm (%)↓ and CD8+ Tm (%)↓ in recipient spleen and lymph nodes

2. IFN‐γ↓ IL‐2↓ and TGF‐β↑ IL‐10↑ in both recipient serum and allograft

(Yan et al., 2013;

Li et al., 2015)

Islet allgraft rejection

Model 1. BALB/→C57BL/6

Model 2. Lewis rats→C57BL/6

Model 1. As2O3 (i.p) (2 μg·g−1·day−1, Days 0–9)

Model 2. As2O3 (i.p) (10 μg·g−1·day−1, Days 0–9)

1. Prolong islet allograft and xenograft survival

1.CD4+ T (%)↓ CD8+ T (%)↓and Foxp3+ Treg (%)↑ in recipient spleen and lymph nodes

2.IFN‐γ↓IL‐2↓ and TGF‐β↑ in recipient serum and allograft

(Gao et al., 2015; Zhao, Xia, et al., 2018)
Chronic GVHD B10.D2 → BALB/c As2O3 (i.p) (5 μg·g−1·day−1, 3 weeks from Day 7 post‐transplantation) 1. Reduction of the fibrotic changes and prevention of the clinical symptoms

1. Activated CD4+ T cells (%)↓ and pDCs (%)↓ in splenocytes through apoptosis

2. Splenic IL‐4↓, IL‐17↓ and serum Anti‐DNA‐topoisomerase 1 autoantibody↓

(Kavian et al, 2012)

Abbreviations: As2O3, arsenic trioxide; HOCl, hypochlorous acid; IkBα,nuclear factor of κ light polypeptide gene enhancer in B‐cell inhibitor, α; NaAsO2, sodium arsenite; OVA, ovalbumin; pDC, plasmacytoid dendritic cell; RA, rheumatoid arthritis; RF, rheumatoid factor; TNBS, 2,4,6‐trinitrobenzene sulfonic acid; SSc, systemic sclerosis; SLE, systemic lupus erythematosus; Tm, memory T cell.

Asthma is a T‐helper type 2 (Th2) lymphocyte‐mediated chronic inflammatory disorder characterized by airway eosinophilia and airway hyperresponsiveness. As2O3 is observed to ameliorate the allergen‐driven airway hyperresponsiveness in a mouse model of asthma, through modulation of the NF‐κB pathway, airway eosinophil recruitment, and functions (Chu et al., 2010; Zhou et al., 2006).

Overall, As(III) has therapeutic potential on several autoimmune and inflammatory diseases through a pro‐apoptotic effect and effector‐cell functional modulation.

4.2. Allogeneic organ/stem cell transplantation

During organ or allogeneic stem cell transplantation, human leukocyte antigen mismatches lead to severe T‐ and B‐cell‐mediated alloreactivity, which may eventually lead to severe inflammatory transplantation complications of allograft rejection or graft‐versus‐host disease (GVHD) respectively. As2O3 has been shown to prolong the allograft survival in immunocompetent mouse heart transplant models, with reduction of the proportions of CD4+ and CD8+ memory T cells and increase of Tregs in recipient spleen and lymph nodes (Lin et al., 2011; Xu et al., 2010; Yan et al., 2009). Meanwhile, IFN‐γ expression was reduced, and TGF‐β expression was increased in both the recipient serum and the graft (Lin et al., 2011; Xu et al., 2010). Further studies have shown that As2O3 inhibits accelerated allograft rejection mediated by alloreactive CD8+ and/or CD4+ memory T cells and prolongs allograft survival (Li et al., 2015; Yan et al., 2013). Moreover, in two models (one allo‐ and one xeno‐) of islet transplantation, As2O3 was able to prolong the graft survival by inhibiting inflammatory reactions and T‐cell responses (Gao et al., 2015; Zhao, Xia, et al., 2018). GVHD occurs when immunocompetent T cells in the graft recognize the recipient as foreign and attacks the target organs. A study revealed that As2O3 can prevent disease occurrence in a murine sclerodermatous GVHD model mediated by overproduction of H2O2 which kills activated CD4+ T cells and pDCs (Kavian, Marut, Servettaz, Laude, et al., 2012). To summarize, As(III) could be a promising drug for alloreactivity through effector‐cell modulations.

4.3. Clinical trials of trivalent arsenic on immune‐mediated diseases

Given the discoveries in basic research and preclinical models, interests grow on As(III) as a clinical therapeutic agent in immune‐mediated diseases. An ongoing randomized clinical trial (NCT02966301) is currently testing As2O3 as first‐line treatment of chronic GVHD. Results of a phase 2a randomized clinical trial (NCT01738360) evaluating the therapeutic efficacy of As2O3 in SLE are expected soon.

5. TRIVALENT ARSENIC AND TUMOUR IMMUNOTHERAPY

The direct effects of As(III) on tumour cells are not discussed here but are reviewed by Emadi and Gore (2010). The past decade has witnessed breakthroughs which bring immunotherapy to the forefront of cancer therapy. As(III) acts in cellular immunotherapy in two ways (summarized in Table 3). On the one hand, it selectively depletes tumour promoting cells. The ratio of effector to regulatory T cells (Teff/Treg) is of great importance in immune modulation, and targeting Tregs to enhance anti‐tumour immune responses has become an important strategy in cancer immunology (Roychoudhuri, Eil, & Restifo, 2015). It was observed that As2O3 reduces the increased Treg numbers and Foxp3 mRNA levels in ex vivo malignant ascites (Hu et al., 2018). In two murine models of colon and liver cancer, As2O3 exerted its anti‐tumour effect in both in situ carcinoma and colon cancer lung metastasis through selective depletion of the infiltrated Tregs (Thomas‐Schoemann et al., 2012; Wang et al., 2017; Wang, Hu, Xu, & Liu, 2016). On the other hand, As(III) enhances cytotoxicity of tumour‐killing cells. It has been shown that As2O3 increases the cytotoxic activity of cytokine‐induced killer (CIK) cells and the IFN‐γ secretion in vitro, which may help in tumour control (Wang et al., 2016; Wang et al., 2017). Moreover, exposure of myeloma cell lines to As2O3 increases lymphokine‐activated killer (LAK)‐mediated killing by up‐regulation of CD38 and CD54 on the myeloma cells, and increased expression of CD31 (CD38 ligand) and CD11a (CD54 ligand) on LAKs, suggesting that the improved killing is mediated by increased adhesion (Deaglio et al., 2001). In addition, exposure of NK and leukemic cells to low doses of As2O3 modulates NK cell receptors and malignant cell ligand profile in a direction that enhances NK cell‐mediated cytolytic activity (Alex et al., 2018; Kim et al., 2008). This effect was proved in a mouse model of APL, where the As2O3 + NK treatment promoted longer survival as compared with As2O3 alone (Alex et al., 2018). Overall, As2O3 could work as an effective adjuvant for cellular immunotherapy in both haematological malignancies and solid tumours.

TABLE 3.

Trivalent arsenic facilitates tumour immunotherapy

Investigated disease Ex vivo sample/mouse model Optimal therapeutic regimen Clinical efficacy Possible mechanism References
Malignant ascites TILs derived from ascites of gastric cancer patients As2O3 (in vitro, 5 or 10 μM for 48 hr) N/A
  1. CD4+ T cell (%)↓ CD8+ T cell (%)↑ Treg (%)↓ with Foxp3 expression↓ in TILs

2. IL‐10↓ TGF‐β↓IFN‐γ↑ TIL cytotoxicity↑

(Hu et al., 2018)
Colon cancer

Model 1. Colon cancer‐bearing mouse (BALB/c, injection s.c. of CT‐26 colon cancer cell)

Model 2. Lung metastasis model of colon cancer (BALB/c, injection i.v. of CT‐26)

Model 1. As2O3 (i.p) 1 μg·g−1 once at Day 10 of tumour injection

Model 2. As2O3 (i.v) 6 μg·g−1 every 2 days for 2 weeks from Day 3 of tumour injection

1. Delay tumour growth

2. Inhibition of lung metastasis and prolong mouse survival

1. Selective Treg depletion with Foxp3 expression↓ in spleen and tumour tissue, due to ROS and RNS accumulation in Tregs.

