Flyer

Archives of Medicine

  • ISSN: 1989-5216
  • Journal h-index: 17
  • Journal CiteScore: 4.25
  • Journal Impact Factor: 3.58
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Awards Nomination 20+ Million Readerbase
Indexed In
  • Genamics JournalSeek
  • China National Knowledge Infrastructure (CNKI)
  • Directory of Research Journal Indexing (DRJI)
  • OCLC- WorldCat
  • Proquest Summons
  • Publons
  • Geneva Foundation for Medical Education and Research
  • Euro Pub
  • Google Scholar
  • Secret Search Engine Labs
Share This Page

Review Article - (2020) Volume 12, Issue 4

Therapeutic Role of Arsenic Trioxide in Cancer Protection

Archana Chaudhary and Rizwanul Haque*

Department of Biotechnology, School of Earth Biological & Environmental Sciences, Central University of South Bihar, Gaya, India

*Corresponding Author:
Dr. Rizwanul Haque
Associate Professor
Department of Biotechnology
School of Earth Biological & Environmental Sciences
Central University of South Bihar, Gaya, India
Tel: 7979054563
E-mail: rhaque@cub.ac.in

Received Date: April 06, 2020; Accepted Date: June 11, 2020; Published Date: June 18, 2020

Citation: Chaudhary A, Haque R (2020) Therapeutic Role of Arsenic Trioxide in Cancer Protection. Arch Med Vol. 12 Iss.3: 14 DOI: 10.36648/1989-5216.12.3.315

Copyright: © 2020 Chaudhary A, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Visit for more related articles at Archives of Medicine

Abstract

The growing frame of Arsenic is a significant public health concern now a day. Although arsenic is a well known carcinogen which leads to the induction of different types of cancers, it also been recently recommended as one of the most effectual novel anticancer agent for the treatment of various types of cancer including acute promyelocytic leukaemia (APL). However the exact metabolic mechanism induces due to arsenic and its compounds are still a question to be answer. The revival of this metalloid in treating APL is a unique story in cancer therapy. Based on these facts, Here we focused the effect of arsenic trioxide alone or its combination with other existing supplements in different types of cancer, and these strategies may enlightening in near future with respect to precious uses of arsenicals as medicines for treating cancer. Along with this it also requires to understand how arsenic emphasizes its toxic effects and how its implication leads to novel therapeutic advances. Hence there is an urgent need for better understanding of paradoxical effect of arsenic which may have a potential to become an appropriate treatment of various types of malignancies. Making concern in the area, in this review we focused its targeted pathways which would be helpful in shedding light on its actual role in relation to toxicity as well as in potential drug.

Keywords

Arsenic; Acute Promyelocytic Leukaemia (APL)

Introduction

Over the years, the relationship between arsenic and malignancy is much growing concern as millions of peoples are potential victims. Arsenic an environmental element, obtained a position in history, both as a recommended poison and as a miracle medicine [1]. However on the other hand inorganic arsenic (As2O3) is the most effective drug for the treatment of APL [2]. The mechanism of cytotoxic potential of arsenic and its methylated metabolites in eradicating cancer is sorely lacking. To describe the better understanding of the toxicity and carcinogenicity along with the use of arsenic in chemotherapy, caution is required considering the poor understanding of its various mechanisms of exerting toxicity.

Literature Review

To provide a deeper understanding of remedial role of arsenic in cancer treatment, in this review we mainly focus on how arsenic in combination with some existing compounds mediate metabolic pathways in its anticancer potential as well as how it possess its therapeutic role for the treatment of numerous malignancies?

Potential targets of Arsenic

Apart from various pathways arsenic imposes intense cellular alteration, including induction of apoptosis, reduction in rate of proliferation, suppress the process of angiogenesis and stimulation of differentiation [3,4]. Arsenic may induces its biological effects by interacting with closely spaced cysteine residues on critical cell proteins. Arsenic achieved a potential success especially in one type of cancer called APL. In majority of APL cases, it is characterized by the t (15: 17) translocation which lead to the formation of PML and RAR gene fusion. This fusion is a representative of (19,20) transcription factor [3]. The above protein helps in the blockage of gene which is responsible of myeloid differentiation. Gene sequences of PML depicts that it has cysteine rich region which helps in arsenic interaction. This PML protein usually localized in nuclear body present inside the nucleus [5]. The interaction of PML-RAR in leukemia leads to the disruption of nuclear bodies which ultimately dispersed the PML proteins into smaller fragments. Treatment with RA also blocks the myeloid differentiation by PML-RAR fusion which implements in ATRA therapy for APL [5]. Arsenic also results in the degradation of PML-RAR fusion protein and it was proved as a novel alternative for the treatment of ATR as in both RA-resistant as well as RA-sensitive APL patients it exhibits complete abrogation [6]. Arsenic trioxide influences the promotion of a nuclear protein which colocalizes with PML in nuclear bodies and represses transcription of a gene called Daxx [7]. Daxx which have a major role in modulation of death related genes transcription in Fas induced apoptosis [8]. So a slight increase in arsenic concentration directly influences PML within nuclear bodies and sufficient to trigger Daxx-dependent apoptosis. Arsenic and its compounds also affects the covalent modification of PML with SUMO-1 which is a ubiquitin like protein, so it involves in the augmentation of PML-containing nuclear bodies in the nucleus therefore it plays a lead role in proapoptotic signal transduction [9]. Hence an increasing dose of arsenic increases the SUMO-1 modification of PML-RAR and ultimately leads to apoptosis.

Response of Arsenic on cellular signaling pathways

Researchers around the country already reported that arsenic involve in pro-apoptotic pathways in several malignant cell lines that may be dependent on PML and P53. Arsenic trioxide upregulates the P53 expression in MBC-1, a B-cell lymphoma gastric cancer cells, which leads to apoptosis fallowed by caspase activation [10-12]. In human T-cell lymphotropic virus type 1 infected cells the increasing dose of arsenic trioxide results in the accumulation of P53, G1 phase arrest, increases the level of Cip1/p21 and p27KIP1, dephosphorylating of retinoblastoma protein and eventually leads to accumulation of P53 which induces apoptosis [13,14]. In human fibroblast cells, arsenite leads to the double strand breaks which also results in phosphorylation or upregulation of P53, so it also assist the increase in expression of P53 downstream proteins (P21 and others) [15,16]. Reports suggests that a arsenic treatment in certain extent insists P53 accumulation mainly due to the involvement of phosphatidylinositol-3-kinase related proteins, within an antaxia-telangiectasia mutated pathways [17-21] Bcl-2 have a major role in the regulation of arsenic mediated apoptosis, arsenic mainly upregulates p53, other growth arrest related genes and apoptosis while it cause downregulation of Bcl-2 in APL patients [22,23]. Arsenic modulates the binding of PML, Bax, p27KIP1 to nuclear bodies [3,24,25], along with PML containing cells, which synergistically move with IFNs (interferons) so that it prompt PML, to trigger cell death.

Effects of Arsenic

It has been demonstrated that sodium arsenite increases the activity of mutagenic components such as, c-Fos and c-Jun which eventually leads to accelerate the transcription factor- AP-1 (DNA binding activity of activator protein-1), simultaneously this AP-1 activates the JNKs which have a major role in in phosphorylation of many transcription factors and enhances the expression of immediate early genes [26,27]. Arsenic mainly activates JNKs pathway by directly inhibiting the constitutive JNK phosphatase. In human embryonic kidney cells arsenic compounds activates the JNK pathway by activating MEKK3and MEKK4 in other case without arsenic JNK activation requires activation of MEKK2 along with MEKK3 and MEKK4 [28]. Arsenic also influences the signaling pathways that are activated by increasing oxidative stress.

In bronchial epithelial cells, members of MAP kinase family which regulates the extra cellular signaling pathways also activates due to arsenic exposure [29,30]. Literature also describes that three PKC family isozymes arbitrates signal transduction because of arsenite introduction in an epidermal cell lines which finally results in AP-1 activation [31]. So it is now almost clear that arsenic compounds have role in in translocation of several PKC isoforms from cytosol to the plasma membrane, these enzymes mainly involves in mediating signal transduction and thus it may be use as a noble target for targeting biological effects of arsenic treatments. More discoveries are needed with specific inhibitors and dominant-negative mutant model helps in dissecting which MAP kinase proteins have major role in arsenic mediated action in particular cell types.

Tyrosine phosphatase

These enzymes possess vicinal thiols and hence being a potential target site for arsenic related compounds. Tyrosine phosphatase is reported as a molecular target for the activity of arsenic [32]. Some experiments also govern that due to arsenic posphotyrosine level increases but this activation causes no changes in level of tyrosine phosphatase activity.

Role in apoptosis

Arsenic trioxides have major role in caspases activation recent research on myeloma cells shows that caspases-9 is mainly activated in arsenic mediated apoptosis in combination with dexamethasone where as in neuroblastoma cell lines and in myeloid leukemia cells arsenic trioxides mediate apoptosis by activating caspases-3 the ultimate mechanism by which arsenic promotes apoptosis may be by inhibiting telomerase activity [33,34]. In NB4 cells arsenic trioxides resulted in downregulation of telomerase genes and it activity [35,36] this may be due to immediate response of arsenic-trioxide on transcription factors such as Sp1 and Myc.

Arsenic and ROS

Arsenic and its related compounds disturb the natural oxidation and oxidative reduction equilibrium by regulating various pathways involves in multiple redox reactions with intimate oxidants and other cellular antioxidant systems. As arsenic have high affinity for thiol groups, proteins with approachable as well as firmly spaced thiol moieties with high thiol disulphide oxidation potentials may be redox sensitive and redox regulation distinctly mediates important cell functions. Arsenic exerts it’s both therapeutic and toxic effects by targeting redox-sensitive enzyme and proteins [37]. Endogenous glutathione and thioredoxin plays a crucial role in regulating the redox signaling and thus protecting cells from damaging effects of arsenic studies also suggests that arsenic paradoxically shares many properties of tumor promotors as effects the several redox sensitive signaling molecules such as AP-1, P52, P21 as well as S-nitro thiols which results in the dysregulation of various cell signaling and gene expression [38].

Other cellular targets of arsenic

Several reports suggests that arsenic below 10 micro molar concentration leads to the blockage of ligand binding to glucocorticoid receptor in a region that contains a dithiol critical to proper ligand binding [39]. It also inhibits the interaction of estradiol and estrogen receptor. An arsenic compound also activates the estrogen receptor and therefore amplifies the estrogen-dependent genes. Studies also demonstrated that arsenic may acts as an environmental estrogen, as in many cell types including MCF-7 arsenic induces apoptosis, this activation also inhibited by a complete antiestrogen [40]. Arsenic also acts as an antiangiogenic [41] model by disrupting the reciprocal stimulatory loop between leukemic cells and endothelial cells [42]. It negatively affect the tumour cell growth by inhibiting angiogenesis. Due to high rate of sulfhydryl content in tubulin cytoskeleton has been reported as a potential cellular target for arsenic [43]. Pyruvate dehydrogenase was earlier reported as important molecular targets of arsenic trioxide which results in the decreased gluconeogenesis and hypoglycemia. Arsenic trioxide at 5micromolar directly inhibits the activity of pyruvate dehydrogenase in vitro.

Synergist Effects of Arsenic with other Natural Compounds in Cancer Protection

Colon cancer

Arsenic in its inorganic form i.e., arsenic trioxide resist the activation of rapamycin: NF-kB (nuclear factor kappa-lightchain- enhancer of activated B cells) is a protein complex that controls transcription of DNA, cytokine production and cell survival, these inhibition ultimately leads to the blocking of phosphorylation as well as degradation of IkB-alpha in HCT-116 cells [44]. This arsenic induced blocked phosphorylation of IkB-alpha leads to apoptosis induction in colon cancer and thus possesses its therapeutic role in cancer protection. Apart from this several reports demonstrated that the combinational therapy of arsenic trioxide and PI3K inhibitor LY294002 have a drastic role in tumor reduction in colon cancer cell lines [45].

Prostate cancer

Arsenic basically inhibits Akt/mTOR signaling pathway which is an intracellular signaling pathway important in regulating the cell cycle [46]. Therefore, it is directly related to its anticancer approach by inhibiting the proliferation of PC-3 by downregulating the Hh signaling pathway and the antitumor effect was further enhanced by a classic Hh pathway inhibitor cyclopamine [47,48]. Researchers applied the synergistic therapy by combining ATO-mTOR inhibitor RAD001 which shows the induced apoptosis and autophagy rate in prostate cancer cells [49]. The induction in autophagy mediated cell death was basically linked by increased Beclin 1mRNAs stability [50,51]. So the combinational therapy of ATO-RAD001 shows much significant role in cancer suppression and tumor proliferation than monotherapy without any enhancement in weight loss.