2. Cytokine induced killer cells (in vitro): cytotoxicity↑IFN‐γ↑

(Thomas‐Schoemann et al., 2012; Wang et al., 2016)
Hepatic cancer Hepatic cancer bearing mouse (KM strain, H22 hepatic cancer cell liver implantation)

As2O3 (i.v)

6 μg·g−1 every 2 days for 2 weeks right after liver tumour implantation

1. Prolong mouse survival

2. Inhibition of tumour growth

3. Reduction of abdominal adhesion and ascites

1. Selective Treg depletion

2. Serum FN‐γ↑IL‐10↓TGF‐β↓

3. IOD of CD3+ T cell↑Foxp3+ cell↓ in tumour

(Wang et al., 2017)
Multiple myeloma Co‐culture of LAK and myeloma cell As2O3 (in vitro, 0.5 μM for 72 hr) N/A 1. Increase of LAK‐mediated lysis via up‐regulation of expressions of CD54 and CD38 on myeloma cells, and CD31(CD38 ligand) and CD11a (CD54 ligand) on LAKs (Deaglio et al., 2001)
Acute promyelocytic leukaemia APL bearing mouse model (FVB/N, APL blast cells from MRP8‐PML‐RARa transgenic FVB/N mouse)

As2O3 (i.p) (5 μg·g−1·day−1, Days 8 to 35 of tumour injection) and

Syngeneic NK cells (i.v) (5 * 105 cells/dose, Days 8, 18, and 28 of tumour injection)

1. Prolong survival comparing As2O3 + NK group and As2O3 group alone

1. Enhancement of NK cell cytolytic activity

2. Alteration of NK cell receptor and ligand Profile on tumour cell

(Alex et al., 2018; Kim et al., 2008)
Hepatic cancer Hepatic cancer bearing mouse (BALB/c, H22 hepatic cancer cell) Intratumoral injection of 100 μg B7H3 plasmid (on Day 14 of tumour injection) and 5 μg As2O3 (every 2 days) until tumour disappear 1. Transplanted hepatoma eradication and regression of distant tumour nodules

1. Generation of anti‐tumour immunity relies largely on CD8+ T cells and NK cells

2. Serum IFN‐γ↑ and CTL activity↑

(Luo et al., 2006)
Bladder cancer Orthotopic bladder cancer model (C3H/HeN, human bladder cancer cells 5637) Intravesical instillation of 0.2‐ml BCG (600 mg·L−1) and 0.2 ml of As2O3 (30 μmol·L−1·week−1) for 4 weeks 1. Reduction of tumour weight and volume

1. Apoptosis of tumour cells↑ via IER3/Nrf2 pathway↓

2. DC (%) in cancer tissue↑Expression of CD83/CD86↑IL‐6/IL‐8 in DCs↑

(Mao et al., 2018)

Abbreviations: APL, acute promyelocytic leukaemia; As2O3, arsenic trioxide; BCG, bacillus Calmette‐Guerin; IOD, integrated OD; LAK, lymphokine activated killers; TIL, tumour‐infiltrating lymphocytes; RNS, reactive nitrogen species.

Moreover, As(III) can synergize with B7‐H3 (CD276), an important immune checkpoint member of the B7 and CD28 families, to eradicate hepatocellular carcinoma in a mouse model, along with the generation of potent systemic antitumor immunity mediated by CD8+ T and NK cells (Luo et al., 2006). As(III) was also used in a mouse model of bladder cancer to facilitate intravesical bacillus Calmette‐Guerin immunotherapy, by targeting the IER3/Nrf2 pathway (Mao et al., 2018).

6. DOSING OF THERAPEUTIC AS(III)

Two different As2O3 dosing schedules have been assessed in Phase 3 clinical trials for APL, with the protocol of 0.15 mg·kg−1·day−1 daily until complete remission (up to 140 doses of 0.15 mg·kg−1) in the Italian‐German trial (Lo‐Coco et al., 2013) and the protocol of 0.3 mg·kg−1 on Days 1 to 5 in Week 1 followed by 0.25 mg·kg−1 twice weekly for 7 weeks (63 doses of 0.25–0.30 mg·kg−1) in the U.K. National Cancer Research Institute trial (Burnett et al., 2015). Despite the discrepancy on the frequency of use, the intensity of As2O3 in the 2 schedules is, however, almost identical with respect to total As2O3 dose in milligrams per kilogram.

The appropriate dosing of As2O3 in the treatment of immune‐mediated diseases is being assessed in clinical trials. In the Phase 2a study evaluating As2O3 in SLE (NCT01738360), patients received 10 doses of As2O3 up to 0.30 mg·kg−1·day−1 within a period of 25 days. In the ongoing Phase 2 trial testing As2O3 in chronic GVHD (NCT02966301), each patient received 11 doses of As2O3 (0.15 mg·kg−1·day−1) over a 4‐week period (one cycle) and one more cycle for consolidation when patients achieved a partial response. Therefore, As2O3 is used in immune‐mediated diseases with similar daily doses but smaller total intensity as compared with that in APL.

Notably, the use of arsenic derivatives is highly embryotoxic and is contraindicated at any stage of pregnancy (Sanz et al., 2019), while As2O3 at 0.15 mg·kg−1·day−1 (standard dose) was well tolerated by paediatric patients with APL and achieved satisfactory outcomes in clinical trials (Kutny et al., 2017). The dose of As2O3 in elderly and obese patients may need to be attenuated while no consensus has been published due to limited experience (Hickey et al., 2019; Lengfelder, Hofmann, & Nolte, 2013).

The pharmacokinetics of As2O3 as measured by plasma concentrations were comparable in different studies (Shen et al., 1997., Fox et al., 2008). In a Phase 1 clinical trial assessing single‐agent As2O3 in APL, at 0.15 mg·kg−1·day−1, total arsenic in the plasma reached the maximum concentration (Cmax) of 0.28 μM (0.11–0.37 μM) within 2 hr after completion of administration and was quickly eliminated below the lower limit of detection (0.07 μM) in 10 hr (Fox et al., 2008). Notably, repeated administration did not alter the pharmacokinetic behaviours of As2O3 (Shen et al., 1997; Fox et al., 2008), indicating no or minimal plasma drug accumulation. Moreover, the pharmacokinetics for As(III) in the bone marrow seems to be similar as compared with peripheral blood (Iriyama et al., 2012).

Concerning the acute adverse events of As(III), despite the therapeutic superiority for ATRA plus As2O3 versus ATRA plus chemotherapy in APL, the former regimen led to less haematological toxicity and fewer infections, but more common cardiac and hepatic toxicities in the two Phase 3 clinical trials (Burnett et al., 2015; Lo‐Coco et al., 2013). Indeed, cardiac toxicity manifesting as QT interval prolongation seems to be the leading adverse event of As2O3 (Roboz et al., 2014). As2O3 induces cardiotoxicity through multiple mechanisms including ROS generation and disruption of intracellular calcium homeostasis (Haybar, Shahrabi, Rezaeeyan, Jodat, & Saki, 2019). Between 11% and 26% of patients receiving As2O3 experienced QT prolongation in different studies (Burnett et al., 2015; Lo‐Coco et al., 2013; Roboz et al., 2014), and it was recommended to withhold As2O3 when QT interval is prolonged longer than 500 ms (Sanz et al., 2019). However, clinically relevant arrhythmias are rare and can be avoided with appropriate precautions such as electrolyte repletion (Roboz et al., 2014). In addition, As2O3‐induced hepatic toxicity featuring mainly as elevated liver alanine transaminase could occur in between 25% and 63% of patients receiving ATRA plus As2O3 and seems to be highly manageable with temporary discontinuation of the treatment (Burnett et al., 2015; Lo‐Coco et al., 2013).

7. ARSENIC RETENTION AND CHRONIC ADVERSE EVENTS

Arsenic (including different arsenic species such as As(III) and As(V)) retention and possible chronic adverse events are concerns surrounding the use of As2O3. In a 5‐year follow‐up study including 85 APL patients treated with As2O3 + ATRA, Hu et al. observed that after cessation of As2O3, the plasma and the urine arsenic concentrations were rapidly decreased to levels below the safety limit recommended by the U.S. Agency for Toxic Substances and Disease Registry even though the concentrations remained slightly greater than in healthy controls. For the nail and hair arsenic contents representing the long‐term exposure and in vivo accumulation of arsenic, the levels were only slightly higher than those in healthy controls. Meanwhile, no adverse events such as secondary carcinoma were observed (Hu et al., 2009). In another 6‐year follow‐up study which enrolled 72 APL patients receiving single‐agent As2O3, no additional toxicities developed after completion of the course of As2O3, and there was no significant difference in the arsenic retention in hair and nail samples of controls and patients who had completed therapy at least two years earlier. Moreover, seven patients (four women and three men) have had eight normal babies without report of abortions, indicating safety profile for fertility after treatment cessation (Mathews et al., 2010). Importantly, in a 12‐year follow‐up study including 265 APL patients treated with As2O3 + ATRA, Zhu et al. reported that plasma and urine, as well as the delayed increased hair and nail arsenic levels all returned to normal after 6 months. Interestingly, the plasma and urine arsenic levels were not higher and even lower than healthy controls on cessation of As2O3. However, incidence of mild liver dysfunction (15.2%) and hepatic steatosis (42.9%) was reported. In addition, only one patient developed breast cancer 3 years after termination of As2O3 (Zhu et al., 2016). Collectively, no significant arsenic retention was observed in these studies, and chronic adverse events, especially secondary carcinoma, are rare.

However, since the longest published follow‐up data for APL patients was 12 years, the very long‐term adverse effects especially tumourigenesis remain to be better observed. Indeed, chronic ingestion of arsenic through drinking contaminated water or inhalation results in the accumulation of arsenic in vital organs, which may lead to diabetes, atherosclerosis, hypertension, ischaemic heart diseases, hepatotoxicity, nephrotoxicity, skin lesions, and cancer of the skin, bladder, and lungs (see Khairul, Wang, Jiang, Wang, & Naranmandura, 2017).

Oxidative stress has been suggested as an important mechanism for arsenic‐induced carcinogenesis (Kitchin & Conolly, 2010). In rat models, As(III) caused an elevation of 8‐hydroxydeoxyguanosine by generation of ROS, stimulated cell proliferation, and induced liver and bladder carcinogenicity (Kinoshita, Wanibuchi, Wei, Yunoki, & Fukushima, 2007; Suzuki, Arnold, Pennington, Kakiuchi‐Kiyota, & Cohen, 2009). In addition, intracellular signal transduction and transcription factors activation are related to arsenic‐induced carcinogenicity (Khairul et al., 2017). Moreover, As(III) interferes with key cellular processes such as DNA damage‐repair and chromosomal structure, leading to genomic instability. At the epigenetic level, As(III) affects global hypomethylation and hypermethylation of specific gene promoters, leading to deregulation of both oncogenic and tumour‐suppressive genes (Sage et al., 2017). These factors may operate together and lead to abnormal cell proliferation, mutations accumulation, and finally development of cancers. Nevertheless, As(III) may compromise the immune system's ability to rid the host of pathogens and tumours (Dangleben et al., 2013). Arsenic‐exposed mice demonstrated depressed humoral and cellular immunity and displayed significantly impaired resistance against B16F10 melanoma, which resulted in sevenfold increased tumour burden (Sikorski, Mccay, White, Bradley, & Munson, 1989). Collectively, attentions should be paid to patients with immune deficiency or defective DNA repair machinery when considering As(III) treatment as it may lead to higher risk of infection and carcinogenesis in the long term.

Millions of people worldwide are exposed to arsenic on a regular basis. However, as compared with environmental exposure which is low in concentration (mostly below 10 μg·kg−1·day−1) and very long in duration (decades; Dangleben et al., 2013), the standard As2O3 treatment is much higher in concentration (0.15 mg·kg−1·day−1) and shorter in duration (weeks). Arsenic‐induced toxicity may differ between these two types of exposure (Zhang, Zhang, Wang, Li, & Zhang, 2018). Moreover, risks of accumulated toxicity due to drug plus environmental exposure may exist and require further studies.

8. CONCLUSIONS AND PERSPECTIVES

As(III), when used in a clinically relevant dose, that is, between 0.07 and 0.28 μM, as found in the plasma of APL patients treated with As2O3 (Fox et al., 2008), can target specific immune cell subsets and exert immunomodulatory effects. The efficacy of As(III) on mouse models of autoimmune and inflammatory diseases highlights its therapeutic potential in humans. It emerges as a promising adjuvant to cancer immunotherapy agents in the treatment of both haematological and solid tumours.

It is noteworthy that the As(III) effect is dose, cellular, and tissue dependent, which requires deeper understanding of this drug in vitro, in vivo, and in clinical practice. For this purpose, comprehensive genomic, proteomic, and metabolomic profiling will be critical for identifying and validating potential molecular targets of As(III) for future therapeutic use.

Several clinical trials with As(III) in autoimmune and inflammatory diseases are ongoing to prove the “bench to bedside” indication of As(III). Notably, during As2O3 use in APL, adverse events including cardiac and hepatic toxicities have been observed, which require precautions also during its usage in immune‐mediated diseases.

8.1. Nomenclature of targets and ligands

Key protein targets and ligands in this article are hyperlinked to corresponding entries in https://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Harding et al., 2018), and are permanently archived in the Concise Guide to PHARMACOLOGY 2019/2020 (Alexander, Cidlowski et al., 2019; Alexander, Fabbro et al., 2019a, 2019b; Alexander, Kelly et al., 2019; Alexander, Mathie et al., 2019).

CONFLICT OF INTEREST

The authors declare no conflicts of interest.

ACKNOWLEDGEMENTS

The authors acknowledge the Association for Training, Education and Research in Hematology, Immunology and Transplantation for the generous and continuous support to the research work. Y.Y. thanks China Scholarship Council for financial support (CSC 201606320257). The authors thank Prof. Ali Bazarbachi for critical reading of the manuscript.

Ye Y, Gaugler B, Mohty M, Malard F. Old dog, new trick: Trivalent arsenic as an immunomodulatory drug. Br J Pharmacol. 2020;177:2199–2214. 10.1111/bph.15011