Oral cancer

This is the most common neoplasm of neck and head which is highly associated with poor prognosis, despite of large number of treatment options are available now days. Arsenic trioxide was reported as drug additive with radiotherapy and a platinum-based anti-neoplastic drug cisplatin (CDDP), these combinations are the most appropriate therapies for oral cancer [52-54]. Several reports demonstrated that ATO +dithithreitol (DTT) leads to the increased rate of proappoptotic molecules Bax and Bak and simultaneously reduce the Bcl-2 and p53 [55], which ultimately leads to the significant cell death of oral cancer cells.

Ovarian cancer

If we talk about the rate of mortality then it is highest for ovarian cancer among all malignant tumours of female genital organs. ATO in combination with CDDP (Cisplatin a chemotherapy medication used to treat a number of cancers) proves to be much effective and it enhances the cytotoxic effect CDDP alone by many folds [56]. Many studies also notified that the effectiveness of ATO can be enhanced with buthionine sulfoximine and ascorbic acid by mediating GSH depletion and oxidative stress-related pathway in cell killing or antitumor induction [57,58].

Cervical cancer

Cervical cancer is one of the most common cancer among females worldwide, which is generally treated with radiotherapy and a combined therapy with chemotherapeutics, such as platinum-based drugs. ATO leads to the suppression of MMP-9 (Matrix metallopeptidase 9 which involves in the degradation of extracellular matrix) and thus this ATO mediated suppression decreases radiationaccelerated lung metastases [59]. Both in vitro and in vivo data depicts that ATO-radiation treatment shows more than expected beneficial role in anticancer effect on cervical cancer. Apart from this ATO also enhances the translocation of apoptotic regulators that is Bax protein and also increases the phosphorylation of Bcl-2, which ultimately leads to the activation of MAPKs as well as JNK pathways [60]. ROS has a major role in ATO-radiation-induced apoptosis [61]. Beside ATO the role of tetra arsenic oxide in exerting anticancer effects on cervical cancer cells has also been demonstrated. These oxides of arsenic in combination with CDDP leads to the maximum reduction of tumour growth specially in case of cervical cancer [62], this therapy also drastically increases the number of apoptotic cells and hence possess a novel anticancer role.

Breast cancer

This type of cancer is one of the leading causes of cancer related deaths among females worldwide. According to various reports it has been proven that ATO leads to reduces the expression of DNA methyltransferase-1 and induce the expression of estrogen receptor alpha, whose expression has been recognized to increase disease-free survival and indicate an overall better prognosis [63]. Surprisingly ATO and antiestrogen tamoxifen (TAM) therapy effectively suppresses tumour growth of human breast cancer cell line MDA-MB-435 S both in vitro and in vivo [64]. Several reports demonstrated that ATO enhances the anticancer activity of rapamycin (a specific mTOR inhibitor) as well as enhances ATO-melatonininduced apoptosis in vivo [65]. Various plants growth regulators such as cotylenin A (CN-A) when applied after coincubation with ATO shows tremendous anticancer activity against breast cancer cells in vitro [66]. Other form of arsenic monomethylarsonous (MMaIII ) and dimethylarsinous acid (DMA III) is more cytotoxic than ATO towards breast cancer.

HCC

Hepatocellular carcinoma (HCC) is the most common primary liver malignancy, and the sixth most common types of cancer worldwide. Sorafenib (mutikinase inhibitors) in combination with ATO leads to induced activation of Akt [67] or its downstream factors, including glycogen synthase kinase -3beta, mTOR, ribosomal protein S6 kinase, and eukaryotic translation initiation factors 4E binding protein and extent the survival rate of patient with advanced HCC [68]. ATO also enhances the anticancer potential of metformin, survivin mutant (T34A), shikonin and andrographolide in HCC cells [69].

Lung cancer

Lung cancer is known for the most common type of cancer worldwide. A nonsteroidal anti-inflammatory drug of the arylalkanoic acid like sulindic in combination with ATO leads to the induction of apoptosis in lung cell lines [70]. This synergistic approach of sulindic and ATO mainly targets mitochondrial pathway, the NF-kB pathways as well as also mediate p53-induced downregulation of survivin and hence leads to the induction of apoptosis in lung cancer cell lines [71]. Studies also demonstrated that ATO- sulindac combinational therapy induces augmentation of cytotoxicity in both human non-small cell lung cancer cell lines (A549) by mediating ROS-induced MAPK phosphorylation via c-jun NH2- terminal kinase-dependent Bcl-xL phosphorylation [72]. Apart from this other structural analog of sulindac i.e., the nonselective cyclooxygenase inhibitors for example indomethacin was also depicts to intensify the arsenic persuaded cytotoxic outcome in A549 cells by arbitrating the exhilaration of ERK/or p38 MAPKs [73]. Recent studies also reported that the combination of fibroblast growth factor receptor (FGFR) inhibitor PD173074 and ATO suppresses tumour proliferation in lung squamous cell carcinoma (SCC)cell line SK-MES-1, here this therapy reduces FGFR1,Akt,Src.c-Raf and Erk by mediating proteasomal degradation [74]. ATO leads to ROS-mediated ER stress and mitochondrial dysfunction and ultimately leads to apoptosis in A549 cells, when it combines with resveratrol.

Gastric cancer

Anti-apoptotic drug such as ABT-737 which inhibit Bcl-2 as well as Bcl-Xl when synergistically mix up with ATO leads to the drastic suppression in the proliferation rate of human gastric cancer cell lines SGC7901 and MGC-803 [75,76].

Pancreatic cancer

Studies reported that sesquiterpene lactone such as parthenolide from the medicinal herb feverfew, enhances apoptosis of human pancreatic cancer cell lines PANC-1 and BxPC-3 by mediating ROS generation and subsequent caspase activation [77]. ATO and parthenolide significantly reduces tumor proliferation rate in PANC-1cell line. Studies also governs that ATO alone have limited efficacy on cytotoxicity in pancreatic ductal adenocarcinoma because of the highcellular ROS scavenging activity [78].

Glioblastoma

Arsenic trioxide increases expression of death receptor 5 (DR5), a death receptor of tumor necrosis factor-related apoptosis inducing ligand (TRAIL) in human glioma cell lines [79]. The combination of ATO and TRAIL synergistically reduces the survival of glioma cells. Radiation therapy in combination with ATO positively regulates the autophagy effects in U118- MG cells by enhancing the mitotic arrest and modulating PI3K/Akt and ERK1/2 signalling pathways [80,81].