REFERENCES

  1. Ablain, J. , Rice, K. , Soilihi, H. , De Reynies, A. , Minucci, S. , & de Thé, H. (2014). Activation of a promyelocytic leukemia‐tumor protein 53 axis underlies acute promyelocytic leukemia cure. Nature Medicine, 20, 167–174. [DOI] [PubMed] [Google Scholar]
  2. Ahn, H. , Kim, J. , Kang, S. G. , Yoon, S. I. , Ko, H. J. , Kim, P. H. , … Lee, G. S. (2018). Mercury and arsenic attenuate canonical and non‐canonical NLRP3 inflammasome activation. Scientific Reports, 8, 13659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alex, A. A. , Ganesan, S. , Palani, H. K. , Balasundaram, N. , David, S. , Lakshmi, K. M. , … Janet, N. B. (2018). Arsenic trioxide enhances the NK cell cytotoxicity against acute promyelocytic leukemia while simultaneously inhibiting its bio‐genesis. Frontiers in Immunology, 9, 1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alexander, S. P. H. , Cidlowski, J. A. , Kelly, E. , Mathie, A. , Peters, J. A. , Veale, E. L. , … CGTP Collaborators (2019). The Concise Guide to PHARMACOLOGY 2019/20: Nuclear hormone receptors. British Journal of Pharmacology, 176, S229–S246. 10.1111/bph.14750 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Alexander, S. P. H. , Fabbro, D. , Kelly, E. , Mathie, A. , Peters, J. A. , Veale, E. L. , … CGTP Collaborators (2019a). The Concise Guide to PHARMACOLOGY 2019/20: Catalytic receptors. British Journal of Pharmacology, 176, S247–S296. 10.1111/bph.14751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Alexander, S. P. H. , Fabbro, D. , Kelly, E. , Mathie, A. , Peters, J. A. , Veale, E. L. , … CGTP Collaborators (2019b). The Concise Guide to PHARMACOLOGY 2019/20: Enzymes. British Journal of Pharmacology, 176, S297–S396. 10.1111/bph.14752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Alexander, S. P. H. , Kelly, E. , Mathie, A. , Peters, J. A. , Veale, E. L. , Faccenda, E. , … CGTP Collaborators (2019). The Concise Guide to PHARMACOLOGY 2019/20: Introduction and Other Protein Targets. British Journal of Pharmacology, 176, S1–S20. 10.1111/bph.14747 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Alexander, S. P. H. , Mathie, A. , Peters, J. A. , Veale, E. L. , Striessnig, J. , Kelly, E. , … CGTP Collaborators (2019). The Concise Guide to PHARMACOLOGY 2019/20: Ion channels. British Journal of Pharmacology, 176, S142–S228. 10.1111/bph.14749 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Antoine, F. , Ennaciri, J. , & Girard, D. (2010). Syk is a novel target of arsenic trioxide (ATO) and is involved in the toxic effect of ATO in human neutrophils. Toxicol Vitr, 24, 936–941. [DOI] [PubMed] [Google Scholar]
  10. Bahari, A. , & Salmani, V. (2017). Environmentally relevant dose of arsenic interferes in functions of human monocytes derived dendritic cells. Toxicology Letters, 275, 118–122. [DOI] [PubMed] [Google Scholar]
  11. Banerjee, N. , Banerjee, S. , Sen, R. , Bandyopadhyay, A. , Sarma, N. , Majumder, P. , … Giri, A. K. (2009). Chronic arsenic exposure impairs macrophage functions in the exposed individuals. Journal of Clinical Immunology, 29, 582–594. [DOI] [PubMed] [Google Scholar]
  12. Baysan, A. , Yel, L. , Gollapudi, S. , Su, H. , & Gupta, S. (2007). Arsenic trioxide induces apoptosis via the mitochondrial pathway by upregulating the expression of Bax and Bim in human B cells. International Journal of Oncology, 30, 313–318. [PubMed] [Google Scholar]
  13. Bhattacharjee, P. , Das, N. , Chatterjee, D. , Banerjee, A. , Das, J. K. , Basu, S. , … Giri, A. K. (2013). Association of NALP2 polymorphism with arsenic induced skin lesions and other health effects. Mutation Research, 755, 1–5. [DOI] [PubMed] [Google Scholar]
  14. Binet, F. , Cavalli, H. , Moisan, E. , & Girard, D. (2006). Arsenic trioxide (AT) is a novel human neutrophil pro‐apoptotic agent: Effects of catalase on AT‐induced apoptosis, degradation of cytoskeletal proteins and de novo protein synthesis. British Journal of Haematology, 132, 349–358. [DOI] [PubMed] [Google Scholar]
  15. Binet, F. , Chiasson, S. , & Girard, D. (2008). Arsenic trioxide induces de novo protein synthesis of annexin‐1 in neutrophils: Association with a heat shock‐like response and not apoptosis. British Journal of Haematology, 140, 454–463. [DOI] [PubMed] [Google Scholar]
  16. Binet, F. , Chiasson, S. , & Girard, D. (2010a). Evidence that endoplasmic reticulum (ER) stress and caspase‐4 activation occur in human neutrophils. Biochemical and Biophysical Research Communications, 391, 18–23. [DOI] [PubMed] [Google Scholar]
  17. Binet, F. , Chiasson, S. , & Girard, D. (2010b). Arsenic trioxide induces endoplasmic reticulum stress‐related events in neutrophils. International Immunopharmacology, 10, 508–512. [DOI] [PubMed] [Google Scholar]
  18. Binet, F. , & Girard, D. (2008). Novel human neutrophil agonistic properties of arsenic trioxide: Involvement of p38 mitogen‐activated protein kinase and/or c‐jun NH2‐terminal MAPK but not extracellular signal‐regulated kinases‐1/2. Journal of Leukocyte Biology, 84, 1613–1622. [DOI] [PubMed] [Google Scholar]
  19. Bishayi, B. , & Sengupta, M. (2003). Intracellular survival of Staphylococcus aureus due to alteration of cellular activity in arsenic and lead intoxicated mature Swiss albino mice. Toxicology, 184, 31–39. [DOI] [PubMed] [Google Scholar]
  20. Bobe, P. , Bonardelle, D. , Benihoud, K. , Opolon, P. , & Chelbi‐Alix, M. K. (2006). Arsenic trioxide: A promising novel therapeutic agent for lymphoproliferative and autoimmune syndromes in MRL/lpr mice. Blood, 108, 3967–3975. [DOI] [PubMed] [Google Scholar]
  21. Bourdonnay, E. , Morzadec, C. , Fardel, O. , & Vernhet, L. (2009). Redox‐sensitive regulation of gene expression in human primary macrophages exposed to inorganic arsenic. Journal of Cellular Biochemistry, 107, 537–547. [DOI] [PubMed] [Google Scholar]
  22. Bourdonnay, E. , Morzadec, C. , Fardel, O. , & Vernhet, L. (2011). Arsenic increases lipopolysaccharide‐dependent expression of interleukin‐8 gene by stimulating a redox‐sensitive pathway that strengthens p38‐kinase activation. Molecular Immunology, 48, 2069–2078. [DOI] [PubMed] [Google Scholar]
  23. Bourdonnay, E. , Morzadec, C. , Sparfel, L. , Galibert, M. D. , Jouneau, S. , Martin‐Chouly, C. , … Vernhet, L. (2009). Global effects of inorganic arsenic on gene expression profile in human macrophages. Molecular Immunology, 46, 649–656. [DOI] [PubMed] [Google Scholar]
  24. Burnett, A. K. , Russell, N. H. , Hills, R. K. , Bowen, D. , Kell, J. , Knapper, S. , … Khwaja, A. (2015). Arsenic trioxide and all‐trans retinoic acid treatment for acute promyelocytic leukaemia in all risk groups (AML17): Results of a randomised, controlled, phase 3 trial. The Lancet Oncology, 16, 1295–1305. [DOI] [PubMed] [Google Scholar]
  25. Chen, G. Q. , Zhu, J. , Shi, X. G. , Ni, J. H. , Zhong, H. J. , Si, G. Y. , … Shen, Z. X. (1996). In vitro studies on cellular and molecular mechanisms of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia As2O3 induces NB4 cell apoptosis with downregulation of Bcl‐2 expression and modulation of PML‐RARα/PML proteins. Blood, 88, 1052–1061. [PubMed] [Google Scholar]
  26. Chou, W. C. , Jie, C. , Kenedy, A. A. , Jones, R. J. , Trush, M. A. , & Dang, C. V. (2004). Role of NADPH oxidase in arsenic‐induced reactive oxygen species formation and cytotoxicity in myeloid leukemia cells. Proceedings of the National Academy of Sciences of the United States of America, 101, 4578–4583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chu, K. H. , Lee, C. C. , Hsin, S. C. , Cai, B. C. , Wang, J. H. , & Chiang, B. L. (2010). Arsenic trioxide alleviates airway hyperresponsiveness and eosinophilia in a murine model of asthma. Cellular & Molecular Immunology, 7, 375–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Cicconi, L. , & Lo‐Coco, F. (2016). Current management of newly diagnosed acute promyelocytic leukemia. Annals of Oncology, 27, 1474–1481. [DOI] [PubMed] [Google Scholar]
  29. Conde, P. , Acosta‐Saavedra, L. C. , Goytia‐Acevedo, R. C. , & Calderon‐Aranda, E. S. (2007). Sodium arsenite‐induced inhibition of cell proliferation is related to inhibition of IL‐2 mRNA expression in mouse activated T cells. Archives of Toxicology, 81, 251–259. [DOI] [PubMed] [Google Scholar]
  30. Dangleben, N. L. , Skibola, C. F. , & Smith, M. T. (2013). Arsenic immunotoxicity: a review. Env. Heal., 12, 73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Dassouki, Z. , Sahin, U. , El Hajj, H. , Jollivet, F. , Kfoury, Y. , Lallemand‐Breitenbach, V. , … Bazarbachi, A. (2015). ATL response to arsenic/interferon therapy is triggered by SUMO/PML/RNF4‐dependent Tax degradation. Blood, 125, 474–482. [DOI] [PubMed] [Google Scholar]
  32. de Thé, H. (2018). Differentiation therapy revisited. Nature Reviews. Cancer, 18, 117–127. [DOI] [PubMed] [Google Scholar]
  33. Deaglio, S. , Canella, D. , Baj, G. , Arnulfo, A. , Waxman, S. , & Malavasi, F. (2001). Evidence of an immunologic mechanism behind the therapeutical effects of arsenic trioxide (As2O3) on myeloma cells. Leukemia Research, 25, 227–235. [DOI] [PubMed] [Google Scholar]
  34. Duan, X. , Gao, S. , Li, J. , Wu, L. , Zhang, Y. , Li, W. , … Li, B. (2017). Acute arsenic exposure induces inflammatory responses and CD4+ T cell subpopulations differentiation in spleen and thymus with the involvement of MAPK, NF‐κB, and Nrf2. Molecular Immunology, 81, 160–172. [DOI] [PubMed] [Google Scholar]
  35. El Hajj, H. , Dassouki, Z. , Berthier, C. , Raffoux, E. , Ades, L. , Legrand, O. , … Zibara, K. (2015). Retinoic acid and arsenic trioxide trigger degradation of mutated NPM1, resulting in apoptosis of AML cells. Blood, 125, 3447–3754. [DOI] [PubMed] [Google Scholar]
  36. El Hajj, H. , El‐Sabban, M. , Hasegawa, H. , Zaatari, G. , Ablain, J. , Saab, S. T. , … Nicot, C. (2010). Therapy‐induced selective loss of leukemia‐initiating activity in murine adult T cell leukemia. The Journal of Experimental Medicine, 207, 2785–2792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. El‐Sabban, M. E. , Nasr, R. , Dbaibo, G. , Hermine, O. , Abboushi, N. , Quignon, F. , … Bazarbachi, A. (2000). Arsenic‐interferon‐α–triggered apoptosis in HTLV‐I transformed cells is associated with Tax down‐regulation and reversal of NF‐κB activation. Blood, 96, 2849–2855. [PubMed] [Google Scholar]
  38. Emadi, A. , & Gore, S. D. (2010). Arsenic trioxide—An old drug rediscovered. Blood Reviews, 24, 191–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Fasci, D. , Anania, V. G. , Lill, J. R. , & Salvesen, G. S. (2015). SUMO deconjugation is required for arsenic‐triggered ubiquitylation of PML. Science Signaling, 8, ra56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Fox, E. , Razzouk, B. I. , Widemann, B. C. , Xiao, S. , O'Brien, M. , Goodspeed, W. , … Adamson, P. C. (2008). Phase 1 trial and pharmacokinetic study of arsenic trioxide in children and adolescents with refractory or relapsed acute leukemia, including acute promyelocytic leukemia or lymphoma. Blood, 111, 566–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Galicia, G. , Leyva, R. , Tenorio, E. P. , Ostrosky‐Wegman, P. , & Saavedra, R. (2003). Sodium arsenite retards proliferation of PHA‐activated T cells by delaying the production and secretion of IL‐2. International Immunopharmacology, 3, 671–682. [DOI] [PubMed] [Google Scholar]
  42. Gao, C. , Jiang, J. , Ma, P. , Cheng, P. , Lian, Y. , Zhao, B. , … Jin, N. (2015). Arsenic trioxide induces T cell apoptosis and prolongs islet allograft survival in mice. Transplantation, 99, 1796–1806. [DOI] [PubMed] [Google Scholar]
  43. Guilliams, M. , Mildner, A. , & Yona, S. (2018). Developmental and functional heterogeneity of monocytes. Immunity, 49, 595–613. [DOI] [PubMed] [Google Scholar]
  44. Gupta, S. , Yel, L. , Kim, D. , Kim, C. , Chiplunkar, S. , & Gollapudi, S. (2003). Arsenic trioxide induces apoptosis in peripheral blood T lymphocyte subsets by inducing oxidative stress: A role of Bcl‐2. Molecular Cancer Therapeutics, 2, 711–719. [PubMed] [Google Scholar]
  45. Harding, S. D. , Sharman, J. L. , Faccenda, E. , Southan, C. , Pawson, A. J. , Ireland, S. , … NC‐IUPHAR (2018). The IUPHAR/BPS Guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucleic Acids Res., 46, D1091–D1106. 10.1093/nar/gkx1121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Haybar, H. , Shahrabi, S. , Rezaeeyan, H. , Jodat, H. , & Saki, N. (2019). Strategies to inhibit arsenic trioxide‐induced cardiotoxicity in acute promyelocytic leukemia. Journal of Cellular Physiology, 234, 14500–14506. [DOI] [PubMed] [Google Scholar]
  47. Hickey, E. , Clemons, B. , Griffin, S. , Cox, J. , Sutphin, S. , Ramlal, R. , & Benitez, L. (2019). Multicenter evaluation of arsenic trioxide dosing in obese patients with low–intermediate risk acute promyelocytic leukemia. Leukemia & Lymphoma, 60(14), 3557–3560. [DOI] [PubMed] [Google Scholar]
  48. Hsu, K. S. , & Kao, H. Y. (2018). PML: Regulation and multifaceted function beyond tumor suppression. Cell & Bioscience, 8, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Hu, J. , Liu, Y. F. , Wu, C. F. , Xu, F. , Shen, Z. X. , Zhu, Y. M. , … Sun, H. P. (2009). Long‐term efficacy and safety of all‐trans retinoic acid/arsenic trioxide‐based therapy in newly diagnosed acute promyelocytic leukemia. Proceedings of the National Academy of Sciences of the United States of America, 106, 3342–3347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Hu, Z. , Hu, S. , Wu, Y. , Li, S. , He, C. , Xing, X. , … Du, X. (2018). Accumulation and suppressive function of regulatory T cells in malignant ascites: Reducing their suppressive function using arsenic trioxide in vitro. Oncology Letters, 15, 5384–5390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Hutchinson, L. M. , Trinh, B. M. , Palmer, R. K. , Preziosi, C. A. , Pelletier, J. H. , Nelson, H. M. , & Gosse, J. A. (2011). Inorganic arsenite inhibits IgE receptor‐mediated degranulation of mast cells. Journal of Applied Toxicology, 31, 231–241. [DOI] [PubMed] [Google Scholar]
  52. Iriyama, N. , Yoshino, Y. , Yuan, B. , Horikoshi, A. , Hirabayashi, Y. , Hatta, Y. , … Takeuchi, J. (2012). Speciation of arsenic trioxide metabolites in peripheral blood and bone marrow from an acute promyelocytic leukemia patient. Journal of Hematology & Oncology, 5, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Jeanne, M. , Lallemand‐Breitenbach, V. , Ferhi, O. , Koken, M. , Le Bras, M. , Duffort, S. , … de Thé, H. (2010). PML/RARA oxidation and arsenic binding initiate the antileukemia response of As2O3 . Cancer Cell, 18, 88–98. [DOI] [PubMed] [Google Scholar]