ATO induced apoptosis is markedly enhanced by using polyunsaturated fatty acid docosahexaenoic acid (DHA) in 12 different ATO- resistant solid tumour cell lines including breast, ovarian, colon, prostate, cervical and pancreatic cancer, without any toxic effect on normal skin fibroblasts, human microvascular endothelial cells and peripheral blood mononuclear cells derived from healthy donors [82].

Promyelocytic leukaemia (APL )

Arsenic trioxide is a chemotherapy drug and is also called Trisenox or ATO. It is a treatment for a type of acute myeloid leukemia called Acute Promyelocytic Leukemia (APL). Researchers are also looking into it as a treatment for other types of cancer. Several reports suggested that arsenic trioxide in amalgamation with all trans retinoic acid resist the cells proliferation rate as well as also induces in vitro apoptosis in verities of cell types such hepatoma, breast cancer and lung cancer cells [83,84]. Apart from this Kryeziu et al. also reported that the combined treatment of erlotinib which is a selective EGFR inhibitor with ATO helps in accumulation of DNA damage by inhibiting EGFR-mediated DA double-strand break repair in mesothelioma, hepatocellular carcinoma, colorectal carcinoma, thyroid carcinoma and cervix carcinoma in vitro [85].

Hematological Malignancies

Acute promyelocytic leukemia (APL)

Different reports demonstrated that the combinational therapy of As4S4 and As3+ induces the degradation of PML/RAR α oncoproteins and subsequent apoptosis [86]. Other reports emphasized that the addition of ATRA-ATO leads to increased rate of differentiation of APL cells by inducing Src family kinase inhibitor PP2 [87]. ATO shows the antileukemic activity and this property of ATO is enhanced by the combination therapy with- granulocyte monocytes colony stimulation factor, a noncalcemic vitamin D analog 19-Nor-125 (OH)2D2,N- (beta-Elemene-13-yl)tryptophan methyl, Gefitiniba selective inhibitor of epidermal growth factor receptor (EGFR) gefitinib, and high-dose vitamin C (ascorbic acid), all of which enhance ATO-induced differentiation of APL cells [88-90].

Acute myeloid leukemia (AML)

When juvenile leukocytes are augmented in an early stage of differentiation this condition is defined as acute myeloid leukemia, which is a malignant disease of bone marrow. Reports suggested that mutation in FLT3 in any AML patient have shorter internal tandem duplication (FLT3-ITTD) and leads to the disease free survival [91,92]. Researchers demonstrated that the combined treatment of ATO and FLT3 specific inhibitor AG1296 induces apoptosis in FLT3-ITD –positive cells, but not in FLT3 wild type cells. Combination of ATRA+ATO exerts synergistic cytotoxicity against FLT3-ITD AML cells via coinhibition of FLT3 signaling pathways in many APL patients [93]. These combination also induces apoptosis of NPM1- mutated AML cells y targeting nucleophosmin (NPMI) oncoprotein, whose mutation possibly represents a therapeutic targets because of high frequency in 30% AML [94]. BSO with ATO also represents anticancer effects by reducing ROS generation in AML cells [95]. In addition of BSO and ATO, dichloroacetate [96], azacytidine [97], rapamycin [98] and aclacinomycin A [99] induces apoptosis in AML cells.

Multiple myeloma (MM)

In MM cell lines relevant doses of ascorbic acid decreases GSH levels and enhances ATO-mediated cell death [100]. ATO + melphalan + ascorbic acid is one of the attractive alternative for refractory MM patients [101]. Several proteasome inhibitors such as bortezomib (BOR) and carfilzomib along with this immunomodulatory drugs such as thalidomide, lenalidomide (LEN), pomalidomide have best effect in MM patient’s survival rate [102]. Reports also given a clue that enhanced cytotoxicity of ATO-BOR is associated with augmented STAT3 inhibition, JNK activation and upregulation of Bim, p21, p27, p53 as well as downregulation of Bcl-2 [103]. ATO also leads to the upregulation of cereblon which is an antimyeloma target of LEN [104], and hence increases the sensitivity of MM cells, and this sensitivity enhances by combination of ATO with vitamin E analog Trolox [105] which is a MEK inhibitor PD325901 [106] a natural quinoid diterpene cryptotanshinone and a phytochemical sulforaphane [107,108].

Lymphoma

Researchers proven that ATO with BOR shows an anticancer effects in mantle cell lymphoma which is an incurable B-cell non-Hodgkin lymphoma [109,110]. Cucurbitacin B, from Trichosanthes kirilowii maxim, in combination with ATO synergistically enhances apoptosis rate by inhibiting STAT3 phosphorylation in Burkitts lymphoma cell lines both in vitro and in vivo [111,112].

Discussion and Conclusion

Arsenic and its related compounds twins several mechanism leads to various signal transduction pathways to effect various cellular response such as growth inhibition, induction of apoptosis, angiogenesis inhibition and many more. Here we shown that arsenical compounds, alone or in combination with other anticancer therapeutics such as molecular targeted drugs, radiation, chemotherapy helps in the induction of apoptosis in several cancer cell types. Now a days due to the advancement in technologies and surpluses of anticancer therapies there is a chance of targeting single molecules and signaling pathways as well as single cellular biological processes may generates a different malignant population of cancer cells, some of which may acquire a certain drug resistance. Therefore a novel therapeutic agents or alternatives are urgently required to overcome drug resistance and improve both the disease outcomes and the quality of life for patients with cancer. Large number clinical trials under way in several types of malignancies and solid tumour to investigate the therapeutic potential of arsenic. As arsenic in its low concentration proved to have therapeutic potential which already governed by APL and hence it is recommended as a possible promise for preclinical model of other types of cancer too. Moreover studies are required to understand the relationship between apoptosis induction and genetic changes in cancer cells due to arsenic which may enhances the studies in selectivity for cancer treatment. Further advancements are required to understand the synergistic anticancer action regarding ATO-based combination therapeutic to develop a novel combined therapy for cancer.

Acknowledgements

First author acknowledge the receipt of financial support under the fellowship CSIR-UGC (ARCHANA/MAY2018-383981) from university grant commission.

28343

References

  1. Adams SL (2008) Arsenic: Medicinal double-edged sword. HemOnc Today, November 10, 2008.
  2. Alimoghaddam K (2014) A review of arsenic trioxide and acute promyelocytic leukemia. Int J Hematol Oncol Stem Cell Res 8: 44–54.
  3. Jr Miller WH, Schipper HM, Lee JS, Singer J, Axman SW (2002) Mechanism of action of arsenic trioxide. Cancer Res 62: 3893-3903.
  4. Antman KH (2001) Introduction : the history of arsenic trioxide in cancer therapy. The Oncologist 6: 1-2.
  5. Davison K, Mann KK,  Miller WH (2002) Arsenic trioxide: Mechanism of action. Semin Hematol 39: 3-7.
  6. Jing Y (2004) The PML-RARalpha fusion protein and targeted therapy for acute promyelocytic leukemia. Leuk Lymphoma 45: 639-648.
  7. Zhang TD, Chen GQ, Wang ZG, Wang ZY, Chen SJ, et al. (2001) Arsenic trioxide, a therapeutic agent for APL. Oncogene 20: 7146-7153.
  8. Lallemand-Breitenbach V, de Thé, H (2010) PML nuclear bodies. Cold Spring Harbor Perspectives in Biology 2: a000661.
  9. Bernardi R, Pandolfi PP (2007) Structure, dynamics and functions of promyelocytic leukaemia nuclear bodies. Nat Rev Mol Cell Biol 8: 1006-1016
  10. Percherancier Y, Germain-Desprez D, Galisson F, Mascle XH, Dianoux L, et al. (2009) Role of SUMO in RNF4-mediated promyelocytic leukemia protein (PML) degradation: sumoylation of PML and phospho-switch control of its SUMO binding domain dissected in living cells. J Biol Chem 284: 16595-16608.
  11. Kang T, Ge M, Wang R, Zhen T, Xiuli Z, et al. (2019) Arsenic sulfide induces RAG1-dependent DNA damage for cell killing by inhibiting NFATc3 in gastric cancer cells. J Exp Clin Cancer Res 38: 487.
  12. Zhi-Bin M, Hong-Yu X, Miao J, You-Lin Y, Lian-Xin L, et al. (2014) Arsenic trioxide induces apoptosis of human gastrointestinal cancer cells. World J Gastroenterol 20: 5505–5510.
  13. Zhong L, Xu F, Chen F (2018) Arsenic trioxide induces the apoptosis and decreases NF-κB expression in lymphoma cell lines. Oncology Letters 16: 6267–6274.
  14. Yih LH, Lee TC (2000) Arsenite induces p53 accumulation through an ATM-dependent pathway in human fibroblasts. Cancer Research 60: 6346-52.
  15. John Lisa, Sauter ER, Herlyn M, Litwin S, Adler-Storthz K (2000) Endogenous p53 gene status predicts the response of human squamous cell carcinomas to wild-type p53. Cancer Gene Ther 7: 749-756.
  16. Ishitsuka K, Hanada S, Uozumi K, Utsunomiya A, Arima T (2000) Arsenic Trioxide and the Growth of Human T-cell Leukemia Virus Type I Infected T-cell Lines. Leuk Lymphoma 37: 649-655.
  17. Williams AB, Schumacher B (2016) p53 in the DNA-Damage-Repair Process. Cold Spring Harbor Perspectives in Medicine 6: a026070.
  18. Yu Y, Zhang D, Huang H, Li J, Zhang M, et al. (2014) NF-κB1 p50 promotes p53 protein translation through miR-190 downregulation of PHLPP1. Oncogene 33: 996-1005.
  19. Levine AJ (1997) p53, the cellular gatekeeper for growth and division. Cell 88: 323-331.
  20. Carr MI, Jones SN (2016) Regulation of the Mdm2-p53 signaling axis in the DNA damage response and tumorigenesis. Transl Cancer Res 5: 707-724.
  21. Shiloh Y (2003) ATM and related protein kinases: safeguarding genome integrity. Nat Rev Cancer 3: 155-168.
  22. Laura Z, Domenico D, Giacomo B (2014) CHK2 kinase in the DNA damage response and beyond. J Mol Cell Biol 6: 442-457.
  23. Zheng Y, Zhou M, Ye A, Li Q, Bai Y, Zhang Q (2010) The conformation change of Bcl-2 is involved in arsenic trioxide-induced apoptosis and inhibition of proliferation in SGC7901 human gastric cancer cells. World J Surg Oncol 8: 31.
  24. Kumar S, Brown A, Tchounwou PB (2018) Trisenox disrupts MDM2-DAXX-HAUSP complex and activates p53, cell cycle regulation and apoptosis in acute leukemia cells. Oncotarget 9: 33138-33148.
  25. Liu L, Li Y, Xiong X, Qi K, Zhang C, et al. (2016) Low dose of arsenic trioxide inhibits multidrug resistant-related P-glycoprotein expression in human neuroblastoma cell line. Int J Oncol 49: 2319-2330.
  26. Lam S, Li Y, Zheng C, Leung LL, Ho JC (2014) E2F1 downregulation by arsenic trioxide in lung adenocarcinoma. Int J Oncol 45: 2033-2043.
  27. Porter AC, Fanger GR, Vaillancourt RR (1999) Signal transduction pathways regulated by arsenate and arsenite. Oncogene 18: 7794-7802.
  28. Wu W, Graves LM, Jaspers I, Devlin RB, Reed W, et al. (1999) Activation of the EGF receptor signaling pathway in human airway epithelial cells exposed to metals. Am J Physiol 277: L924-L931.
  29. Chen NY, Ma WY, Huang C, Ding M, Dong Z (2000) Activation of PKC is required for arsenite-induced signal transduction. J Environ Pathol Toxicol Oncol 19: 297-305.
  30. Koul HK, Pal M, Koul S (2013) Role of p38 MAP Kinase Signal Transduction in Solid Tumors. Genes Cancer 4: 342-359.
  31. Li D, Wei Y, Xu S, Niu Q, Zhang M, et al. (2018) A systematic review and meta-analysis of bidirectional effect of arsenic on ERK signaling pathway. Mol Med Rep 17: 4422-4432.
  32. Cosentino-Gomes D, Rocco-Machado N, Meyer-Fernandes JR (2012) Cell signaling through protein kinase C oxidation and activation. Int J Mol Sci 13: 10697-10721.
  33. Shen S, Li XF, Cullen WR, Weinfeld M, Le XC (2013) Arsenic binding to proteins. Chem Rev 113: 7769-7792.
  34. Hayashi T, Hideshima T, Akiyama H, Richardson P, Schlossman RL, et al. (2002) Arsenic trioxide inhibits growth of human multiple myeloma cells in the bone marrow microenvironment. Molecular Cancer Therapeutics 1: 851-860.