  54. Jiao, Z. X. , Leng, Y. , Xia, J. J. , Wu, H. Q. , Jin, N. , Fu, J. Z. , … Qi, Z. Q. (2016). As2O3 combined with leflunomide prolongs heart xenograft survival via suppressing the response of Th1, Th2, and B cells in a rat model. Xenotransplantation, 23, 237–248. [DOI] [PubMed] [Google Scholar]
  55. Kavian, N. , Marut, W. , Servettaz, A. , Laude, H. , Nicco, C. , Chéreau, C. , … Batteux, F. (2012). Arsenic trioxide prevents murine sclerodermatous graft‐versus‐host disease. Journal of Immunology, 188, 5142–5149b. [DOI] [PubMed] [Google Scholar]
  56. Kavian, N. , Marut, W. , Servettaz, A. , Nicco, C. , Chéreau, C. , Lemaréchal, H. , … Batteux, F. (2012). Reactive oxygen species‐mediated killing of activated fibroblasts by arsenic trioxide ameliorates fibrosis in a murine model of systemic sclerosis. Arthritis and Rheumatism, 64, 3430–3440. [DOI] [PubMed] [Google Scholar]
  57. Kchour, G. , Rezaee, S. R. , Farid, R. , Ghantous, A. , Rafatpanah, H. , Tarhini, M. , … Nasser, R. (2013). The combination of arsenic, interferon‐α, and zidovudine restores an ‘immunocompetent‐like’ cytokine expression profile in patients with adult T‐cell leukemia lymphoma. Retrovirology, 10, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Kchour, G. , Tarhini, M. , Kooshyar, M. M. , El Hajj, H. , Wattel, E. , Mahmoudi, M. , … Rezaee, S. R. (2009). Phase 2 study of the efficacy and safety of the combination of arsenic trioxide, interferon α, and zidovudine in newly diagnosed chronic adult T‐cell leukemia/lymphoma (ATL). Blood, 113, 6528–6532. [DOI] [PubMed] [Google Scholar]
  59. Khairul, I. , Wang, Q. Q. , Jiang, Y. H. , Wang, C. , & Naranmandura, H. (2017). Metabolism, toxicity and anticancer activities of arsenic compounds. Oncotarget, 8, 23905–23926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kim, J. Y. , Bae, J. H. , Lee, S. H. , Lee, E. Y. , Chung, B. S. , Kim, S. H. , & Kang, C. D. (2008). Induction of NKG2D ligands and subsequent enhancement of NK cell‐mediated lysis of cancer cells by arsenic trioxide. Journal of Immunotherapy, 31, 475–486. [DOI] [PubMed] [Google Scholar]
  61. Kim, Y. E. , & Ahn, J. H. (2015). Positive role of promyelocytic leukemia protein in type I interferon response and its regulation by human cytomegalovirus. PLoS Pathogens, 11, 1–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kinoshita, A. , Wanibuchi, H. , Wei, M. , Yunoki, T. , & Fukushima, S. (2007). Elevation of 8‐hydroxydeoxyguanosine and cell proliferation via generation of oxidative stress by organic arsenicals contributes to their carcinogenicity in the rat liver and bladder. Toxicology and Applied Pharmacology, 221, 295–305. [DOI] [PubMed] [Google Scholar]
  63. Kitchin, K. T. , & Conolly, R. (2010). Arsenic‐induced carcinogenesissoxidative stress as a possible mode of action and future research needs for more biologically based risk assessment. Chemical Research in Toxicology, 23, 327–335. [DOI] [PubMed] [Google Scholar]
  64. Kutny, M. A. , Alonzo, T. A. , Gerbing, R. B. , Wang, Y. C. , Raimondi, S. C. , Hirsch, B. A. , … Gregory, J. J. Jr. (2017). Arsenic trioxide consolidation allows anthracycline dose reduction for pediatric patients with acute promyelocytic leukemia: Report from the children's oncology group phase III historically controlled trial AAML0631. Journal of Clinical Oncology, 35, 3021–3029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Lallemand‐Breitenbach, V. , & de Thé, H. (2018). PML nuclear bodies from architecture to function. Current Opinion in Cell Biology, 52, 154–161. [DOI] [PubMed] [Google Scholar]
  66. Lallemand‐Breitenbach, V. , Zhu, J. , Puvion, F. , Koken, M. , Honoré, N. , Doubeikovsky, A. , … de Thé, H. (2001). Role of promyelocytic leukemia (Pml) sumolation in nuclear body formation, 11s proteasome recruitment, and as2O3‐induced Pml or Pml/retinoic acid receptor α degradation. The Journal of Experimental Medicine, 193, 1361–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Lee, Y. S. , Kim, D. , Lee, E. K. , Kim, S. , Choi, C. S. , & Jun, H. S. (2015). Sodium meta‐arsenite prevents the development of autoimmune diabetes in NOD mice. Toxicology and Applied Pharmacology, 284, 254–261. [DOI] [PubMed] [Google Scholar]
  68. Lemarie, A. , Bourdonnay, E. , Morzadec, C. , Fardel, O. , & Vernhet, L. (2008). Inorganic arsenic activates reduced NADPH oxidase in human primary macrophages through a Rho kinase/p38 kinase pathway. Journal of Immunology, 180, 6010–6017. [DOI] [PubMed] [Google Scholar]
  69. Lemarie, A. , Morzadec, C. , Bourdonnay, E. , Fardel, O. , & Vernhet, L. (2006). Human macrophages constitute targets for immunotoxic inorganic arsenic. Journal of Immunology, 177, 3019–3027. [DOI] [PubMed] [Google Scholar]
  70. Lemarie, A. , Morzadec, C. , Merino, D. , Micheau, O. , Fardel, O. , & Vernhet, L. (2006). Arsenic trioxide induces apoptosis of human monocytes during macrophagic differentiation through nuclear factor‐κB‐related survival pathway down‐regulation. The Journal of Pharmacology and Experimental Therapeutics, 316, 304–314. [DOI] [PubMed] [Google Scholar]
  71. Lengfelder, E. , Hofmann, W. K. , & Nolte, F. (2013). Management of elderly patients with acute promyelocytic leukemia: Progress and problems. Annals of Hematology, 92, 1181–1188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Leung, J. , Pang, A. , Yuen, W. H. , Kwong, Y. L. , & Tse, E. W. (2007). Relationship of expression of aquaglyceroporin 9 with arsenic uptake and sensitivity in leukemia cells. Blood, 109, 740–746. [DOI] [PubMed] [Google Scholar]
  73. Li, C. , Guan, T. , Gao, C. , Lin, Y. , Yan, G. , Zhu, M. , … Qi, Z. (2015). Arsenic trioxide inhibits accelerated allograft rejection mediated by alloreactive CD8+ memory T cells and prolongs allograft survival time. Transplant Immunology, 33, 30–36. [DOI] [PubMed] [Google Scholar]
  74. Li, J. , Zhao, L. , Zhang, Y. , Li, W. , Duan, X. , Chen, J. , … Li, B. (2017). Imbalanced immune responses involving inflammatory molecules and immune‐related pathways in the lung of acute and subchronic arsenic‐exposed mice. Env. Res, 159, 381–393. [DOI] [PubMed] [Google Scholar]
  75. Lin, Y. , Dai, H. , Su, J. , Yan, G. , Xi, Y. , Ekberg, H. , … Qi, Z. (2011). Arsenic trioxide is a novel agent for combination therapy to prolong heart allograft survival in allo‐primed T cells transferred mice. Transplant Immunology, 25, 194–201. [DOI] [PubMed] [Google Scholar]
  76. Lo, Y. H. , Huang, Y. W. , Wu, Y. H. , Tsai, C. S. , Lin, Y. C. , Mo, S. T. , … Lai, M. Z. (2013). Selective inhibition of the NLRP3 inflammasome by targeting to promyelocytic leukemia protein in mouse and human. Blood, 121, 3185–3194. [DOI] [PubMed] [Google Scholar]
  77. Lo‐Coco, F. , Avvisati, G. , Vignetti, M. , Thiede, C. , Orlando, S. M. , Iacobelli, S. , … Specchia, G. (2013). Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. The New England Journal of Medicine, 369, 111–121. [DOI] [PubMed] [Google Scholar]
  78. Lu, W. , Mehraj, V. , Vyboh, K. , Cao, W. , Li, T. , & Routy, J. P. (2015). CD4:CD8 ratio as a frontier marker for clinical outcome, immune dysfunction and viral reservoir size in virologically suppressed HIV‐positive patients. Journal of the International AIDS Society, 18, 20052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Luo, L. , Qiao, H. , Meng, F. , Dong, X. , Zhou, B. , Jiang, H. , … Sun, X. (2006). Arsenic trioxide synergizes with B7H3‐mediated immunotherapy to eradicate hepatocellular carcinomas. International Journal of Cancer, 118, 1823–1830. [DOI] [PubMed] [Google Scholar]
  80. Macoch, M. , Morzadec, C. , Fardel, O. , & Vernhet, L. (2013). Inorganic arsenic impairs differentiation and functions of human dendritic cells. Toxicology and Applied Pharmacology, 266, 204–213. [DOI] [PubMed] [Google Scholar]
  81. Macoch, M. , Morzadec, C. , Génard, R. , Pallardy, M. , Kerdine‐Römer, S. , Fardel, O. , & Vernhet, L. (2015). Nrf2‐dependent repression of interleukin‐12 expression in human dendritic cells exposed to inorganic arsenic. Free Radical Biology & Medicine, 88, 381–390. [DOI] [PubMed] [Google Scholar]
  82. Maier, N. K. , Crown, D. , Liu, J. , Leppla, S. H. , & Moayeri, M. (2014). Arsenic trioxide and other arsenical compounds inhibit the NLRP1, NLRP3, and NAIP5/NLRC4 inflammasomes. Journal of Immunology, 192, 763–770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Malloy, M. T. , McIntosh, D. J. , Walters, T. S. , Flores, A. , Goodwin, J. S. , & Arinze, I. J. (2013). Trafficking of the transcription factor Nrf2 to promyelocytic leukemia‐nuclear bodies: Implications for degradation of nrf2 in the nucleus. The Journal of Biological Chemistry, 288, 14569–14583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Mao, M. H. , Huang, H. B. , Zhang, X. L. , Li, K. , Liu, Y. L. , & Wang, P. (2018). Additive antitumor effect of arsenic trioxide combined with intravesical bacillus Calmette‐Guerin immunotherapy against bladder cancer through blockade of the IER3/Nrf2 pathway. Biomedicine & Pharmacotherapy, 107, 1093–1103. [DOI] [PubMed] [Google Scholar]
  85. Martelli, M. P. , Gionfriddo, I. , Mezzasoma, F. , Milano, F. , Pierangeli, S. , Mulas, F. , … Vetro, C. (2015). Arsenic trioxide and all‐trans retinoic acid target NPM1 mutant oncoprotein levels and induce apoptosis in NPM1‐mutated AML cells. Blood, 125, 3455–3465. [DOI] [PubMed] [Google Scholar]
  86. Mathews, V. , George, B. , Lakshmi, K. M. , Viswabandya, A. , Bajel, A. , Balasubramanian, P. , … Chandy, M. (2010). Single‐agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: Long‐term follow‐up data. Journal of Clinical Oncology, 28, 3866–3871. [DOI] [PubMed] [Google Scholar]
  87. Mei, Y. , Zheng, Y. , Wang, H. , Gao, J. , Liu, D. , Zhao, Y. , & Zhang, Z. (2011). Arsenic trioxide induces apoptosis of fibroblast‐like synoviocytes and represents antiarthris effect in experimental model of rheumatoid arthritis. The Journal of Rheumatology, 38, 36–43. [DOI] [PubMed] [Google Scholar]
  88. Morzadec, C. , Bouezzedine, F. , Macoch, M. , Fardel, O. , & Vernhet, L. (2012). Inorganic arsenic impairs proliferation and cytokine expression in human primary T lymphocytes. Toxicology, 300, 46–56. [DOI] [PubMed] [Google Scholar]
  89. Morzadec, C. , Macoch, M. , Robineau, M. , Sparfel, L. , Fardel, O. , & Vernhet, L. (2012). Inorganic arsenic represses interleukin‐17A expression in human activated Th17 lymphocytes. Toxicology and Applied Pharmacology, 262, 217–222. [DOI] [PubMed] [Google Scholar]
  90. Morzadec, C. , Macoch, M. , Sparfel, L. , Kerdine‐Romer, S. , Fardel, O. , & Vernhet, L. (2014). Nrf2 expression and activity in human T lymphocytes: Stimulation by T cell receptor activation and priming by inorganic arsenic and tert‐butylhydroquinone. Free Radical Biology & Medicine, 71, 133–145. [DOI] [PubMed] [Google Scholar]
  91. Niwa‐Kawakita, M. , Ferhi, O. , Soilihi, H. , Le Bras, M. , Lallemand‐Breitenbach, V. , & de The, H. (2017). PML is a ROS sensor activating p53 upon oxidative stress. The Journal of Experimental Medicine, 214, 3197–3206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Roboz, G. J. , Ritchie, E. K. , Carlin, R. F. , Samuel, M. , Gale, L. , Provenzano‐Gober, J. L. , … Kligfield, P. D. (2014). Prevalence, management, and clinical consequences of QT interval prolongation during treatment with arsenic trioxide. Journal of Clinical Oncology, 32, 3723–3728. [DOI] [PubMed] [Google Scholar]
  93. Rousselot, P. , Labaume, S. , Marolleau, J. P. , Larghero, J. , Noguera, M. H. , Brouet, J. C. , & Fermand, J. P. (1999). Arsenic trioxide and melarsoprol induce apoptosis in plasma cell lines and in plasma cells from myeloma patients. Cancer Research, 59, 1041–1048. [PubMed] [Google Scholar]
  94. Roychoudhuri, R. , Eil, R. L. , & Restifo, N. P. (2015). The interplay of effector and regulatory T cells in cancer. Current Opinion in Immunology, 33, 101–111. [DOI] [PubMed] [Google Scholar]
  95. Sage, A. P. , Minatel, B. C. , Ng, K. W. , Stewart, G. L. , Dummer, T. J. , Lam, W. L. , & Martinez, V. D. (2017). Oncogenomic disruptions in arsenic‐induced carcinogenesis. Oncotarget, 8, 25736–25755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Sahin, U. , Ferhi, O. , Jeanne, M. , Benhenda, S. , Berthier, C. , Jollivet, F. , … Lallemand‐Breitenbach, V. (2014). Oxidative stress‐induced assembly of PML nuclear bodies controls sumoylation of partner proteins. The Journal of Cell Biology, 204, 931–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Sakurai, T. , Ohta, T. , & Fujiwara, K. (2005). Inorganic arsenite alters macrophage generation from human peripheral blood monocytes. Toxicology and Applied Pharmacology, 203, 145–153. [DOI] [PubMed] [Google Scholar]
  98. Sanz, M. A. , Fenaux, P. , Tallman, M. S. , Estey, E. H. , Löwenberg, B. , Naoe, T. , … Mathews, V. (2019). Management of acute promyelocytic leukemia: Updated recommendations from an expert panel of the European LeukemiaNet. Blood, 133, 1630–1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Scherer, M. , & Stamminger, T. (2016). Emerging Role of PML Nuclear Bodies in Innate Immune Signaling. J Virol 90, 5850–5854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Schieber, M. , & Chandel, N. S. (2014). ROS function in redox signaling and oxidative stress. Current Biology, 24, R453–R462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Sengupta, M. , & Bishayi, B. (2002). Effect of lead and arsenic on murine macrophage response. Drug and Chemical Toxicology, 25, 459–472. [DOI] [PubMed] [Google Scholar]
  102. Shen, S. , Li, X. F. , Cullen, W. R. , Weinfeld, M. , & Le, X. C. (2013). Arsenic binding to proteins. Chemical Reviews, 113, 7769–7792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Shen, Z. X. , Chen, G. Q. , Ni, J. H. , Li, X. S. , Xiong, S. M. , Qiu, Q. Y. , … Chen, Y. (1997). Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL) II. Clinical efficacy and pharmacokinetics in relapsed patients. Blood, 89, 3354–3360. [PubMed] [Google Scholar]
  104. Shim, J. , Kennedy, R. H. , Weatherly, L. M. , Hutchinson, L. M. , Pelletier, J. H. , Hashmi, H. N. , … Gosse, J. A. (2016). Arsenic inhibits mast cell degranulation via suppression of early tyrosine phosphorylation events. Journal of Applied Toxicology, 36, 1446–1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Sikorski, E. E. , Mccay, J. A. , White, K. L. , Bradley, S. G. , & Munson, A. E. (1989). Immunotoxicity of the semiconductor gallium arsenide in female B6C3F1 mice. Toxicological Sciences, 13, 843–858. [DOI] [PubMed] [Google Scholar]
  106. Singer, M. , Trugnan, G. , & Chelbi‐Alix, M. K. (2011). Arsenic trioxide reduces 2,4,6‐trinitrobenzene sulfonic acid‐induced murine colitis via nuclear factor‐κB down‐regulation and caspase‐3 activation. Innate Immunity, 17, 365–374. [DOI] [PubMed] [Google Scholar]