  35. Ishitsuka K, Ikeda R, Suzuki S, Ohno N, Utsunomiya A, et al. (1999) The inductive pathways of apoptosis and G1 phase accumulation by arsenic trioxide in an adult T-cell leukemia cell line, MT-1. Blood 94: 263b.
  36. Xu W, Wang Y, Tong H, Qian W, Jin J (2014) Downregulation of hTERT: an important As2O3 induced mechanism of apoptosis in myelodysplastic syndrome. PloS One 9: e113199.
  37. Chou WC, Hawkins AL, Barrett JF, Griffin CA, Dang CV (2001) Arsenic inhibition of telomerase transcription leads to genetic instability. The Journal of Clinical Investigation 108: 1541-1547.
  38. Chen YC, Lin-Shiau SY, Lin JK (1998) Involvement of reactive oxygen species and caspase 3 activation in arsenite-induced apoptosis. J Cell Physiol 177: 324-333.
  39. Pace C, Dagda R, Angermann J (2017) Antioxidants Protect against Arsenic Induced Mitochondrial Cardio-Toxicity. Toxics 5: 38.
  40. Telford WG, Fraker PJ (1997) Zinc reversibly inhibits steroid binding to murine glucocorticoid receptor. Biochem Biophys Res Commun 238: 86-89.
  41. Stoica A, Pentecost E, Martin MB (2000) Effects of arsenite on estrogen receptor-α expression and activity in MCF-7 breast cancer cells. Endocrinology 141: 3595-3602.
  42. Roboz GJ, Dias S, Lam G, Lane WJ, Soignet SL, et al. (2000) Arsenic trioxide induces dose- and time-dependent apoptosis of endothelium and may exert an antileukemic effect via inhibition of angiogenesis. Blood 96: 1525-1530.
  43. Lew YS, Brown SL, Griffin RJ, Song CW, Kim JH (1999) Arsenic trioxide causes selective necrosis in solid murine tumors by vascular shutdown. Cancer Res 59: 6033-6037.
  44. Li YM, Broome JD (1999) Arsenic targets tubulins to induce apoptosis in myeloid leukemia cells. Cancer Res 59: 776-780.
  45. Felix K, Manna SK, Wise K, Barr J, Ramesh GT (2005) Low levels of arsenite activates nuclear factor-kappaB and activator protein-1 in immortalized mesencephalic cells. J Biochem Mol Toxicol 19: 67-77.
  46. Cai X, Yu K, Zhang L, Li Y, Li Q, et al. (2015) Synergistic inhibition of colon carcinoma cell growth by Hedgehog-Gli1 inhibitor arsenic trioxide and phosphoinositide 3-kinase inhibitor LY294002. Onco Targets Ther 8: 877-883.
  47. Chen QY, Costa M (2018) PI3K/Akt/mTOR signaling pathway and the biphasic effect of arsenic in carcinogenesis. Mol Pharmacol 94: 784-792.
  48. Bao C, Kramata P, Lee HJ, Suh N (2018) Regulation of Hedgehog signaling in cancer by natural and dietary compounds. Mol Nutr Food Res 62: 10.
  49. Xiong Y, Guo Y, Gao Y, Li S, Dai Z, et al. (2015) Synergism between arsenic trioxide and cydopamine in the inhibition of PC3 cell survival via the Hedgehog signaling pathway. Neoplasma p. 62.
  50. Tai S, Xu L, Xu M, Zhang L, Zhang Y, et al. (2017) Combination of Arsenic trioxide and Everolimus (Rad001) synergistically induces both autophagy and apoptosis in prostate cancer cells. Oncotarget 8: 11206-11218.
  51. Kang R, Zeh HJ, Lotze MT, Tang D (2011) The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ 18: 571-580.
  52. Menon MB, Dhamija S (2018) Beclin 1 Phosphorylation at the Center of Autophagy Regulation. Front Cell Dev Biol 6: 137.
  53. Dasari S, Tchounwou PB (2014) Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol 740: 364-378.
  54. Ning Z, Wu ZM, McGowan E, Shi J, Zu-Bei H, et al. (2009) Arsenic trioxide and cisplatin synergism increase cytotoxicity in human ovarian cancer cells: Therapeutic potential for ovarian cancer. Cancer Sci 100: 2459-64.
  55. Florea AM. Büsselberg D (2011) Cisplatin as an anti-tumor drug: cellular mechanisms of activity, drug resistance and induced side effects. Cancers 3: 1351-1371.
  56. Tsai CW, Yang MD, Hsia TC, Chang WS, Hsu CM, Hsieh YH, et al. ( 2017) Dithiothreitol enhanced arsenic-trioxide-induced cell apoptosis in cultured oral cancer cells via mitochondrial dysfunction and endoplasmic reticulum stress. Environ Toxicol 32: 17-27.
  57. Zhang N, Wu ZM, McGowan E, Shi J, Hong ZB, et al. ( 2009) Arsenic trioxide and cisplatin synergism increase cytotoxicity in human ovarian cancer cells: Therapeutic potential for ovarian cancer. Cancer Sci 100: 2459-2464.
  58. Liu N, Tai S, Ding B, Thor RK, Bhuta S, et al. ( 2012) Arsenic trioxide synergizes with everolimus (Rad001) to induce cytotoxicity of ovarian cancer cells through increased autophagy and apoptosis. Endocr Relat Cancer 19: 711-723.
  59. Ong PS, Chan SY, Ho PC (2011) Differential augmentative effects of buthionine sulfoximine and ascorbic acid in As2O3 -induced ovarian cancer cell death: Oxidative stress-independent and -dependent cytotoxic potentiation. Int J Oncol 38: 1731-1739.
  60. Wei LH, Lai KP, Chen CA, Chia-Hsien C, Yun-Ju H, et al. ( 2005) Arsenic trioxide prevents radiation-enhanced tumor invasiveness and inhibits matrix metalloproteinase-9 through downregulation of nuclear factor kappaB. Oncogene 24: 390-398.
  61. Kang YH, Lee SJ (2008) Role of p38 MAPK and JNK in enhanced cervical cancer cell killing by the combination of arsenic trioxide and ionizing radiation. Oncol Rep 20: 637-643.
  62. Kim YW, Bae SM, Battogtokh G, Bang HJ, Ahn WS. (2012) Synergistic anti-tumor effects of combination of photodynamic therapy and arsenic compound in cervical cancer cells: In vivo and in vitro studies. PLoS One 7: e38583.
  63. Byun JM, Jeong DH, Lee DS, Kim JR, Park SG, Kang MS, et al. (2013) Tetraarsenic oxide and cisplatin induce apoptotic synergism in cervical cancer. Oncol Rep 29: 1540-1546
  64. Zhang W, Wang L, Fan Q, Xinai Wu, Feng Wang, et al. ( 2011) Arsenic trioxide re-sensitizes ERα-negative breast cancer cells to endocrine therapy by restoring ERα expression in vitro and in vivo. Oncol Rep 26: 621-628.
  65. Liu H, Tao X, Ma F, Qiu J, Wu C, Wang M ( 2012) Radiosensitizing effects of arsenic trioxide on MCF-7 human breast cancer cells exposed to 89 strontium chloride. Oncol Rep 28: 1894-1902.
  66. Guilbert C, Annis MG, Dong Z, Siegel PM, Miller WH Jr, et al. (2013) Arsenic trioxide overcomes rapamycin-induced feedback activation of AKT and ERK signaling to enhance the anti-tumor effects in breast cancer. PLoS One 8: e85995.
  67. Kasukabe T, Okabe-Kado J, Kato N, Honma Y, Kumakura S ( 2015) Cotylenin A and arsenic trioxide cooperatively suppress cell proliferation and cell invasion activity in human breast cancer cells. Int J Oncol 46: 841-848.
  68. Zhai B, Jiang X, He C, Dali Z, Lixin M, et al. (2015) Arsenic trioxide potentiates the anticancer activities of sorafenib against hepatocellular carcinoma by inhibiting Akt activation. Tumour Biol 36: 2323-2334.
  69. Chen C, Zhang Y, Wang Y, Huang D, Xi Y, Qi Y (2011) Genistein potentiates the effect of arsenic trioxide against human hepatocellular carcinoma: Role of Akt and nuclear factor-κB. Cancer Lett 301: 75-84.
  70. Huang A, Yue D, Liao D, Cheng L, Ma J, et al. (2016) Survivin T34A increases the therapeutic efficacy of arsenic trioxide in mouse hepatocellular carcinoma models. Oncol Rep 36: 3283-3290.
  71. Jiang TT, Brown SL, Kim JH (2004) Combined effect of arsenic trioxide and sulindac sulfide in A549 human lung cancer cells in vitro. J Exp Clin Cancer Res 23: 259-262.
  72. Jin HO, Yoon SI, Seo SK, Hyung-Chahn L, Sang-Hyeok W, et al. (2006) Synergistic induction of apoptosis by sulindac and arsenic trioxide in human lung cancer A549 cells via reactive oxygen species-dependent down-regulation of survivin. Biochem Pharmacol 72: 1228-1236.
  73. Han YH, Kim SZ, Kim SH, Park WH (2008) Induction of apoptosis in arsenic trioxide-treated lung cancer A549 cells by buthionine sulfoximine. Mol Cells 26: 158-164.
  74. Li H, Zhu X, Zhang Y, Xiang J, Chen H (2009) Arsenic trioxide exerts synergistic effects with cisplatin on non-small cell lung cancer cells via apoptosis induction. J Exp Clin Cancer Res 28: 110.
  75. Lam SK, Leung LL, Li YY, Zheng CY, Ho JC (2016) Combination effects of arsenic trioxide and fibroblast growth factor receptor inhibitor in squamous cell lung carcinoma. Lung Cancer 101: 111-119.
  76. Sun XP, Zhang X, He C, Qiao H, Jiang X, et al. (2012) ABT-737 synergizes with arsenic trioxide to induce apoptosis of gastric carcinoma cells in vitro and in vivo. J Int Med Res 40: 1251-1264.
  77. Cang S, Iragavarapu C, Savooji J, Song Y, Liu D (2015) ABT-199 (venetoclax) and BCL-2 inhibitors in clinical development. J Hematol Oncol 8: 129.
  78. Wang W, Adachi M, Zhang R, Zhou J, Zhu D (2009) A novel combination therapy with arsenic trioxide and parthenolide against pancreatic cancer cells. Pancreas 38: e114-e123.
  79. Lang M, Wang X, Wang H, Jie Dong, Chungen Lan, et al. (2016) Arsenic trioxide plus PX-478 achieves effective treatment in pancreatic ductal adenocarcinoma. Cancer Lett 378: 87-96.
  80. Kim EH, Yoon MJ, Kim SU, Kwon TK, Sohn S, et al. (2008) Arsenic trioxide sensitizes human glioma cells, but not normal astrocytes, to TRAIL-induced apoptosis via CCAAT/ enhancer-binding protein homologous protein-dependent DR5 up-regulation. Cancer Res 68: 266-275
  81. Chiu HW, Lin JH, Chen YA, Ho SY, Wang YJ (2010) Combination treatment with arsenic trioxide and irradiation enhances cell-killing effects in human fibrosarcoma cells in vitro and in vivo through induction of both autophagy and apoptosis. Autophagy 6: 353-365.
  82. Chiu HW, Lin W, Ho SY, Wang YJ (2011) Synergistic effects of arsenic trioxide and radiation in osteosarcoma cells through the induction of both autophagy and apoptosis. Radiat Res 175: 547-560.
  83. Baumgartner M, Sturlan S, Roth E, Wessner B, Bachleitner-Hofmann T (2004) Enhancement of arsenic trioxide-mediated apoptosis using docosahexaenoic acid in arsenic trioxideresistant solid tumor cells. Int J Cancer 112: 707-712.
  84. Ravandi F, Estey E, Jones D, et al. (2009) Effective treatment of acute promyelocytic leukemia with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab ozogamicin. J Clin Oncol 27: 504-510.
  85. Abaza Y, Kantarjian H, Garcia-Manero G, Elihu E, Gautam B, et al. (2017) Longterm outcome of acute promyelocytic leukemia treated with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab. Blood 129: 1275-1283.
  86. Kryeziu K, Jungwirth U, Hoda MA, Franziska F, Siegfried K, et al. (2013) Synergistic anticancer activity of arsenic trioxide with erlotinib is based on inhibition of EGFR-mediated DNA double-strand break repair. Mol Cancer Ther 12: 1073-1084.
  87. Lo-Coco F, Avvisati G, Vignetti M, Christian T, Sonia MO, et al. (2013) Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med 369: 111-121.
  88. Tarkanyi I, Dudognon C, Hillion J, Pendino F, Lanotte M, et al. (2005) Retinoid/arsenic combination therapy of promyelocytic leukemia: Induction of telomerase-dependent cell death. Leukemia 19: 1806-1811.
  89. de Thé H, Chen Z (2010) Acute promyelocytic leukaemia: Novel insights into the mechanisms of cure. Nature Reviews. Cancer 10: 775-783.
  90. Rogers CS, Yedjou CG, Sutton DJ, Tchounwou PB (2014) Vitamin D3 potentiates the antitumorigenic effects of arsenic trioxide in human leukemia (HL-60) cells. Exp Hematol Oncol 3: 9.
  91. Muto A, Kizaki M, Kawamura C, et al. (2001) A novel differentiation-inducing therapy for acute promyelocytic leukemia with a combination of arsenic trioxide and GM-CSF. Leukemia 15: 1176-1184.
  92. Kumagai T, Shih LY, Hughes SV, Desmond JC, O'Kelly J, et al. (2005) 19-Nor1,25 (OH)2D2 (a novel, noncalcemic vitamin D analogue), combined with arsenic trioxide, has potent antitumor activity against myeloid leukemia. Cancer Res 65: 2488-2497.
  93. Thiede C, Steudel C, Mohr B, Markus S, Ulrike S, et al. (2002) Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: Association with FAB subtypes and identification of subgroups with poor prognosis. Blood 99: 4326-4335.
  94. Takahashi S, Harigae H, Yokoyama H, Izumi I, Shouri A, et al. (2006) Synergistic effect of arsenic trioxide and flt3 inhibition on cells with flt3 internal tandem duplication. Int J Hematol 84: 256-261.
  95. El-Hajj H, Dassouki Z, Berthier C, Raffoux E, Ades L, et al. (2015) Retinoic acid and arsenic trioxide trigger degradation of mutated NPM1, resulting in apoptosis of AML cells. Blood 125: 3447-3454.
  96. Tanaka Y, Komatsu T, Shigemi H, Yamauchi T, Fujii Y (2014) BIMEL is a key effector molecule in oxidative stress-mediated apoptosis in acute myeloid leukemia cells when combined with arsenic trioxide and buthionine sulfoximine. BMC Cancer 14: 27
  97. Emadi A, Sadowska M, Carter-Cooper B, Bhatnagar V, van der Merwe I, et al. (2015) Perturbation of cellular oxidative state induced by dichloroacetate and arsenic trioxide for treatment of acute myeloid leukemia. Leuk Res 39: 719-729.
  98. Chau D, Ng K, Chan TS, Cheng YY, Fong B, Tam S, et al. (2015) Azacytidine sensitizes acute myeloid leukemia cells to arsenic trioxide by up-regulating the arsenic transporter aquaglyceroporin 9. J Hematol Oncol 8: 46.
  99. Dembitz V, Lalic H, Ostojic A, Vrhovac R, Banfic H, et al. (2015) The mechanism of synergistic effects of arsenic trioxide and rapamycin in acute myeloid leukemia cell lines lacking typical t (15;17) translocation. Int J Hematol 102: 12-24.
  100. Ye Y, Xu X, Zhang M, Dafa Q, Xiaochun B, et al. (2015) Low-dose arsenic trioxide combined with aclacinomycin a synergistically enhances the cytotoxic effect on human acute myelogenous leukemia cell lines by induction of apoptosis. Leuk Lymphoma 56: 3159-3167
  101. Grad JM, Bahlis NJ, Reis I, Oshiro MM, Dalton WS, et al. (2001) Ascorbic acid enhances arsenic trioxide-induced cytotoxicity in multiple myeloma cells. Blood 98: 805-913.
  102. Berenson JR, Boccia R, Siegel D, et al. (2006) Efficacy and safety of melphalan, arsenic trioxide and ascorbic acid combination therapy in patients with relapsed or refractory multiple myeloma: A prospective, multicenter, phase II, single-arm study. Br J Haematol 135: 174-183.
  103. Stewart AK (2012) Novel therapeutics in multiple myeloma. Hematol 17: S105-S108.
  104. Wen J, Feng Y, Huang W, Chen H, Liao B, et al. (2010) Enhanced antimyeloma cytotoxicity by the combination of arsenic trioxide and bortezomib is further potentiated by p38 MAPK inhibition. Leuk Res 34: 85-92.
  105. Jian Y, Gao W, Geng C, Zhou H, Leng Y, et al. (2017) Arsenic trioxide potentiates sensitivity of multiple myeloma cells to lenalidomide by upregulating cereblon expression levels. Oncol Lett 14: 3243-3248.
  106. Diaz Z, Colombo M, Mann KK, Su H, Smith KN, et al. (2005) Trolox selectively enhances arsenic-mediated oxidative stress and apoptosis in APL and other malignant cell lines. Blood 105: 1237-1245.
  107. Lunghi P, Giuliani N, Mazzera L, Lombardi G, Ricca M, et al. (2008) Targeting MEK/MAPK signal transduction module potentiates ATO-induced apoptosis in multiple myeloma cells through multiple signaling pathways. Blood 112: 2450-2462.
  108. Liu P, Xu S, Zhang M, Wang WW, Zhang YF, et al. (2013) Anticancer activity in human multiple myeloma U266 cells: Synergy between cryptotanshinone and arsenic trioxide. Metallomics 5: 871-878.
  109. Shin DS, Kim HN, Shin KD, Young JY, Seung-Jun K, et al. (2009) Cryptotanshinone inhibits constitutive signal transducer and activator of transcription 3 function through blocking the dimerization in DU145 prostate cancer cells. Cancer Res 69: 193-202.
  110. Jung HJ, Chen Z, McCarty N (2012) Synergistic anticancer effects of arsenic trioxide with bortezomib in mantle cell lymphoma. Am J Hematol 87: 1057-1064.
  111. Zhao LL, Liu YF, Peng LJ, Fei AM, Cui W, et al. (2015) Arsenic trioxide rewires mantle cell lymphoma response to bortezomib. Cancer Med 4: 1754-1766.
  112. Ding X, Chi J, Yang X, Hao J, Liu C, et al. (2017) Cucurbitacin B synergistically enhances the apoptosis-inducing effect of arsenic trioxide by inhibiting STAT3 phosphorylation in lymphoma Ramos cells. Leuk Lymphoma 58: 2439-2451.