  107. Singh, N. , Kumar, D. , Lal, K. , Raisuddin, S. , & Sahu, A. P. (2010). Adverse health effects due to arsenic exposure: Modification by dietary supplementation of jaggery in mice. Toxicology and Applied Pharmacology, 242, 247–255. [DOI] [PubMed] [Google Scholar]
  108. Soto‐Pena, G. A. , & Vega, L. (2008). Arsenic interferes with the signaling transduction pathway of T cell receptor activation by increasing basal and induced phosphorylation of Lck and Fyn in spleen cells. Toxicology and Applied Pharmacology, 230, 216–226. [DOI] [PubMed] [Google Scholar]
  109. Srivastava, R. K. , Li, C. , Chaudhary, S. C. , Ballestas, M. E. , Elmets, C. A. , Robbins, D. J. , … Athar, M. (2013). Unfolded protein response (UPR) signaling regulates arsenic trioxide‐mediated macrophage innate immune function disruption. Toxicology and Applied Pharmacology, 272, 879–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Srivastava, R. K. , Li, C. , Wang, Y. , Weng, Z. , Elmets, C. A. , Harrod, K. S. , … Athar, M. (2016). Activating transcription factor 4 underlies the pathogenesis of arsenic trioxide‐mediated impairment of macrophage innate immune functions. Toxicology and Applied Pharmacology, 308, 46–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Suzuki, S. , Arnold, L. L. , Pennington, K. L. , Kakiuchi‐Kiyota, S. , & Cohen, S. M. (2009). Effects of co‐administration of dietary sodium arsenite and an NADPH oxidase inhibitor on the rat bladder epithelium. Toxicology, 261, 41–46. [DOI] [PubMed] [Google Scholar]
  112. Tenorio, E. P. , & Saavedra, R. (2005). Differential effect of sodium arsenite during the activation of human CD4+ and CD8+ T lymphocytes. International Immunopharmacology, 5, 1853–1869. [DOI] [PubMed] [Google Scholar]
  113. Thomas‐Schoemann, A. , Batteux, F. , Mongaret, C. , Nicco, C. , Chéreau, C. , Annereau, M. , … Alexandre, J. (2012). Arsenic trioxide exerts antitumor activity through regulatory T cell depletion mediated by oxidative stress in a murine model of colon cancer. Journal of Immunology, 189, 5171–5177. [DOI] [PubMed] [Google Scholar]
  114. Tohyama, N. , Tanaka, S. , Onda, K. , Sugiyama, K. , & Hirano, T. (2013). Influence of anticancer agents on cell survival, proliferation, and CD4+CD25+Foxp3+ regulatory T cell‐frequency in human peripheral‐blood mononuclear cells activated by T cell‐mitogen. International Immunopharmacology, 15, 160–166. [DOI] [PubMed] [Google Scholar]
  115. VanDenBerg, K. R. , Freeborn, R. A. , Liu, S. , Kennedy, R. C. , Zagorski, J. W. , & Rockwell, C. E. (2017). Inhibition of early T cell cytokine production by arsenic trioxide occurs independently of Nrf2. PLoS ONE, 12, e0185579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Vega, L. , Montes de Oca, P. , Saavedra, R. , & Ostrosky‐Wegman, P. (2004). Helper T cell subpopulations from women are more susceptible to the toxic effect of sodium arsenite in vitro. Toxicology, 199, 121–128. [DOI] [PubMed] [Google Scholar]
  117. Vega, L. , Ostrosky‐Wegman, P. , Fortoul, T. I. , Diaz, C. , Madrid, V. , & Saavedra, R. (1999). Sodium arsenite reduces proliferation of human activated T‐cells by inhibition of the secretion of interleukin‐2. Immunopharmacology and Immunotoxicology, 21, 203–220. [DOI] [PubMed] [Google Scholar]
  118. Wan, Y. Y. , & Flavell, R. A. (2009). How diverse—CD4 effector T cells and their functions. Journal of Molecular Cell Biology, 1, 20–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Wang, L. , Hu, X. , Xu, Y. , & Liu, Z. (2016). Arsenic trioxide inhibits lung metastasis of mouse colon cancer via reducing the infiltration of regulatory T cells. Tumour Biology, 37, 15165–15173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Wang, L. , Wang, R. , Fan, L. , Liang, W. , Liang, K. , Xu, Y. , … Ye, Q. (2017). Arsenic trioxide is an immune adjuvant in liver cancer treatment. Molecular Immunology, 81, 118–126. [DOI] [PubMed] [Google Scholar]
  121. Wang, Z. Y. , & Chen, Z. (2008). Acute promyelocytic leukemia from highly fatal to highly curable. Blood, 111, 2505–2515. [DOI] [PubMed] [Google Scholar]
  122. Wei, Z. , Zhang, X. , Wang, J. , Wang, Y. , Yang, Z. , & Fu, Y. (2018). The formation of canine neutrophil extracellular traps induced by sodium arsenic in polymorphonuclear neutrophils. Chemosphere, 196, 297–302. [DOI] [PubMed] [Google Scholar]
  123. Xu, S. , Chen, J. , Wang, F. , Kang, X. , Lan, T. , Wang, F. , … Xing, J. (2010). Arsenic trioxide combined with co‐stimulatory molecule blockade prolongs survival of cardiac allografts in alloantigen‐primed mice. Transplant Immunology, 24, 57–63. [DOI] [PubMed] [Google Scholar]
  124. Xu, W. , Li, X. , Quan, L. , Yao, J. , Mu, G. , Guo, J. , & Wang, Y. (2018). Arsenic trioxide decreases the amount and inhibits the function of regulatory T cells, which may contribute to its efficacy in the treatment of acute promyelocytic leukemia. Leukemia & Lymphoma, 59, 650–659. [DOI] [PubMed] [Google Scholar]
  125. Yan, G. , Xi, Y. , Xu, S. , Lin, Y. , Chen, J. , Dai, H. , … Qi, Z. (2013). Inhibition of accelerated rejection mediated by alloreactive CD4+ memory T cells and prolonged allograft survival by arsenic trioxide. Immunological Investigations, 42, 438–454. [DOI] [PubMed] [Google Scholar]
  126. Yan, S. , Zhang, Q. Y. , Zhou, B. , Xue, L. , Chen, H. , Wang, Y. , & Zheng, S. S. (2009). Arsenic trioxide attenuated the rejection of major histocompatibility complex fully‐mismatched cardiac allografts in mice. Transpl. Proc, 41, 1855–1858. [DOI] [PubMed] [Google Scholar]
  127. Yu, H.‐S. , Chen, G.‐S. , Liao, W.‐T. , Chang, K.‐L. , & Yu, C.‐L. (2002). Arsenic induces tumor necrosis factor α release and tumor necrosis factor receptor 1 signaling in T helper cell apoptosis. The Journal of Investigative Dermatology, 119, 812–819. [DOI] [PubMed] [Google Scholar]
  128. Zhang, J. , Zhang, Y. , Wang, W. , Li, C. , & Zhang, Z. (2018). Double‐sided personality: Effects of arsenic trioxide on inflammation. Inflammation, 41, 1128–1134. [DOI] [PubMed] [Google Scholar]
  129. Zhang, M. , Qi, Y. , Li, H. , Cui, J. , Dai, L. , Frank, J. A. , … Chen, G. (2016). AIM2 inflammasome mediates Arsenic‐induced secretion of IL‐1 β and IL‐18. Oncoimmunology, 5, e1160182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Zhao, B. , Xia, J. J. , Wang, L. M. , Gao, C. , Li, J. L. , Liu, J. Y. , … Zhao, B. H. (2018). Immunosuppressive effect of arsenic trioxide on islet xenotransplantation prolongs xenograft survival in mice. Cell Death & Disease, 9, 408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Zhao, J. J. , Yang, S. W. , Zhang, F. , Yuan, X. L. , Zhu, Z. M. , Fang, B. J. , & Sony, Y. P. (2018). As2O3 up‐regulates the proportion of CD4+CD25+CD127 low Tregs in peripheral blood of patients with severe aplastic anemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi, 26, 854–858. [DOI] [PubMed] [Google Scholar]
  132. Zhou, L. F. , Zhu, Y. , Cui, X. F. , Xie, W. P. , Hu, A. H. , & Yin, K. S. (2006). Arsenic trioxide, a potent inhibitor of NF‐κB, abrogates allergen‐induced airway hyperresponsiveness and inflammation. Respiratory Research, 7, 146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Zhu, H. , Hu, J. , Chen, L. , Zhou, W. , Li, X. , Wang, L. , … Chen, Y. (2016). The 12‐year follow‐up of survival, chronic adverse effects, and retention of arsenic in patients with acute promyelocytic leukemia. Blood, 128, 1525–1528. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES