Next Article in Journal
Polymer Scaffolds for Biomedical Applications in Peripheral Nerve Reconstruction
Next Article in Special Issue
Protective Role of Vanillic Acid against Diethylnitrosamine- and 1,2-Dimethylhydrazine-Induced Hepatocarcinogenesis in Rats
Previous Article in Journal
Functional Pyrazolo[1,5-a]pyrimidines: Current Approaches in Synthetic Transformations and Uses As an Antitumor Scaffold
Previous Article in Special Issue
Crosstalk of Cancer Signaling Pathways by Cyclic Hexapeptides and Anthraquinones from Rubia cordifolia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Anti-Leukemic Activity of Natural Compounds

1
Department of Hematology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
2
“Aurel Ardelean” Institute of Life Sciences, Vasile Godis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
3
Department of Gastroenterology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
4
Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, Rebreanu 86, 310414 Arad, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2021, 26(9), 2709; https://doi.org/10.3390/molecules26092709
Submission received: 21 March 2021 / Revised: 26 April 2021 / Accepted: 30 April 2021 / Published: 5 May 2021
(This article belongs to the Special Issue Antitumoral Properties of Natural Products Ⅱ)

Abstract

:
The use of biologically active compounds has become a realistic option for the treatment of malignant tumors due to their cost-effectiveness and safety. In this review, we aimed to highlight the main natural biocompounds that target leukemic cells, assessed by in vitro and in vivo experiments or clinical studies, in order to explore their therapeutic potential in the treatment of leukemia: acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia (CLL). It provides a basis for researchers and hematologists in improving basic and clinical research on the development of new alternative therapies in the fight against leukemia, a harmful hematological cancer and the leading cause of death among patients.

1. Introduction

Cancer is one of the leading causes of death worldwide and a major challenge for the public health system [1]. The incidence of cancer is constantly increasing and is estimated to increase by 70% over the next 20 years [2].
Conventional anticancer therapies have limited efficacy and are associated with many side effects, such as hepatotoxicity, myelosuppression, or tumor lysis syndrome [3]. Chemotherapy and radiation therapy are frequently correlated with side effects, such as hair loss, loss of appetite, diarrhea, vomiting, liver damage, and neurological disorders [4]. Therefore, it is necessary to find new therapeutic approaches with high efficacy and fewer side effects. The main treatments used in leukemia are radiotherapy, hyperthermia, and chemotherapy. Conventional drug treatment is associated with cytotoxicity and systemic side effects. Therefore, efforts in cancer treatment are focused on finding strategies that can specifically target tumor cells without affecting normal cells [5]. Understanding the molecular mechanisms involved in hematologic cancers is useful in developing of the new therapeutic strategies that target various molecular abnormalities. Recently, there has been an increase in molecularly targeted therapies approved by the FDA in various types of leukemia, but there are insufficient data on the use of these drugs. Thus, in the case of AML, several agents are available for various clinical stages, but the best response rates were obtained by combining new molecularly-targeted treatments with conventional induction chemotherapy [6]. However, the patients experience short-term nausea/vomiting, diarrhea, hair loss, mouth sores, infection, rash; and for the long-term, organ dysfunction, chemobrain, fatigue, neuropathy, as well as resistance of leukemia cells to chemotherapy drugs [7,8,9], highlighting the need for the development of less toxic and targeted therapies.
Recent advances in understanding carcinogenesis have led to the synthesis of new drugs that target specific receptors [10]. The development of new antitumor agents is an important strategy in the fight against cancer [11]. The development of new anticancer agents derived from natural sources is currently being pursued [12]. Secondary metabolites from plants, such as flavonoids, alkaloids, terpenoids, saponins, and others, are important sources of anticancer agents [13,14,15]. Different types of herbal formulations, such as flavonoids and various enzymes, play an important role in cancer by preventing DNA damage and increasing the level of antioxidants in the body with lower side effects [16]. Lately, many phytochemicals isolated from different parts of the plant have been tested by in vitro and in vivo experiments to find biological effects against different diseases, such as cancer.
Over 60% of anti-tumor drugs that have shown high efficacy in clinical use have been obtained from plants, aquatic organisms, and microorganisms. The anticancer effect of these natural products is mediated by various mechanisms, as apoptosis, modulation of the immune system, and inhibition of angiogenesis [17].
There are several plant-derived compounds used in the treatment of hematologic cancers. The vinca alkaloids, vincristine and vinblastine, the first US FDA-approved anticancer agents in plants, are used to treat lymphomas, including Hodgkin’s disease and acute lymphoblastic leukemias in combination with chemotherapy [18,19]. Etoposides, a compound used in the treatment of various types of leukemias and lymphomas, and teniposides used in various types of hematological cancers, either alone or in combination with chemotherapeutic drugs, are semi-synthetic plant derivatives [20,21].
Cancer chemoprevention is a new approach to cancer management. This therapeutic strategy uses non-cytotoxic drugs and natural agents to inhibit carcinogenesis [22] and block progression to invasive cancer [10]. Secondary metabolites in plants, enzymes, and other compounds play an important role in combating various types of cancer [23]. Chemoprevention includes DNA damage protection, which initiates the process of neoplastic transformation or can reverse the progression of preinvasive lesions. The effectiveness of this approach has been highlighted by epidemiological observations, in experimental models of animal carcinogenesis, knock-out models, tumor cell lines, and clinical studies [10].
In this review, we aimed to highlight the main biologically active compounds which target leukemic cells, assessed by in vitro and in vivo experiments or clinical studies, in order to explore their therapeutic potential in treatment of leukemia.
The biologically active compounds with antileukemic activity presented in the below tables are of plant origin and they are widespread in the plant kingdom. For example: luteolin is a flavone found in carrots, celery, peppers, cabbage, broccoli, onion leaves, apple skins, parsley, basil, thyme, and mint [24,25]; quercetin is found in many fruits and vegetables such as apples, cherries, berries, onions, asparagus, and red leaf lettuce [26]; apigenin is contained in Artemisia [27], Achillea [28,29], Matricaria [30], and Tanacetum [31] genera; epigallocatechin-gallate (EGCG) is the main constituent of green tea [32]; curcumin is a phenolic compound found in the rhizomes of Curcuma longa L., commonly known as turmeric [33]; thymoquinone is a monoterpene isolated from Nigella sativa seeds [34]. It is also found in high concentration in the Monarda fistulosa plant, also known as wild bergamot [35]; emodin is a natural anthraquinone derivative [36] extracted from various plants, such as Rheum officinale and Polygonam cuspidatum [37]; parthenolide is a sesquiterpene lactone extracted from the leaves of the medicinal plant Tanacetum parthenium [38].
This wide range of natural compounds with anti-leukemic potential provides a basis for researchers and hematologists in improving basic and clinical research on the development of new alternative therapies in the fight against leukemia.

2. Natural Compounds in Acute Myeloid Leukemia (AML)

Acute myeloid leukemia (AML) is the most common type of acute leukemia among adults [5]. This is an aggressive hematological malignancy characterized by an extremely proliferative accumulation of immature and dysfunctional myeloid cells [39] which infiltrates bone marrow, blood, and other tissues [40]. Additionally, leukemic cells show an increase proliferative capacity and altered hematopoietic differentiation [41].
Although most patients with AML experienced partial remission after conventional treatment, such as chemotherapy, they face a number of problems, such as the risk of recurrence, malignant cell resistance, and side effects that diminish the therapeutic value of these treatments [42]. Recurrence is common and the chances of survival are lower for a long term in most cases [43].
The main difficulty in the treatment of AML is chemoresistance, and CD34 + AML cells indicate poor prognosis and resistance to spontaneous apoptosis [44]. The emergence of multidrug resistance (MDR) in chemotherapeutic agents is an important obstacle in the treatment of AML. The discovery of new therapeutic agents that can be used to overcome MDR is becoming a challenge in clinical practice [37].
To date, polyphenols having cytotoxic effect on AML cells were identified [45]. Deng et al. (2017) demonstrated that luteolin extracted from Reseda odorata L., inhibited the growth of leukemic cell lines by inducing apoptosis through blocking of the RSK1 pathway, as well as by inhibiting their ability to migrate [46]. Other studies demonstrated a selective inhibitory activity against Fms-like tyrosine kinase 3 (FLT3), a highly expressed tyrosine kinase receptor in patients with AML and induced a strong cytotoxic effect in MV4-11 leukemic cells [47].
Quercetin has been shown to have an antitumor effect in various experimental models using tumor cell lines, including AML [48,49,50]. The antitumor activity of quercetin has been correlated with its ability to inhibit proliferation and induced cell death in AML cells [48,51]. Quercetin induced AML cell apoptosis through Fas-mediated extrinsic pathways [51] and mitochondrial-derived intrinsic pathways [48]. It also had antitumor effect in acute T-cell lymphoblastic leukemia (ALL) and chronic myeloid leukemia (CML) [52,53].
Delphinidin showed antiproliferative effects against human acute promyelocytic leukemia (APL) NB4 cell line, a subtype of acute myeloid leukemia. Delphinidin had a cytotoxic effect on NB4 cells, induced activation of caspase-8 and -9 and -3 and decreased Bid expression and mitochondrial membrane potential (ΔΨm). Delphinidine-induced cytotoxicity was more pronounced in NB4 cells compared to normal peripheral blood mononuclear cells (PBMNCs) [54].
Genistein has been shown to have antiproliferative activity on tumor cells, being an alternative therapy for the treatment of patients with AML [55].
Parthenolide induced specific toxicity to leukemic cells and leukemic stem cells (LSCs) without causing damage to normal hematopoietic cells [56]. Parthenolide has been shown to be effective in inducing specific apoptosis to LSCs in AML. Due to poor bioavailability, the antileukemic activity of parthenolide has not been demonstrated in vivo [57,58]. In order to increase water solubility, parthenolide analogs have been developed [59] that showed high bioavailability and bioactivity in vivo [57]. The chemically modified parthenolide analog, dimethylamino-parthenolide, showed an oral bioavailability of ~70% compared to intravenous administration in experimental models performed in mice and dogs and an improvement in the selective eradication of AML and of their progenitor stem cells [57].
Martínez-Castillo et al. (2018) studied the effects of curcumin in two cell lines derived from chronic and acute myeloid leukemia, respectively, HL-60 and K562 cells. K562 cells showed a higher sensitivity to cytostatic and cytotoxic effects of curcumin compared to HL-60 cells. Curcumin induced G1 phase blockade in HL-60 cells and G2/M phase blockade in K562 cells. Curcumin induced apoptosis in cell lines derived from chronic and acute myeloid leukemia by distinct cellular mechanisms. Thus, curcumin-induced apoptosis in HL-60 cells was caspase-dependent, whereas in K562 cells, they underwent apoptosis in a caspase-independent manner [60].
Boswellic acid acetate, a 1:1 mixture of α-boswellic acid acetate and β-boswellic acid acetate, isolated from Boswellia carterri, showed cytotoxic effects against six myeloid leukemia cell lines. This cytotoxic effect was mediated by the induction of apoptosis. Over 50% of cells underwent apoptosis after treatment with 20 mg/mL boswellic acid acetate for 24 h [61].
The main pharmacological effects exerted by natural compounds against acute myeloid leukemia (AML) are summarized in Table 1.
The natural compounds with anti-tumoral activity against acute mieloid leukemia (AML) by in vitro and in vivo experiments or synergic activity with antineoplastig drugs, are summarized in Figure 1.

3. Natural Compounds in Chronic Myeloid Leukemia (CML)

Chronic myeloid leukemia (CML), BCR-ABL1-positive, also known as chronic myelogenous leukemia, is defined as a myeloproliferative neoplasm consisting predominantly of proliferating granulocytes [83]. This has an incidence of 1–2 cases per 100,000 adults [84]. Approximately 95% of patients with CML have t (9; 22) translocation (q34; q11.2) [85]. CML affects both peripheral blood and bone marrow [83].
Fusion of the Abelson gene (ABL1) on chromosome 9 with the cluster breakpoint region (BCR) on chromosome 22 generates the oncoprotein BCR-ABL, an active tyrosine kinase that induces cytokine-independent cell proliferation, which causes excessive accumulation of myeloid cells in hematopoietic tissues [86]. The Bcr-Abl oncoprotein activates several downstream pathways, responsible for inducing cell proliferation, loss of adhesion, cell differentiation blocking, and inhibition apoptosis [87,88].
The main pharmacological effects exerted by natural compounds against chronic myeloid leukemia (CML) are summarized in Table 2.
The natural compounds with anti-tumoral activity against chronic myeloid leukemia (AML) by in vitro and in vivo experiments, are summarized in Figure 2.

4. Natural Compounds in Acute Lymphoblastic Leukemia (ALL)

Acute T-cell lymphoblastic leukemia (T-ALL) is an aggressive malignant blood disorder [112]. Currently, the T-ALL treatment protocols include high doses of chemotherapeutics, which have significant toxic side effects [113,114]. Natural products with various biological activities and specific selectivity have served as important sources of antitumor agents that have been developed for clinical use [115].
Anthocyanins, a subclass of flavonoids, are glycosides of anthocyanidins [116]. Blueberries are an important source of anthocyanins [117]. Anthocyanins showed, anti-mutagenesis and anti-carcinogenesis activity [118,119]. They have been shown to have a strong antitumor effect by inducing a pro-apoptotic mitochondrial-mediated response [120].
Anthocyanins from blueberry extract (Antho 50) induced apoptosis in Jurkat cells by decreasing the expression of Polycomb group proteins. This effect was mediated by an increase in intracellular ROS and depolarization of the mitochondrial membrane [117]. In another study, two anthocyanins extracted from blackcurrant juice, delphinidin-3-O-glucoside and delphinidin-3-O-rutinoside, induced apoptosis in human Jurkat leukemic cells [121]. Additionally, blackcurrant juice and blackcurrant extract inhibited proliferation, induced cell cycle arrest in the G2/M phase, and apoptosis in Jurkat cells. These effects have been associated with increased expression of p73 and caspase 3, Akt and Bad dephosphorylation, and down-regulation of UHRF1 and Bcl-2 [121].
The main pharmacological effects exerted by natural compounds against acute lymphoblastic leukemia (ALL) are summarized in Table 3.
The natural compounds with anti-tumoral activity against acute lymphoblastic leukemia (ALL) by in vitro and in vivo experiments or antagonizing activity against cytotoxicity of antineoplastic drugs, are summarized in Figure 3.

5. Natural Compounds in Chronic Lymphocytic Leukemia (CLL)

Chronic lymphocytic leukemia (CLL) is the most common type of hematologic cancer in the western countries (22–30%) [134,135]. CLL is a monoclonal lymphoproliferative disorder characterized by the proliferation and accumulation of morphologically mature, but immunologically dysfunctional B-cell lymphocytes [136]. CLL B cells interact with their microenvironment, and B cell survival is enhanced by contact with bone marrow stromal cells. Therefore, the lifespan of B cells increases, causing their abnormal accumulation [137]. The main sites of the disease include peripheral blood, spleen, lymph nodes, and bone marrow [136]. It mainly affects adults [138].
Although there are many therapeutic protocols, CLL is still an incurable disease [138]. Current treatment options include conventional chemotherapy, monoclonal antibodies, and hematopoietic transplantation [139]. These standard treatment methods are not sufficient to eliminate all CLL cells and have a number of side effects. Additionally, standard treatment promotes the development of resistance to treatment and most treated patients relapsed. Therefore, it is necessary to develop new therapeutic strategies that could eliminate apoptosis-resistant CLL cells. Recently, there has been a growing interest in the use of agents derived from natural compounds for cancer therapy [140].
Bcl-2 plays a key role in regulating cellular responses to treatment due to its pro- and anti-apoptotic properties [141]. The anti-apoptotic protein Bcl-2 is overexpressed in several hematological malignancies, including CLL. This overexpression is considered to be responsible for defective apoptosis in CLL [142].
The effects of polyphenols on cell proliferation, gene regulation, and apoptosis have been studied on several cancer cell lines [143].
Alhosin et al. (2015) demonstrated that a standardized blueberry extract containing 50% anthocyanins (Antho 50) had the ability to induce apoptosis in CLL B cells via the Bcl-2/Bad pathway. They evaluated the pro-apoptotic effect of Antho 50 on CLL B cells from 30 patients and on peripheral blood mononuclear cells (PBMCs) from healthy subjects. The main phenolic compounds in cranberry extract responsible for the pro-apoptotic effect in CLL B cells were delphinidin-3-O-glucoside and delphinidin-3-O-rutinoside. Antho 50-induced apoptosis has been associated with caspase-3 activation, down-regulation of UHRF1, dephosphorylation of Akt and Bad, and down-regulation of Bcl-2 [144].
Luteolin significantly induced apoptosis in chronic lymphocytic leukemia (CLL) cell lines by increasing caspase activity and triggering the intrinsic apoptotic pathway [145].
The main pharmacological effects exerted by natural compounds against chronic lymphocytic leukemia (CLL) are summarized in Table 4.
The natural compounds with anti-tumoral activity against chronic lymphocytic leukemia (CLL) by in vitro and in vivo experiments or antagonizing activity against cytotoxicity of antineoplastic drugs, are summarized in Figure 4.

6. Clinical Trials and Synergic Activity with Conventional Anti-Leukemic Drugs

Several clinical studies are published in database ClinicalTrials.Gov regarding the anti-tumor action of biactive compounds and synergies with anti-neoplastic therapy of leukemias.
The effect of genistein was tested in a phase I/II clinical study in combination with decitabine in pediatric relapsed refractory malignancies. Genistein was administered orally twice daily from day 2 to day 21, followed by a 7-day break (clinical trial number: NCT02499861). The aim of the research includes assessment of a tolerated dose of the combination of intravenous decitabine with oral genistein for children with refractory or recurrent solid malignancies and leukemia. The adverse events of the combination therapy and clinical benefit in phase IIa of the study measured by either volumetric MRI for solid tumor or by bone marrow aspiration or biopsy for leukemia at the end of cycles 2, 4, 6, 9, and 12 were assessed. To date, the results are not yet published in the database ClinicalTrials.Gov.
The efficacy of concomitant administration of curcumin and colecalciferol was investigated in a phase II trial in the treatment of patients with chronic lymphocytic leukemia in stage 0-II, previously untreated and small lymphocytic lymphoma (clinical trial number: NCT02100423).
Given that green tea extract contains ingredients that can slow the growth of certain cancers, its effect was tested in a phase I/II trial in the treatment of patients with chronic lymphocytic leukemia in stage 0, I, or II (clinical trial number: NCT00262743). In the phase I trial, patients were given orally 400 to 2000 mg of green tea extract (Polyphenon E) twice a day for 6 months [158]. In the phase II trial, oral administration of 2000 mg of Polyphenon E twice daily for 6 months was well tolerated [159]. Most patients experienced a decrease in absolute lymphocyte count (LAC) and lymphadenopathy following treatment with Polyphenon E [158,159].

7. Conclusions

In this review, we presented the natural compounds that have shown an anti-leukemic activity in experimental studies on different cell lines or primary cultures, preclinical and clinical studies, results that could propose them in subsequent therapeutic protocols of different types of leukemia: acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphocytic leukemia (ALL), and chronic lymphocytic leukemia (CLL). Mechanistically, they demonstrated the ability to induce cell cycle blockage and apoptosis or autophagy in cancer cells, as well as inhibition of proliferation/migration and tumor progression, antagonizing activity of cytotoxicity exerted by antineoplastic drugs, or exerted synergy with conventional therapy. Although in vitro results are promising, most bioactive compounds have not yet been tested in preclinical or clinical studies. Moreover, some of the compounds are not soluble and therefore have a reduced bioavailability when administered orally (e.g., flavonoids), which reduces their potential. Therefore, special formulations or chemical modification are needed to increase the bioactive potential. Overall, nature provides a wide range of bioactive compounds with anti-leukemic potential, and extensive research is still needed for them to be considered viable therapeutic options for the treatment of various types of leukemia.

Author Contributions

Conceptualization C.C., A.C., A.S., and A.H.; methodology C.C., A.C., A.S., and A.H.; validation C.C., A.C., A.S., and A.H.; investigation, C.C., A.C., A.S., A.H., and E.M.; writing—original draft preparation, C.C., A.C., A.S., and A.H.; writing—review and editing, C.C., A.C., A.S., A.H., and E.M.; funding acquisition, C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Talib, W.H.; Alsalahat, I.; Daoud, S.; Abutayeh, R.F.; Mahmod, A.I. Plant-derived natural products in cancer research: Extraction, mechanism of action, and drug formulation. Molecules 2020, 25, 5319. [Google Scholar] [CrossRef]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  3. Watts, J.; Nimer, S. Recent advances in the understanding and treatment of acute myeloid leukemia. F1000Research 2018, 7, 1196. [Google Scholar] [CrossRef] [Green Version]
  4. Rafiq, S.; Raza, M.H.; Younas, M.; Naeem, F.; Adeeb, R.; Iqbal, J.; Anwar, P.; Sajid, U.; Manzoor, H.M. Molecular targets of curcumin and future therapeutic role in leukemia. JBM 2018, 6, 33–50. [Google Scholar] [CrossRef] [Green Version]
  5. Naimi, A.; Entezari, A.; Hagh, M.F.; Hassanzadeh, A.; Saraei, R.; Solali, S. Quercetin sensitizes human myeloid leukemia KG-1 cells against TRAIL-induced apoptosis. J. Cell Physiol. 2019, 234, 13233–13241. [Google Scholar] [CrossRef] [PubMed]
  6. Kayser, S.; Levis, M.J. Advances in targeted therapy for acute myeloid leukaemia. Br. J. Haematol. 2018, 180, 484–500. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, J.; Gu, Y.; Chen, B. Mechanisms of drug resistance in acute myeloid leukemia. Onco Targets Ther. 2019, 12, 1937–1945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Crossnohere, N.L.; Richardson, D.R.; Reinhart, C.; O’Donoghue, B.; Love, S.M.; Smith, B.D.; Bridges, J.F.P. Side effects from acute myeloid leukemia treatment: Results from a national survey. Curr. Med. Res. Opin. 2019, 35, 1965–1970. [Google Scholar] [CrossRef] [PubMed]
  9. Yu, Z.; Liu, L.; Shu, Q.; Li, D.; Wang, R. Leukemia stem cells promote chemoresistance by inducing downregulation of lumican in mesenchymal stem cells. Oncol. Lett. 2019, 18, 4317–4327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Krishnan, K.; Campbell, S.; Abdel-Rahman, F.; Whaley, S.; Stone, W.L. Cancer chemoprevention drug targets. Curr. Drug Targets 2003, 4, 45–54. [Google Scholar] [CrossRef]
  11. Raguz, S.; Yagüe, E. Resistance to chemotherapy: New treatments and novel insights into an old problem. Br. J. Cancer 2008, 99, 387–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Dias, D.A.; Urban, S.; Roessner, U. A historical overview of natural products in drug discovery. Metabolites 2012, 2, 303–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Yin, S.J.; Zhang, L.; Zhang, L.; Wan, J.; Song, W.; Jiang, X.; Park, Y.D.; Si, Y.X. Metabolic responses and arginine kinase expression of juvenile cuttlefish (Sepia pharaonis) under salinity stress. Int. J. Biol. Macromol. 2018, 113, 881–888. [Google Scholar] [CrossRef]
  14. Avato, P.; Migoni, D.; Argentieri, M.; Fanizzi, F.P.; Tava, A. Activity of saponins from Medicago species against HeLa and MCF-7 cell lines and their capacity to potentiate cisplatin effect. Anti Cancer Agents Med. Chem. 2017, 17, 1508–1518. [Google Scholar] [CrossRef] [PubMed]
  15. Joshi, P.; Vishwakarma, R.A.; Bharate, S.B. Natural alkaloids as P-gp inhibitors for multidrug resistance reversal in cancer. Eur. J. Med. Chem. 2017, 138, 273–292. [Google Scholar] [CrossRef] [PubMed]
  16. Oberley, T.D.; Oberley, L.W. Antioxidant enzyme level in cancer. Histol. Histopathol. 1997, 12, 525–535. [Google Scholar]
  17. Rayan, A.; Raiyn, J.; Falah, M. Nature is the best source of anticancer drugs: Indexing natural products for their anticancer bioactivity. PLoS ONE 2017, 12, e0187925. [Google Scholar] [CrossRef]
  18. Chabner, B.A. Goodman & Gilman’s The Pharmacological Basis of Therapeutics, 11th ed.; Brunton, L.L., Lazo, J.S., Parker, K.L., Eds.; McGraw-Hill: New York, NY, USA, 2006; pp. 1257–1262. [Google Scholar] [CrossRef]
  19. Guérritte, F. Anticancer Agents from Natural Products, 1st ed.; Cragg, G.M., Kingston, D.G.I., Newman, D.J., Eds.; CRC/Taylor & Francis Press: Boca Raton, FL, USA, 2005; pp. 123–135. [Google Scholar] [CrossRef]
  20. Lee, K.H. Anticancer Agents from Natural Products, 1st ed.; Cragg, G.M., Kingston, D.G.I., Newman, D.J., Eds.; CRC/Taylor & Francis Press: Boca Raton, FL, USA, 2005; pp. 71–87. [Google Scholar] [CrossRef]
  21. Hande, K.R. Etoposide: Four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer. 1998, 34, 1514–1521. [Google Scholar] [CrossRef]
  22. Silalahi, J. Anticancer and health protective properties of citrus fruit components. Asia Pac. J. Clin. Nutr. 2002, 11, 79–84. [Google Scholar] [CrossRef] [PubMed]
  23. Kellof, G.J. Perspective on cancer chemoprevention research and drug development. Adv. Cancer Res. 2000, 78, 199–334. [Google Scholar] [CrossRef]
  24. Ahmed, S.; Khan, H.; Fratantonio, D.; Hasan, M.M.; Sharifi, S.; Fathi, N.; Ullah, H.; Rastrelli, L. Apoptosis induced by luteolin in breast cancer: Mechanistic and therapeutic perspectives. Phytomedicine 2019, 59, 152883. [Google Scholar] [CrossRef]
  25. Aziz, N.; Kim, M.Y.; Cho, J.Y. Anti-inflammatory effects of luteolin: A review of in vitro, in vivo, and in silico studies. J. Ethnopharmacol. 2018, 225, 342–358. [Google Scholar] [CrossRef]
  26. Costa, L.G.; Garrick, J.M.; Roquè, P.J.; Pellacani, C. Mechanisms of Neuroprotection by Quercetin: Counteracting Oxidative Stress and More. Oxid. Med. Cell Longev. 2016, 2016, 2986796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Ornano, L.; Venditti, A.; Donno, Y.; Sanna, C.; Ballero, M.; Bianco, A. Phytochemical analysis of non-volatile fraction of Artemisia caerulescens subsp. densiflora (Viv.) (Asteraceae), an endemic species of La Maddalena Archipelago (Sardinia–Italy). Nat. Prod. Res. 2016, 30, 920–925. [Google Scholar] [CrossRef] [PubMed]
  28. Venditti, A.; Maggi, F.; Vittori, S.; Papa, F.; Serrilli, A.M.; Di Cecco, M.; Bianco, A. Antioxidant and α-glucosidase inhibitory activities of Achillea tenorii. Pharm. Biol. 2015, 53, 1505–1510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Venditti, A.; Guarcini, L.; Bianco, A.; Rosselli, S.; Bruno, M.; Senatore, F. Phytochemical analysis of Achillea ligustica all. from Lipari Island (Aeolian islands). Nat. Prod. Res. 2016, 30, 912–919. [Google Scholar] [CrossRef] [PubMed]
  30. Sharifi-Rad, M.; Nazaruk, J.; Polito, L.; Morais-Braga, M.F.B.; Rocha, J.E.; Coutinho, H.D.M.; Salehi, B.; Tabanelli, G.; Montanari, C.; Del Mar Contreras, M.; et al. Matricaria genus as a source of antimicrobial agents: From farm to pharmacy and food applications. Microbiol. Res. 2018, 215, 76–88. [Google Scholar] [CrossRef] [PubMed]
  31. Venditti, A.; Frezza, C.; Sciubba, F.; Serafini, M.; Bianco, A.; Cianfaglione, K.; Maggi, F. Volatile components, polar constituents and biological activity of tansy daisy (Tanacetum macrophyllum (Waldst. et Kit.) Schultz Bip. Ind. Crop. Prod. 2018, 118, 225–235. [Google Scholar] [CrossRef]
  32. Steinmann, J.; Buer, J.; Pietschmann, T.; Steinmann, E. Anti-infective properties of epigallocatechin-3-gallate (EGCG), a component of green tea. Br. J. Pharmacol. 2013, 168, 1059–1073. [Google Scholar] [CrossRef] [Green Version]
  33. Russo, E.R.; Facincani, I.; Nakazato, K.C.; Coimbra, T.M.; Crevelin, E.J.; Pereira, A.M.S.; Carmona, F. Oral administration of powdered dried rhizomes of Curcuma longa L. (turmeric, Zingiberaceae) is effective in the treatment of doxorubicin-induced kidney injury in rats. Phytother. Res. 2018, 32, 2408–2416. [Google Scholar] [CrossRef] [Green Version]
  34. Arroo, R.R.J.; Alfa, H.H. Chemical properties of thymoquinone, a monoterpene isolated from the seeds of Nigella sativa Linn. Pharmacol Res. 2018, 133, 151. [Google Scholar] [CrossRef]
  35. Pang, J.; Shen, N.; Yan, F.; Zhao, N.; Dou, L.; Wu, L.C.; Seiler, C.L.; Yu, L.; Yang, K.; Bachanova, V.; et al. Thymoquinone exerts potent growth-suppressive activity on leukemia through DNA hypermethylation reversal in leukemia cells. Oncotarget 2017, 8, 34453–34467. [Google Scholar] [CrossRef] [Green Version]
  36. Tang, T.; Yin, L.; Yang, J.; Shan, G. Emodin, an anthraquinone derivative from Rheum officinale Baill, enhances cutaneous wound healing in rats. Eur. J. Pharmacol. 2007, 567, 177–185. [Google Scholar] [CrossRef]
  37. Chen, Y.; Gan, D.; Huang, Q.; Luo, X.; Lin, D.; Hu, J. Emodin and its combination with cytarabine induce apoptosis in resistant acute myeloid leukemia cells in vitro and in vivo. Cell Physiol. Biochem. 2018, 48, 2061–2073. [Google Scholar] [CrossRef]
  38. Sztiller-Sikorska, M.; Czyz, M. Parthenolide as cooperating agent for anti-cancer treatment of various malignancies. Pharmaceuticals 2020, 13, 194. [Google Scholar] [CrossRef]
  39. Shi, H.; Li, X.Y.; Chen, Y.; Zhang, X.; Wu, Y.; Wang, Z.X.; Chen, P.H.; Dai, H.Q.; Feng, J.; Chatterjee, S.; et al. Quercetin induces apoptosis via downregulation of vascular endothelial growth factor/Akt signaling pathway in acute myeloid leukemia cells. Front. Pharmacol. 2020, 11, 534171. [Google Scholar] [CrossRef]
  40. Döhner, H.; Weisdorf, D.J.; Bloomfield, C.D. Acute myeloid leukemia. N. Engl. J. Med. 2015, 373, 1136–1152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Calgarotto, A.K.; Maso, V.; Junior, G.C.F.; Nowill, A.E.; Filho, P.L.; Vassallo, J.; Saad, S.T.O. Antitumor activities of quercetin and green tea in xenografts of human leukemia HL60 cells. Sci. Rep. 2018, 8, 3459. [Google Scholar] [CrossRef] [Green Version]
  42. Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.; Buechner, T.; Dombret, H.; Ebert, B.; Fenaux, P.; Larson, R.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Guzman, M.L.; Rossi, R.M.; Karnischky, L.; Li, X.; Peterson, D.R.; Howard, D.S.; Jordan, C.T. The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cells. Blood 2005, 105, 4163–4169. [Google Scholar] [CrossRef] [PubMed]
  44. Kouhpeikar, H.; Butler, A.E.; Bamian, F.; Barreto, G.E.; Majeed, M.; Sahebkar, A. Curcumin as a therapeutic agent in leukemia. J. Cell Physiol. 2019, 234, 12404–12414. [Google Scholar] [CrossRef] [PubMed]
  45. Pesakhov, S.; Khanin, M.; Studzinski, G.P.; Danilenko, M. Distinct combinatorial effects of the plant polyphenols curcumin, carnosic acid, and silibinin on proliferation and apoptosis in acute myeloid leukemia cells. Nutr. Cancer 2010, 62, 811–824. [Google Scholar] [CrossRef] [PubMed]
  46. Deng, L.; Jiang, L.; Lin, X.; Tseng, K.F.; Lu, Z.; Wang, X. Luteolin, a novel p90 ribosomal S6 kinase inhibitor, suppresses proliferation and migration in leukemia cells. Oncol. Lett. 2017, 13, 1370–1378. [Google Scholar] [CrossRef] [PubMed]
  47. Chin, Y.W.; Kong, J.Y.; Han, S.Y. Flavonoids as receptor tyrosine kinase FLT3 inhibitors. Bioorg. Med. Chem. Lett. 2013, 23, 1768–1770. [Google Scholar] [CrossRef] [PubMed]
  48. Lee, W.J.; Hsiao, M.; Chang, J.L.; Yang, S.F.; Tseng, T.H.; Cheng, C.W.; Chow, J.M.; Lin, K.H.; Lin, Y.W.; Liu, C.C.; et al. Quercetin induces mitochondrial-derived apoptosis via reactive oxygen species-mediated ERK activation in HL-60 leukemia cells and xenograft. Arch. Toxicol. 2015, 89, 1103–1117. [Google Scholar] [CrossRef]
  49. Srivastava, S.; Somasagara, R.R.; Hegde, M.; Nishana, M.; Tadi, S.K.; Srivastava, M.; Choudhary, B.; Raghavan, S.C. Quercetin, a natural flavonoid interacts with DNA, arrests cell cycle and causes tumor regression by activating mitochondrial pathway of apoptosis. Sci. Rep. 2016, 6, 24049. [Google Scholar] [CrossRef] [Green Version]
  50. Ren, M.X.; Deng, X.H.; Ai, F.; Yuan, G.Y.; Song, H.Y. Effect of quercetin on the proliferation of the human ovarian cancer cell line SKOV-3 in vitro. Exp. Ther. Med. 2015, 10, 579–583. [Google Scholar] [CrossRef] [Green Version]
  51. Lee, W.J.; Chen, Y.R.; Tseng, T.H. Quercetin induces FasL-related apoptosis, in part, through promotion of histone H3 acetylation in human leukemia HL-60 cells. Oncol. Rep. 2011, 25, 583–591. [Google Scholar] [CrossRef]
  52. Avci, C.B.; Yilmaz, S.; Dogan, Z.O.; Saydam, G.; Dodurga, Y.; Ekiz, H.A.; Kartal, M.; Sahin, F.; Baran, Y.; Gunduz, C. Quercetin-induced apoptosis involves increased hTERT enzyme activity of leukemic cells. Hematology 2011, 16, 303–307. [Google Scholar] [CrossRef] [Green Version]
  53. Larocca, L.M.; Teofili, L.; Leone, G.; Sica, S.; Pierelli, L.; Menichella, G.; Scambia, G.; Benedetti Panici, P.; Ricci, R.; Piantelli, M.; et al. Antiproliferative activity of quercetin on normal bone marrow and leukaemic progenitors. Br. J. Haematol. 1991, 79, 562–566. [Google Scholar] [CrossRef]
  54. Yuan, B.; Okusumi, S.; Yoshino, Y.; Moriyama, C.; Tanaka, S.; Hirano, T.; Takagi, N.; Toyoda, H. Delphinidin induces cytotoxicity and potentiates cytocidal effect in combination with arsenite in an acute promyelocytic leukemia NB4 cell line. Oncol. Rep. 2015, 34, 431–438. [Google Scholar] [CrossRef] [Green Version]
  55. Raynal, N.J.; Momparler, L.; Charbonneau, M.; Momparler, R.L. Antileukemic activity of genistein, a major isoflavone present in soy products. J. Nat. Prod. 2008, 71, 3–7. [Google Scholar] [CrossRef] [PubMed]
  56. Pajak, B.; Gajkowska, B.; Orzechowski, A. Molecular basis of parthenolide-dependent proapoptotic activity in cancer cells. Folia Histochem. Cytobiol. 2008, 46, 129–135. [Google Scholar] [CrossRef] [Green Version]
  57. Guzman, M.L.; Rossi, R.M.; Neelakantan, S.; Li, X.; Corbett, C.A.; Hassane, D.C.; Becker, M.W.; Bennett, J.M.; Sullivan, E.; Lachowicz, J.L.; et al. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood 2007, 110, 4427–4435. [Google Scholar] [CrossRef]
  58. Curry, E.A., 3rd; Murry, D.J.; Yoder, C.; Fife, K.; Armstrong, V.; Nakshatri, H.; O’Connell, M.; Sweeney, C.J. Phase I dose escalation trial of feverfew with standardized doses of parthenolide in patients with cancer. Investig. New Drugs. 2004, 22, 299–305. [Google Scholar] [CrossRef]
  59. Kolev, J.N.; O’Dwyer, K.M.; Jordan, C.T.; Fasan, R. Discovery of potent parthenolide-based antileukemic agents enabled by late-stage P450-mediated C-H functionalization. ACS Chem. Biol. 2014, 9, 164–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Martínez-Castillo, M.; Villegas-Sepúlveda, N.; Meraz-Rios, M.A.; Hernández-Zavala, A.; Berumen, J.; Coleman, M.A.; Orozco, L.; Cordova, E.J. Curcumin differentially affects cell cycle and cell death in acute and chronic myeloid leukemia cells. Oncol. Lett. 2018, 15, 6777–6783. [Google Scholar] [CrossRef]
  61. Xia, L.; Chen, D.; Han, R.; Fang, Q.; Waxman, S.; Jing, Y. Boswellic acid acetate induces apoptosis through caspase-mediated pathways in myeloid leukemia cells. Mol. Cancer Ther. 2005, 4, 381–388. [Google Scholar] [CrossRef]
  62. Britschgi, A.; Simon, H.U.; Tobler, A.; Fey, M.F.; Tschan, M.P. Epigallocatechin-3-gallate induces cell death in acute myeloid leukaemia cells and supports all-trans retinoic acid-induced neutrophil differentiation via death-associated protein kinase 2. Br. J. Haematol. 2010, 149, 55–64. [Google Scholar] [CrossRef]
  63. Papież, M.A.; Bukowska-Straková, K.; Krzysciak, W.; Baran, J. (−)-Epicatechin enhances etoposide-induced antileukaemic effect in rats with acute myeloid leukaemia. Anticancer Res. 2012, 32, 2905–2913. [Google Scholar]
  64. Alvarez, M.C.; Maso, V.; Torello, C.O.; Ferro, K.P.; Saad, S.T.O. The polyphenol quercetin induces cell death in leukemia by targeting epigenetic regulators of pro-apoptotic genes. Clin. Epigenetics 2018, 10, 139. [Google Scholar] [CrossRef] [PubMed]
  65. Maso, V.; Calgarotto, A.K.; Franchi, G.C., Jr.; Nowill, A.E.; Filho, P.L.; Vassallo, J.; Saad, S.T. Multitarget effects of quercetin in leukemia. Cancer Prev. Res. 2014, 7, 1240–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lee, S.J.; Yoon, J.H.; Song, K.S. Chrysin inhibited stem cell factor (SCF)/c-Kit complex-induced cell proliferation in human myeloid leukemia cells. Biochem. Pharmacol. 2007, 74, 215–225. [Google Scholar] [CrossRef] [PubMed]
  67. Narasimhan, K.; Lee, Y.M.; Lim, T.K.; Port, S.A.; Han, J.H.; Chen, C.S.; Lin, Q. Genistein exerts anti-leukemic effects on genetically different acute myeloid leukemia cell lines by inhibiting protein synthesis and cell proliferation while inducing apoptosis—Molecular insights from an iTRAQ™ quantitative proteomics study. Oncoscience 2015, 2, 111–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Gu, R.; Zhang, M.; Meng, H.; Xu, D.; Xie, Y. Gallic acid targets acute myeloid leukemia via Akt/mTOR-dependent mitochondrial respiration inhibition. Biomed. Pharmacother. 2018, 105, 491–497. [Google Scholar] [CrossRef] [PubMed]
  69. Jin, U.H.; Song, K.H.; Motomura, M.; Suzuki, I.; Gu, Y.H.; Kang, Y.J.; Moon, T.C.; Kim, C.H. Caffeic acid phenethyl ester induces mitochondria-mediated apoptosis in human myeloid leukemia U937 cells. Mol. Cell. Biochem. 2008, 310, 43–48. [Google Scholar] [CrossRef] [PubMed]
  70. Papież, M.A.; Krzyściak, W.; Szade, K.; Bukowska-Straková, K.; Kozakowska, M.; Hajduk, K.; Bystrowska, B.; Dulak, J.; Jozkowicz, A. Curcumin enhances the cytogenotoxic effect of etoposide in leukemia cells through induction of reactive oxygen species. Drug Des. Devel. Ther. 2016, 10, 557–570. [Google Scholar] [CrossRef] [Green Version]
  71. Peng, D.Y.; Song, H.; Liu, L.B. Resveratrol-downregulated phosphorylated liver kinase B1 is involved in senescence of acute myeloid leukemia stem cells. J. Huazhong Univ. Sci. Technolog. Med. Sci. 2015, 35, 485–489. [Google Scholar] [CrossRef]
  72. Su, Y.C.; Li, S.C.; Wu, Y.C.; Wang, L.M.; Chao, K.S.; Liao, H.F. Resveratrol downregulates interleukin-6-stimulated sonic hedgehog signaling in human acute myeloid leukemia. Evid. Based. Complement. Alternat. Med. 2013, 2013, 547430. [Google Scholar] [CrossRef]
  73. Yaseen, A.; Chen, S.; Hock, S.; Rosato, R.; Dent, P.; Dai, Y.; Grant, S. Resveratrol sensitizes acute myelogenous leukemia cells to histone deacetylase inhibitors through reactive oxygen species-mediated activation of the extrinsic apoptotic pathway. Mol. Pharmacol. 2012, 82, 1030–1041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hsiao, P.C.; Chou, Y.E.; Tan, P.; Lee, W.J.; Yang, S.F.; Chow, J.M.; Chen, H.Y.; Lin, C.H.; Lee, L.M.; Chien, M.H. Pterostilbene simultaneously induced G0/G1-phase arrest and MAPK-mediated mitochondrial-derived apoptosis in human acute myeloid leukemia cell lines. PLoS ONE 2014, 9, e105342. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, Y.; Hui, H.; Li, Z.; Wang, H.M.; You, Q.D.; Lu, N. Gambogic acid induces growth inhibition and differentiation via upregulation of p21waf1/cip1 expression in acute myeloid leukemia cells. J. Asian Nat. Prod. Res. 2014, 16, 1000–1008. [Google Scholar] [CrossRef] [PubMed]
  76. Yuan, Z.; Wang, H.; Hu, Z.; Huang, Y.; Yao, F.; Sun, S.; Wu, B. Quercetin inhibits proliferation and drug resistance in KB/VCR oral cancer cells and enhances its sensitivity to vincristine. Nutr. Cancer. 2015, 67, 126–136. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, E.A.; Angka, L.; Rota, S.G.; Hanlon, T.; Mitchell, A.; Hurren, R.; Wang, X.M.; Gronda, M.; Boyaci, E.; Bojko, B.; et al. Targeting mitochondria with avocatin B induces selective leukemia cell death. Cancer Res. 2015, 75, 2478–2488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Zahedpanah, M.; Shaiegan, M.; Ghaffari, S.H.; Nikbakht, M.; Nikugoftar, M.; Mohammadi, S. Parthenolide induces apoptosis in committed progenitor AML cell line U937 via reduction in osteopontin. Rep. Biochem. Mol. Biol. 2016, 4, 82–88. [Google Scholar] [PubMed]
  79. Chen, Y.; Li, J.; Hu, J.; Zheng, J.; Zheng, Z.; Liu, T.; Lin, Z.; Lin, M. Emodin enhances ATRA-induced differentiation and induces apoptosis in acute myeloid leukemia cells. Int. J. Oncol. 2014, 45, 2076–2084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Musalli, M.G.; Hassan, M.; Sheikh, R.A.; Kalantan, A.A.; Halwani, M.; Zeyadi, M.; Hosawi, S.; Alhosin, M. Thymoquinone induces cell proliferation inhibition and apoptosis in acute myeloid leukemia cells: Role of apoptosis-related WT1 and BCL2 genes. Eur. J. Cell Sci. 2019, 1, 2–9. [Google Scholar] [CrossRef]
  81. Ahmed, N.; Laverick, L.; Sammons, J.; Zhang, H.; Maslin, D.J.; Hassan, H.T. Ajoene, a garlic-derived natural compound, enhances chemotherapy-induced apoptosis in human myeloid leukaemia CD34-positive resistant cells. Anticancer Res. 2001, 21, 3519–3523. [Google Scholar]
  82. Bai, L.Y.; Weng, J.R.; Chiu, C.F.; Wu, C.Y.; Yeh, S.P.; Sargeant, A.M.; Lin, P.H.; Liao, Y.M. OSU-A9, an indole-3-carbinol derivative, induces cytotoxicity in acute myeloid leukemia through reactive oxygen species-mediated apoptosis. Biochem. Pharmacol. 2013, 86, 1430–1440. [Google Scholar] [CrossRef]
  83. Eden, R.E.; Coviello, J.M. Chronic Myelogenous Leukemia; StatPearls Publishing: Treasure Island, FL, USA, 2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK531459/ (accessed on 3 March 2021).
  84. Rohrbacher, M.; Hasford, J. Epidemiology of chronic myeloid leukaemia (CML). Best Pract. Res. Clin. Haematol. 2009, 22, 295–302. [Google Scholar] [CrossRef]
  85. Medina, J.; Kantarjian, H.; Talpaz, M.; O’Brien, S.; Garcia-Manero, G.; Giles, F.; Rios, M.B.; Hayes, K.; Cortes, J. Chromosomal abnormalities in Philadelphia chromosome-negative metaphases appearing during imatinib mesylate therapy in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase. Cancer 2003, 98, 1905–1911. [Google Scholar] [CrossRef]
  86. Sillaber, C.; Mayerhofer, M.; Agis, H.; Sagaster, V.; Mannhalter, C.; Sperr, W.R.; Geissler, K.; Valent, P. Chronic myeloid leukemia: Pathophysiology, diagnostic parameters, and current treatment concepts. Wien Klin. Wochenschr. 2003, 115, 485–504. [Google Scholar] [CrossRef]
  87. Mencalha, A.L.; Correa, S.; Abdelhay, E. Role of calcium-dependent protein kinases in chronic myeloid leukemia: Combined effects of PKC and BCR-ABL signaling on cellular alterations during leukemia development. Onco Targets Ther. 2014, 7, 1247–1254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Steelman, L.S.; Pohnert, S.C.; Shelton, J.G.; Franklin, R.A.; Bertrand, F.E.; McCubrey, J.A. JAK/STAT, Raf/MEK/ERK, PI3K/Akt and BCR-ABL in cell cycle progression and leukemogenesis. Leukemia 2004, 18, 189–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Solmaz, S.; Adan Gokbulut, A.; Cincin, B.; Ozdogu, H.; Boga, C.; Cakmakoglu, B.; Kozanoglu, I.; Baran, Y. Therapeutic potential of apigenin, a plant flavonoid, for imatinib-sensitive and resistant chronic myeloid leukemia cells. Nutr. Cancer. 2014, 66, 599–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Zaric, M.; Mitrovic, M.; Nikolic, I.; Baskic, D.; Popovic, S.; Djurdjevic, P.; Milosavljevic, Z.; Zelen, I. Chrysin induces apoptosis in peripheral blood lymphocytes isolated from human chronic lymphocytic leukemia. Anticancer Agents Med. Chem. 2015, 15, 189–195. [Google Scholar] [CrossRef] [PubMed]
  91. Hassanzadeh, A.; Hosseinzadeh, E.; Rezapour, S.; Vahedi, G.; Haghnavaz, N.; Marofi, F. Quercetin promotes cell cycle arrest and apoptosis and attenuates the proliferation of human chronic myeloid leukemia cell line-K562 through interaction with HSPs (70 and 90), MAT2A and FOXM1. Anticancer Agents Med. Chem. 2019, 19, 1523–1534. [Google Scholar] [CrossRef] [PubMed]
  92. Li, W.; Zhao, Y.; Tao, B.; Zhang, Y. Effects of quercetin on hedgehog signaling in chronic myeloid leukemia KBM7 cells. Chin. J. Integr. Med. 2014, 20, 776–781. [Google Scholar] [CrossRef] [PubMed]
  93. Mutlu Altundağ, E.; Yılmaz, A.M.; Koçtürk, S.; Taga, Y.; Yalçın, A.S. Synergistic induction of apoptosis by quercetin and curcumin in chronic myeloid leukemia (K562) cells. Nutr. Cancer. 2018, 70, 97–108. [Google Scholar] [CrossRef] [PubMed]
  94. Carlo-Stella, C.; Dotti, G.; Mangoni, L.; Regazzi, E.; Garau, D.; Bonati, A.; Almici, C.; Sammarelli, G.; Savoldo, B.; Rizzo, M.T.; et al. Selection of myeloid progenitors lacking BCR/ABL mRNA in chronic myelogenous leukemia patients after in vitro treatment with the tyrosine kinase inhibitor genistein. Blood 1996, 88, 3091–3100. [Google Scholar] [CrossRef]
  95. Xiao, X.; Jiang, K.; Xu, Y.; Peng, H.; Wang, Z.; Liu, S.; Zhang, G. (−)-Epigallocatechin-3-gallate induces cell apoptosis in chronic myeloid leukaemia by regulating Bcr/Abl-mediated p38-MAPK/JNK and JAK2/STAT3/AKT signalling pathways. Clin. Exp. Pharmacol. Physiol. 2019, 46, 126–136. [Google Scholar] [CrossRef] [PubMed]
  96. Huang, Y.; Kumazoe, M.; Bae, J.; Yamada, S.; Takai, M.; Hidaka, S.; Yamashita, S.; Kim, Y.; Won, Y.; Murata, M.; et al. Green tea polyphenol epigallocatechin-O-gallate induces cell death by acid sphingomyelinase activation in chronic myeloid leukemia cells. Oncol. Rep. 2015, 34, 1162–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Feriotto, G.; Tagliati, F.; Giriolo, R.; Casciano, F.; Tabolacci, C.; Beninati, S.; Khan, M.T.H.; Mischiati, C. Caffeic acid enhances the anti-leukemic effect of imatinib on chronic myeloid leukemia cells and triggers apoptosis in cells sensitive and resistant to imatinib. Int. J. Mol. Sci. 2021, 22, 1644. [Google Scholar] [CrossRef] [PubMed]
  98. Bandyopadhyay, G.; Biswas, T.; Roy, K.C.; Mandal, S.; Mandal, C.; Pal, B.C.; Bhattacharya, S.; Rakshit, S.; Bhattacharya, D.K.; Chaudhuri, U.; et al. Chlorogenic acid inhibits Bcr-Abl tyrosine kinase and triggers p38 mitogen-activated protein kinase-dependent apoptosis in chronic myelogenous leukemic cells. Blood 2004, 104, 2514–2522. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, C.G.; Zhong, L.; Liu, Y.L.; Shi, X.J.; Shi, L.Q.; Zeng, L.; Liu, B.Z. Emodin exerts an antiapoptotic effect on human chronic myelocytic leukemia K562 cell lines by targeting the PTEN/PI3K-AKT signaling pathway and deleting BCR-ABL. Integr. Cancer Ther. 2017, 16, 526–539. [Google Scholar] [CrossRef]
  100. Chen, J.; Zhou, M.; Zhang, Q.; Xu, J.; Ouyang, J. Anticancer effect and apoptosis induction of gambogic acid in human leukemia cell line K562 in vitro. Med. Sci. Monit. 2015, 21, 1604–1610. [Google Scholar] [CrossRef] [Green Version]
  101. Shi, X.; Chen, X.; Li, X.; Lan, X.; Zhao, C.; Liu, S.; Huang, H.; Liu, N.; Liao, S.; Song, W.; et al. Gambogic acid induces apoptosis in imatinib-resistant chronic myeloid leukemia cells via inducing proteasome inhibition and caspase-dependent Bcr-Abl downregulation. Clin. Cancer Res. 2014, 20, 151–163. [Google Scholar] [CrossRef] [Green Version]
  102. Taverna, S.; Giallombardo, M.; Pucci, M.; Flugy, A.; Manno, M.; Raccosta, S.; Rolfo, C.; De Leo, G.; Alessandro, R. Curcumin inhibits in vitro and in vivo chronic myelogenous leukemia cells growth: A possible role for exosomal disposal of miR-21. Oncotarget 2015, 6, 21918–21933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Wu, X.P.; Xiong, M.; Xu, C.S.; Duan, L.N.; Dong, Y.Q.; Luo, Y.; Niu, T.H.; Lu, C.R. Resveratrol induces apoptosis of human chronic myelogenous leukemia cells in vitro through p38 and JNK-regulated H2AX phosphorylation. Acta Pharmacol. Sin. 2015, 36, 353–361. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, B.; Liu, J.; Gong, Z. Resveratrol induces apoptosis in K562 cells via the regulation of mitochondrial signaling pathways. Int. J. Clin. Exp. Med. 2015, 8, 16926–16933. [Google Scholar] [PubMed]
  105. Can, G.; Cakir, Z.; Kartal, M.; Gunduz, U.; Baran, Y. Apoptotic effects of resveratrol, a grape polyphenol, on imatinib-sensitive and resistant K562 chronic myeloid leukemia cells. Anticancer Res. 2012, 32, 2673–2678. [Google Scholar]
  106. Roy, M.; Sarkar, R.; Mukherjee, A.; Mukherjee, S. Inhibition of crosstalk between Bcr-Abl and PKC signaling by PEITC, augments imatinib sensitivity in chronic myelogenous leukemia cells. Chem. Biol. Interact. 2015, 242, 195–201. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, Y.; Wei, S.; Wang, J.; Fang, Q.; Chai, Q. Phenethyl isothiocyanate inhibits growth of human chronic myeloid leukemia K562 cells via reactive oxygen species generation and caspases. Mol. Med. Rep. 2014, 10, 543–549. [Google Scholar] [CrossRef] [Green Version]
  108. Safa, M.; Jafari, L.; Alikarami, F.; Manafi Shabestari, R.; Kazemi, A. Indole-3-carbinol induces apoptosis of chronic myelogenous leukemia cells through suppression of STAT5 and Akt signaling pathways. Tumour Biol. 2017, 39. [Google Scholar] [CrossRef] [Green Version]
  109. Chun-Guang, W.; Jun-Qing, Y.; Bei-Zhong, L.; Dan-Ting, J.; Chong, W.; Liang, Z.; Dan, Z.; Yan, W. Anti-tumor activity of emodin against human chronic myelocytic leukemia K562 cell lines in vitro and in vivo. Eur. J. Pharmacol. 2010, 627, 33–41. [Google Scholar] [CrossRef]
  110. Ozkan, T.; Hekmatshoar, Y.; Pamuk, H.; Ozcan, M.; Yaman, G.; Yagiz, G.C.; Akdemir, C.; Sunguroglu, A. Cytotoxic effect of 6-Shogaol in Imatinib sensitive and resistant K562 cells. Mol. Biol. Rep. 2021, 48, 1625–1631. [Google Scholar] [CrossRef] [PubMed]
  111. Flores-Lopez, G.; Moreno-Lorenzana, D.; Ayala-Sanchez, M.; Aviles-Vazquez, S.; Torres-Martinez, H.; Crooks, P.A.; Guzman, M.L.; Mayani, H.; Chávez-González, A. Parthenolide and DMAPT induce cell death in primitive CML cells through reactive oxygen species. J. Cell. Mol. Med. 2018, 22, 4899–4912. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, T.; Du, J.; Kong, D.; Yang, G.; Zhou, Q.; You, F.; Lin, Y.; Wang, Y. Gambogic acid inhibits proliferation and induces apoptosis of human acute T-cell leukemia cells by inducing autophagy and downregulating β-catenin signaling pathway: Mechanisms underlying the effect of Gambogic acid on T-ALL cells. Oncol. Rep. 2020, 44, 1747–1757. [Google Scholar] [CrossRef] [PubMed]
  113. Van Vlierberghe, P.; Ferrando, A. The molecular basis of T cell acute lymphoblastic leukemia. J. Clin. Investig. 2012, 122, 3398–3406. [Google Scholar] [CrossRef] [Green Version]
  114. Goldberg, J.M.; Silverman, L.B.; Levy, D.E.; Dalton, V.K.; Gelber, R.D.; Lehmann, L.; Cohen, H.J.; Sallan, S.E.; Asselin, B.L. Childhood T-cell acute lymphoblastic leukemia: The Dana-Farber Cancer Institute acute lymphoblastic leukemia consortium experience. J. Clin. Oncol. 2003, 21, 3616–3622. [Google Scholar] [CrossRef] [PubMed]
  115. Zi, C.T.; Gao, Y.S.; Yang, L.; Feng, S.Y.; Huang, Y.; Sun, L.; Jin, Y.; Xu, F.Q.; Dong, F.W.; Li, Y.; et al. Design, synthesis, and biological evaluation of novel biotinylated podophyllotoxin derivatives as potential antitumor agents. Front. Chem. 2019, 7, 434. [Google Scholar] [CrossRef]
  116. Popović, D.; Đukić, D.; Katić, V.; Jović, Z.; Jović, M.; Lalić, J.; Golubović, I.; Stojanović, S.; Ulrih, N.P.; Stanković, M.; et al. Antioxidant and proapoptotic effects of anthocyanins from bilberry extract in rats exposed to hepatotoxic effects of carbon tetrachloride. Life Sci. 2016, 157, 168–177. [Google Scholar] [CrossRef] [PubMed]
  117. León-González, A.J.; Sharif, T.; Auger, C.; Abbas, M.; Fuhrmann, G.; Schini-Kerth, V.B. Anthocyanin-rich bilberry extract induces apoptosis in acute lymphoblastic leukemia cells via redox-sensitive epigenetic modifications. J. Funct. Foods 2018, 44, 227–234. [Google Scholar] [CrossRef]
  118. Sorrenti, V.; Di Giacomo, C.; Acquaviva, R.; Bognanno, M.; Grilli, E.; D’Orazio, N.; Galvano, F. Dimethylarginine dimethylaminohydrolase/nitric oxide synthase pathway in liver and kidney: Protective effect of cyanidin 3-O-β-D-glucoside on ochratoxin-A toxicity. Toxins 2012, 4, 353–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Schumacher, M.; Hautzinger, A.; Rossmann, A.; Holzhauser, S.; Popovic, D.; Hertrampf, A.; Oesterle, D.; Spiller, C.; Boll, M.; Wenzel, U. Potential role of P-gp for flavone-induced diminished apoptosis and increased adenoma size in the small intestine of APC(min/+) mice. Cancer Investig. 2011, 29, 396–404. [Google Scholar] [CrossRef]
  120. Feng, R.; Ni, H.M.; Wang, S.Y.; Tourkova, I.L.; Shurin, M.R.; Harada, H.; Yin, X.M. Cyanidin-3-rutinoside, a natural polyphenol antioxidant, selectively kills leukemic cells by induction of oxidative stress. J. Biol. Chem. 2007, 282, 13468–13476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. León-González, A.J.; Sharif, T.; Kayali, A.; Abbas, M.; Dandache, I.; Etienne-Selloum, N.; Kevers, C.; Pincemail, J.; Auger, C.; Chabert, P.; et al. Delphinidin-3-O-glucoside and delphinidin-3-O-rutinoside mediate the redox-sensitive caspase 3-related pro-apoptotic effect of blackcurrant juice on leukaemia Jurkat cells. J. Funct. Foods 2015, 17, 847–856. [Google Scholar] [CrossRef]
  122. Shi, Y.; Su, X.; Cui, H.; Yu, L.; Du, H.; Han, Y. Combination of quercetin and Adriamycin effectively suppresses the growth of refractory acute leukemia. Oncol. Lett. 2019, 18, 153–160. [Google Scholar] [CrossRef] [Green Version]
  123. Goto, H.; Yanagimachi, M.; Goto, S.; Takeuchi, M.; Kato, H.; Yokosuka, T.; Kajiwara, R.; Yokota, S. Methylated chrysin reduced cell proliferation, but antagonized cytotoxicity of other anticancer drugs in acute lymphoblastic leukemia. Anticancer Drugs 2012, 23, 417–425. [Google Scholar] [CrossRef] [PubMed]
  124. Ghasemi-Pirbaluti, M.; Pourgheysari, B.; Shirzad, H.; Sourani, Z.; Beshkar, P. The inhibitory effect of epigallocatechin gallate on the viability of T lymphoblastic leukemia cells is associated with increase of caspase-3 level and Fas expression. Indian J. Hematol. Blood Transfus. 2018, 34, 253–260. [Google Scholar] [CrossRef]
  125. Kuttikrishnan, S.; Siveen, K.S.; Prabhu, K.S.; Khan, A.Q.; Ahmed, E.I.; Akhtar, S.; Ali, T.A.; Merhi, M.; Dermime, S.; Steinhoff, M.; et al. Curcumin induces apoptotic cell death via inhibition of PI3-kinase/AKT pathway in B-precursor acute lymphoblastic leukemia. Front. Oncol. 2019, 9, 484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. William, B.M.; Goodrich, A.; Peng, C.; Li, S. Curcumin inhibits proliferation and induces apoptosis of leukemic cells expressing wild-type or T315I-BCR-ABL and prolongs survival of mice with acute lymphoblastic leukemia. Hematology 2008, 13, 333–343. [Google Scholar] [CrossRef] [PubMed]
  127. Ge, J.; Liu, Y.; Li, Q.; Guo, X.; Gu, L.; Ma, Z.G.; Zhu, Y.P. Resveratrol induces apoptosis and autophagy in T-cell acute lymphoblastic leukemia cells by inhibiting Akt/mTOR and activating p38-MAPK. Biomed. Environ. Sci. 2013, 26, 902–911. [Google Scholar] [CrossRef]
  128. Ramezani, G.; Pourgheysari, B.; Shirzad, H.; Sourani, Z. Pterostilbene increases Fas expression in T-lymphoblastic leukemia cell lines. Res. Pharm. Sci. 2019, 14, 55–63. [Google Scholar] [CrossRef] [PubMed]
  129. Sourani, Z.; Pourgheysari, B.; Rafieian-Kopaei, M.; Shirzad, H.; Shirzad, M. The effect of gallic acid on Jurkat cell line. J. HerbMed Pharmacol. 2015, 4, 129–132. [Google Scholar]
  130. Diamanti, P.; Cocs, C.V.; Moppett, J.P.; Blair, A. Parthenolide eliminates leukemia-initiating cell populations and improves survival in xenografts of childhood acute lymphoblastic leukemia. Blood 2013, 121, 1384–1393. [Google Scholar] [CrossRef] [Green Version]
  131. Soltani, A.; Pourgheysari, B.; Shirzad, H.; Sourani, Z. Antiproliferative and apoptosis-inducing activities of thymoquinone in lymphoblastic leukemia cell line. Indian J. Hematol. Blood Transfus. 2017, 33, 516–524. [Google Scholar] [CrossRef]
  132. Salim, L.Z.; Mohan, S.; Othman, R.; Abdelwahab, S.I.; Kamalidehghan, B.; Sheikh, B.Y.; Ibrahim, M.Y. Thymoquinone induces mitochondria-mediated apoptosis in acute lymphoblastic leukaemia in vitro. Molecules 2013, 18, 11219–11240. [Google Scholar] [CrossRef]
  133. Safa, M.; Tavasoli, B.; Manafi, R.; Kiani, F.; Kashiri, M.; Ebrahimi, S.; Kazemi, A. Indole-3-carbinol suppresses NF-κB activity and stimulates the p53 pathway in pre-B acute lymphoblastic leukemia cells. Tumour Biol. 2015, 36, 3919–3930. [Google Scholar] [CrossRef]
  134. Hallek, M. Chronic lymphocytic leukemia: 2013 update on diagnosis, risk stratification and treatment. Am. J. Hematol. 2013, 88, 803–816. [Google Scholar] [CrossRef]
  135. Stephens, J.M.; Gramegna, P.; Laskin, B.; Botteman, M.F.; Pashos, C.L. Chronic lymphocytic leukemia: Economic burden and quality of life: Literature review. Am. J. Ther. 2005, 12, 460–466. [Google Scholar] [CrossRef] [PubMed]
  136. Mukkamalla, S.K.R.; Taneja, A.; Malipeddi, D.; Master, S.R. Chronic Lymphocytic Leukemia; StatPearls Publishing: Treasure Island, FL, USA, 2020. Available online: https://pubmed.ncbi.nlm.nih.gov/29261864/ (accessed on 2 March 2021).
  137. Golombick, T.; Diamond, T.H.; Manoharan, A.; Ramakrishna, R. B-cell disorders and curcumin. Integr. Cancer Ther. 2017, 16, 255–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Rozman, C.; Montserrat, E. Chronic lymphocytic leukemia. N. Engl. J. Med. 1995, 333, 1052–1057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Byrd, J.C.; Stilgenbauer, S.; Flinn, I.W. Chronic lymphocytic leukemia. Hematol. Am. Soc. Hematol. Educ. Program. 2004, 163–183. [Google Scholar] [CrossRef] [PubMed]
  140. Gokbulut, A.A.; Apohan, E.; Baran, Y. Resveratrol and quercetin-induced apoptosis of human 232B4 chronic lymphocytic leukemia cells by activation of caspase-3 and cell cycle arrest. Hematology 2013, 18, 144–150. [Google Scholar] [CrossRef] [Green Version]
  141. Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef] [PubMed]
  142. Balakrishnan, K.; Gandhi, V. Bcl-2 antagonists: A proof of concept for CLL therapy. Invest. New Drugs. 2013, 31, 1384–1394. [Google Scholar] [CrossRef] [Green Version]
  143. Hamblin, T. Natural products and the treatment of leukemia. Leuk. Res. 2006, 30, 649–650. [Google Scholar] [CrossRef] [PubMed]
  144. Alhosin, M.; León-González, A.J.; Dandache, I.; Lelay, A.; Rashid, S.K.; Kevers, C.; Pincemail, J.; Fornecker, L.M.; Mauvieux, L.; Herbrecht, R.; et al. Bilberry extract (Antho 50) selectively induces redox-sensitive caspase 3-related apoptosis in chronic lymphocytic leukemia cells by targeting the Bcl-2/Bad pathway. Sci. Rep. 2015, 5, 8996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Sak, K.; Kasemaa, K.; Everaus, H. Potentiation of luteolin cytotoxicity by flavonols fisetin and quercetin in human chronic lymphocytic leukemia cell lines. Food Funct. 2016, 7, 3815–3824. [Google Scholar] [CrossRef]
  146. Hashemi, M.; Nouri Long, M.; Entezari, M.; Nafisi, S.; Nowroozii, H. Anti-mutagenic and pro-apoptotic effects of apigenin on human chronic lymphocytic leukemia cells. Acta Med. Iran. 2010, 48, 283–288. [Google Scholar] [PubMed]
  147. Lee, Y.K.; Bone, N.D.; Strege, A.K.; Shanafelt, T.D.; Jelinek, D.F.; Kay, N.E. VEGF receptor phosphorylation status and apoptosis is modulated by a green tea component, epigallocatechin-3-gallate (EGCG), in B-cell chronic lymphocytic leukemia. Blood 2004, 104, 788–794. [Google Scholar] [CrossRef] [Green Version]
  148. Salimi, A.; Roudkenar, M.H.; Seydi, E.; Sadeghi, L.; Mohseni, A.; Pirahmadi, N.; Pourahmad, J. Chrysin as an anti-cancer agent exerts selective toxicity by directly inhibiting mitochondrial complex II and V in CLL B-lymphocytes. Cancer Investig. 2017, 35, 174–186. [Google Scholar] [CrossRef]
  149. Roman, V.; Billard, C.; Kern, C.; Ferry-Dumazet, H.; Izard, J.C.; Mohammad, R.; Mossalayi, D.M.; Kolb, J.P. Analysis of resveratrol-induced apoptosis in human B-cell chronic leukaemia. Br. J. Haematol. 2002, 117, 842–851. [Google Scholar] [CrossRef] [PubMed]
  150. Golombick, T.; Diamond, T.; Manoharan, A.; Ramakrishna, R. The effect of curcumin (as meriva) on absolute lymphocyte count (ALC), NK cells and T cell populations in patients with stage 0/1 chronic lymphocytic leukemia. J. Cancer Ther. 2015, 06, 566–571. [Google Scholar] [CrossRef] [Green Version]
  151. Hayun, R.; Okun, E.; Berrebi, A.; Shvidel, L.; Bassous, L.; Sredni, B.; Nir, U. Rapamycin and curcumin induce apoptosis in primary resting B chronic lymphocytic leukemia cells. Leuk. Lymphoma 2009, 50, 625–632. [Google Scholar] [CrossRef]
  152. Loisel, S.; Le Ster, K.; Meyer, M.; Berthou, C.; Youinou, P.; Kolb, J.P.; Billard, C. Therapeutic activity of two xanthones in a xenograft murine model of human chronic lymphocytic leukemia. J. Hematol. Oncol. 2010, 3, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Liu, H.; Gu, L.B.; Tu, Y.; Hu, H.; Huang, Y.R.; Sun, W. Emodin ameliorates cisplatin-induced apoptosis of rat renal tubular cells in vitro by activating autophagy. Acta Pharmacol. Sin. 2016, 37, 235–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Steele, A.J.; Jones, D.T.; Ganeshaguru, K.; Duke, V.M.; Yogashangary, B.C.; North, J.M.; Lowdell, M.W.; Kottaridis, P.D.; Mehta, A.B.; Prentice, A.G.; et al. The sesquiterpene lactone parthenolide induces selective apoptosis of B-chronic lymphocytic leukemia cells in vitro. Leukemia 2006, 20, 1073–1079. [Google Scholar] [CrossRef] [PubMed]
  155. Marín, G.; Mansilla, E. Parthenolide has apoptotic and cytotoxic selective effect on B-chronic lymphocytic leukemia cells. J. Appl. Biomed. 2006, 4, 135–139. [Google Scholar] [CrossRef] [Green Version]
  156. Arditti, F.D.; Rabinkov, A.; Miron, T.; Reisner, Y.; Berrebi, A.; Wilchek, M.; Mirelman, D. Apoptotic killing of B-chronic lymphocytic leukemia tumor cells by allicin generated in situ using a rituximab-alliinase conjugate. Mol. Cancer Ther. 2005, 4, 325–331. [Google Scholar]
  157. Perez-Chacon, G.; Martinez-Laperche, C.; Rebolleda, N.; Somovilla-Crespo, B.; Muñoz-Calleja, C.; Buño, I.; Zapata, J.M. Indole-3-carbinol synergizes with and restores fludarabine sensitivity in chronic lymphocytic leukemia cells irrespective of p53 activity and treatment resistances. Clin. Cancer Res. 2016, 22, 134–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Shanafelt, T.D.; Call, T.G.; Zent, C.S.; LaPlant, B.; Bowen, D.A.; Roos, M.; Secreto, C.R.; Ghosh, A.K.; Kabat, B.F.; Lee, M.J.; et al. Phase I trial of daily oral Polyphenon E in patients with asymptomatic Rai stage 0 to II chronic lymphocytic leukemia. J. Clin. Oncol. 2009, 27, 3808–3814. [Google Scholar] [CrossRef] [PubMed]
  159. Shanafelt, T.D.; Call, T.G.; Zent, C.S.; Leis, J.F.; LaPlant, B.; Bowen, D.A.; Roos, M.; Laumann, K.; Ghosh, A.K.; Lesnick, C.; et al. Phase 2 trial of daily, oral Polyphenon E in patients with asymptomatic, Rai stage 0 to II chronic lymphocytic leukemia. Cancer 2013, 119, 363–370. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Natural compounds against acute myeloid leukemia (AML).
Figure 1. Natural compounds against acute myeloid leukemia (AML).
Molecules 26 02709 g001
Figure 2. Natural compounds against chronic myeloid leukemia (CML).
Figure 2. Natural compounds against chronic myeloid leukemia (CML).
Molecules 26 02709 g002
Figure 3. Natural compounds against acute lymphoblastic leukemia (ALL).
Figure 3. Natural compounds against acute lymphoblastic leukemia (ALL).
Molecules 26 02709 g003
Figure 4. Natural compounds against chronic lymphocytic leukemia (CLL).
Figure 4. Natural compounds against chronic lymphocytic leukemia (CLL).
Molecules 26 02709 g004
Table 1. Pharmacological effects of natural compounds in acute myeloid leukemia (AML).
Table 1. Pharmacological effects of natural compounds in acute myeloid leukemia (AML).
Bioactive CompoundIn Vitro/In Vivo/Clinical StudyCancer Cell Line and Animal ModelBioactive EffectReferences
LuteolinIn vitroMOLM-13 and Kasumi-1 cells-inhibited leukemic cell proliferation and induced apoptosis by inhibition of the RSK1 pathways
-triggered RSK-dependent antileukemic responses with dephosphorylation of Bad or KIBRA
[46]
EGCGIn vitroNB4 and HL60 cells-induced cell death in myeloid leukemic cells
-↑ DAPK2 levels in AML cells
-EGCG/ATRA cotreatment of myeloid leukemic cells enhanced neutrophil differentiation
[62]
(−)-EpicatechinIn vivoBrown Norway rats↑ the in vivo apoptotic effect of etoposide
↑ the oxidative stress induced by etoposide in leukemic rats
[63]
QuercetinIn vitroMV4-11 and HL-60 cells-promoted AML cell death
-induced caspase-dependent apoptosis in AML cells
-induced apoptosis via mitochondrial pathway
-suppressed VEGFR2 and PI3K/Akt signaling pathway
[39]
QuercetinIn vitroHL60 and U937 cells-down-regulated DNMTs and STAT3
-induced H3 and H4 global acetylation
-enriched H3ac and H4ac in the promoter region of the apoptosis pathway genes and increased their transcription levels
↓ the protein expression of class I HDACs in leukemia cells
-caused proteasome-mediated protein degradation of HADCs in leukemia cells
-down-regulated DNMTs and HADCs at the protein levels, in xenograft models
[64]
QuercetinIn vitrohuman myeloid leukemia KG-1 cells-cytotoxicity effect against KG-1 cells
-augmented the TRAIL-induced cell death in KG-1 cells
↑ mRNA expression levels of DR genes in acute myeloid KG-1 cells
↓ mRNA expression of apoptosis inhibitor genes in the acute myeloid KG-1 cells
↓ mRNA expression of NF-κB (p65 subunit) gene in the acute myeloid KG-1 cells
[5]
QuercetinIn vitroP39 cells-induced apoptosis in P39 leukemia cells
↓ Bcl-2, Bcl-xL, Mcl-1 down-regulation
↓ Bax
-induced mitochondrial translocation, triggering cytochrome c release and caspases activation
[65]
In vivoNOD.CB17-Prkdcscid/J
mice
-induced the expression of FasL protein
↑ cell arrest in G1 phase of the cell cycle
↓ in CDK2, CDK6, cyclin D, cyclin E, and cyclin A proteins
↓ Rb phosphorylation
↑ p21 and p27 expression
-induced autophagosome formation in P39 cell line
↓ tumor volume in P39 xenografts in vivo
Quercetin and green teaIn vivoNOD/SCID mice↓ tumor growth in HL-60 xenografts accompanied by decreased expression of anti-apoptotic proteins, Bcl-2, BCL-xL, and Mcl-1 and increased expression of Bax, a pro-apoptotic protein
-induced apoptosis of leukemic cells
-induced activation of caspase-3
-induced cell cycle arrest of leukemic cells
-mediated G1 phase cell cycle arrest in HL-60 xenografts
-induced conversion of LC3-I to LC3-II
↑ autophagy in leukemic cells
[41]
ChrysinIn vitroMO7e cells-inhibited SCF/c-Kit complex-induced cell proliferation in human myeloid leukemia cells
-inhibited SCF-induced phosphorylation of c-Kit
-inhibited cell proliferation in MO7e cells by blocking c-Kit phosphorylation
[66]
GenisteinIn vitroMV4-11 and HL-60 cells-arrested the mTOR pathway leading to down-regulation of protein synthesis
-induced cell death via apoptosis
-regulatory effects on the cell cycle of the two cell lines, with the induction of G2⁄M phase arrest in HL-60 cells but not in MV4-11 cells
[67]
Gallic acidIn vitroTHP-1 and MV411 cells-induced caspase-dependent apoptosis of AML cell lines, primary MNC and CD34 stem/progenitors isolated from AML patients via caspase-dependent pathway
-enhanced cytarabine and daunorubicin efficacy in vitro cell culture system and in vivo xenograft model
-inhibited mitochondrial respiration in AML cells, leading to decreased ATP production and oxidative stress
-acted on AML cells via Akt/mTOR-dependent inhibition of mitochondrial respiration
[68]
Caffeic acid phenyl ester (CAPE)In vitroU937 cells↓ cell viability of U937 cells
-induced the mitochondria-mediated apoptosis-release of cytochrome C, reduction of Bcl-2 expression, increase of Bax expression, activation/cleavage of caspase-3, and activation/cleavage of PARP
[69]
CurcuminIn vitroHL-60 cells-potentiated the cytotoxic effect of etoposide
-intensified apoptosis and phosphorylation of the histone H2AX induced by this cytostatic drug in leukemic HL-60 cells
-curcumin modified the cytotoxic action of etoposide in HL-60 cells through intensification of ROS production
-enhanced the antileukemic activity of etoposide in BNML rats and induced apoptosis of BNML cells more efficiently than etoposide alone, but this treatment protected nonleukemic B-cells from apoptosis
[70]
In vivoBrown Norway rats with acute myeloid leukemia (BNML)
ResveratrolIn vitroCD34+ CD38 KG1a cells↓ pLKB1 in CD34+ CD38 KG1a cells
↑ the expression of SIRT1 in CD34+ CD38 KG1a cells
-induced senescence and apoptosis of CD34+CD38 KG1a cells
[71]
ResveratrolIn vitroHL-60 cells↓ CSC-related Shh expression, Gli-1 nuclear translocation, and cell viability in IL-6-treated HL-60 cells
-had synergistic effect with Shh inhibitor cyclopamine on inhibiting cell growth
[72]
Resveratrol In vitroU937 and MV-4-11 cells-interacted synergistically with HDACIs in human myeloid leukemia cells
-coadministration with HDACIs led to enhanced DNA damage, mitochondrial injury, and caspase-3, caspase-9, and caspase-8 activation
-blocked HDACI-mediated RelA acetylation and NF-κB activation
-induced S-phase accumulation and sensitized leukemia cells to HDACIs
[73]
PterostilbeneIn vitroMV4-11 HL-60, U937, and THP-1 AML cells-suppressed cell proliferation in various AML cell lines
-induced G0/G1-phase arrest when expressions of cyclin D3 and CDK2/6 were inhibited
-induced cell apoptosis occurred through activation of caspases-8/-9/-3, and a MMP-dependent pathway
-treatment of HL-60 cells with PTER induced sustained activation of ERK1/2 and JNK1/2, and inhibition of both MAPKs by their specific inhibitors significantly abolished the PTER-induced activation of caspases-8/-9/-3
-PTER-induced cell growth inhibition was only partially reversed by the caspase-3-specific inhibitor, Z-DEVE-FMK
-promoted disruption of LMP and release activated cathepsin B
-induced HL-60 cell death via MAPKs-mediated mitochondria apoptosis pathway
[74]
Gambogic acidIn vitroU937 and HL-60 cells-had cytotoxic effect on AML cells
-inhibited cell growth and promoted differentiation in U937 and HL-60 cells
↑ the expression of p21waf1/cip1 in the two cell lines
[75]
3-O-acetyl-11-keto-β-boswellic acid (AKBA)In vitroHL-60 cells-inhibited dose-dependent proliferation of HL-60 and apoptosis rate of HL-60 cells
-changed the cell cycle by increasing of G(1) phase and decreasing of S phase
-anti-proliferation and apoptosis-inducing effects on HL-60 cells
[76]
Boswellic acid acetateIn vitroNB4, SKNO-1, nK562, U937, ML-1, and HL-60 cells-inhibited cell growth and induced cell toxicity of myeloid leukemia cell lines
-induced apoptosis through a p53-independent pathway by activation of caspase-8 induced proteolysis of Bid
↓ mitochondrial membrane potential without production of hydrogen peroxide
↑ the levels of DR4 and DR5 mRNA in apoptotic cells
[61]
Avocatin BIn vitroOCI-AML2 cells↓ human primary AML cell viability without effect on normal peripheral blood stem cells
-selectively toxic toward leukemia progenitor and stem cells
-induced mitochondria-mediated apoptosis
-inhibited fatty acid oxidation and ↓ NADPH levels, resulting in ROS-dependent leukemia cell death
[77]
ParthenolideIn vitroU937 cells-inhibited growth of U937 cells
-induced apoptosis in U937 cells
↓ the CD38+ population of U937 cells
↓ osteopontin gene expression in U937 cells
[78]
Parthenolide In vitroAML cells, bcCML cells, normal bone marrow, and umbilical cord blood cells-induced apoptosis in primary human AML cells and bcCML cells sparing normal hematopoietic cells
-targeted preferentially leukemic but not normal progenitor and stem cell activity
[43]
In vivoNonobese diabetic/severe NOD/SCID mice-the molecular mechanism of PTL mediated apoptosis is associated with inhibition of NF-κB, proapoptotic activation of p53, and increased ROS
-the activity of PTL triggers LSC-specific apoptosis
EmodinIn vitroAML HL-60/ADR cells-induced growth inhibition and apoptotic effects in resistant HL-60/ADR cells in vitro as well as in the HL-60/H3 xenograft models in vivo
↑ chemosensitivity of AML cells to Ara-C, inhibited leukemic cell growth, and improved survival in mouse xenograft model of AML
[37]
In vivoBALB/C-nude mice
EmodinIn vitroNB4, MR2 and primary AML cells -inhibited cell proliferation in NB4 cells, MR2 cells, and primary AML cells
-enhanced differentiation induction of ATRA in retinoid-responsive NB4 cells as well as in retinoid-resistant MR2 cells
-induced cell apoptosis in NB4 cells, MR2 cells, and primary AML cells
-the apoptotic induction in AML cells was associated with the activation of caspase cascades involving caspase-9, caspase-3, and PARP cleavage
-induced the activation of the caspase-dependent pathway
-induced the degradation of RARα protein in NB4 and MR2 cells
-inhibited activation of the PI3K/Akt signaling pathway in AML cells
-inhibited p-Akt at Ser473 as efficiently as mTOR at Ser2448
-suppressed the phosphoration of mTOR downstream targets, 4E-BP1 and p70S6K
[79]
ThymoquinoneIn vitroHL-60 cells↓ HL-60 cell viability
-induced apoptosis in HL-60 cells
↓ the expression of WT1 and BCL2 genes
[80]
Ajoene In vitroKG1 cells↓ bcl-2-expression
↑ the inhibitory effect of the two chemotherapeutic drugs, cytarabine and fludarabine, on Bcl-2-expression in KGI cells
-the two drugs, cytarabine and fludarabine, ↑ the activated caspase-3 level in KGI myeloid leukemia cells
-ajoene enhanced the activation of caspase-3 in both cytarabine- and fludarabine-treated KGI cells
[81]
OSU-A9In vitroHL-60 and THP-1 cells and primary leukemia cells from AML patients-induced cytotoxicity in AML cell lines and primary leukemia cells from AML patients
↓ cyclin A and cyclin B1 in AML cell lines
-induced apoptosis, caspase activation, and PARP cleavage in AML cell lines
-induced autophagy but not autophagic cell death in AML cell lines
-OSU-A9-mediated cytotoxicity and hypophosphorylation of Akt were dependent on the generation of ROS
-suppressed the growth of THP-1 xenograft tumors and prolonged the survival of tumor-bearing athymic nude mice
[82]
In vivoathymic nude mice
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; RSK1—ribosomal S6 kinase 1; RSK (ribosomal S6 kinase); Bad—Bcl-2-associated death promoter; KIBRA—kidney/brain protein; EGCG—epigallocatechin-3-gallate; HL-60—human promyelocytic leukemia; DAPK2—death-associated protein kinase 2; 67LR—67 kDa laminin receptor; ATRA—all-trans retinoic acid; VEGFR2—vascular endothelial growth factor receptor 2; PI3K/Akt signaling pathway—phosphatidylinositol 3-kinase/protein kinase B signaling pathway; DNMTs—DNA methyl transferases; STAT3—signal transducer and activator of transcription 3; HDACs—histone deacetylases; TRAIL—apoptosis-inducing ligand; mRNA—messenger ribonucleic acid; NF-κB—nuclear factor-κB; Bcl-2—B-cell lymphoma-2; Bcl-xL—B-cell lymphoma-extra-large; Mcl-1—myeloid cell leukemia 1; Bax—Bcl-2-associated X protein; CDK2—cyclin-dependent kinase 2; CDK6—cyclin-dependent kinase 6; Rb—retinoblastoma protein; SCF—stem cell factor; mTOR—mechanistic target of rapamycin; MNC—mononuclear cells; ATP—adenosine triphosphate; Akt—protein kinase B; CAPE—caffeic acid phenyl ester; PARP—poly(ADP-ribose) polymerase; BNML—Brown Norway rats with acute myeloid leukemia; pLKB1—phosphorylated liver kinase B1; SIRT1—Sirtuin 1; IL-6—interleukin 6; CSC—cancer stem cell; Shh—sonic hedgehog; Gli-1—glioma-associated oncogene homolog 1; HDACIs—histone deacetylase inhibitors; PTER—pterostilbene; MMP—mitochondrial membrane permeabilization; ERK1/2—extracellular signal-regulated kinase 1/2; JNK1/2—c-Jun N-terminal protein kinase 1/2; MAPKs—mitogen-activated protein kinases; Z-VAD-FMK—carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone; LMP—lysosomal membrane permeabilization; AKBA—3-O-acetyl-11-keto-β-boswellic acid; DR4 and DR5—death receptors 4 and 5; NADPH—nicotinamide adenine dinucleotide phosphate; ROS—reactive oxygen species; PTL—parthenolide; bcCML—blast crisis CML; LSCs—leukemia stem cells; Ara-C—cytarabine; RARα—retinoic acid receptor α; p-Akt—Akt phosphoration; WT1—Wilms’ tumor 1 gene.
Table 2. Pharmacological effects of natural compounds in chronic myeloid leukemia (CML).
Table 2. Pharmacological effects of natural compounds in chronic myeloid leukemia (CML).
Bioactive CompoundIn Vitro/In Vivo/Clinical StudyCancer Cell Line and Animal ModelBioactive EffectReferences
Apigenin In vitroK652 and K562/IMA3 cells-induced cytotoxic and apoptotic effects in K562 and K562/IMA3 cells
-induced loss of mitochondrial membrane potential in both K562 and K562/IMA3 cells
↑ caspase-3 activity in both K562 and K562/IMA3 Cells
-arrested cell cycle progression in G2/M phase in K562 cells
-induced S phase arrest in K562/IMA3 cells
-regulated a set of genes in K652 and K562/IMA3 cells
[89]
Chrysin In vitroMOLT-4 and JVM-13 cell lines, B-CLL cells derived from 28 patients and PBMC from 16 healthy subjects↓ the viability of of leukemic cells
-induced apoptosis of peripheral blood lymphocytes isolated from human CLL patients via mitochondrial pathway
-induced the activation of proapoptotic Bax
↓ the expression of antiapoptotic Bcl-2 protein
-released cytochrome c from mitochondria into cytosol
-activated caspase-3, subsequently leading to the activation of apoptosis of B-CLL cells
[90]
Quercetin In vitroK-562 cells-induced apoptosis in K-562 cells
-abrogated K-562 cells proliferation
↓ genes expression of HSP70, Bcl-X(L), and FOXM1
-improved Bax, caspase-3, and caspase-8 expression
[91]
QuercetinIn vitroKBM7 cells-inhibited KBM7 cell proliferation
-induced cell apoptosis
-blocked cell cycle at G1 phase
↓ the mRNA and protein expression of Smoothened and Glioma1 (Gli1)
↓ Bcl-2 and cyclin D1
↑ p53 and caspase-3 expression
-inhibited Hh signaling and its downstream targets in the KBM7 cells
[92]
Quercetin and curcumin K562 cells-induced changes in several genes in 10 different pathways related to cell proliferation, apoptosis, cell cycle, inflammation, hypoxia, and oxidative stress
↓ CDKN1B, AKT1, IFN-γ
↑ BTG2, CDKN1A, FAS
[93]
Genistein In vitroCML and CFU-Mix
BFU-E and CFU-GM hematopoietic progenitors
-suppressed colony formation
-suppressed progenitor cell growth
↓ marrow BCR/ABL+ progenitors
-exerted a strong antiproliferative effect on CFU-Mix, BFU-E, and CFU-GM
↓ the percentage of leukemic LTC-IC
-induced apoptosis of CML mononuclear and CD34+
[94]
EGCGIn vitroK562, K562R, KCL-22, BaF3/p210 and BaF3/p210T315I cell lines-inhibited the proliferation of CML cell lines and primary CML cells
↓ the mitochondrial membrane permeability of CML cell lines
-induced the apoptosis of CML cells through caspase-independent and AIF-mediated cell death pathways
-suppressed the expression of Bcr/Abl and phospho-Bcr/Abl in CML cell
-regulated Bcr/Abl downstream JAK2/STAT3/AKT and p38-MAPK/JNK signaling pathways in CML
[95]
EGCGIn vitroKU812 cells-induced ASM activation and lipid raft clustering in CML cells
-induced phosphorylation of protein kinase Cδ at Ser664
-induced cell death via the cGMP/ASM pathway in CML cells
[96]
Caffeic acidIn vitroK562 cells-induced mitochondrial membrane depolarization, genomic DNA fragmentation, and phosphatidylserine exposure, hallmarks of apoptosis
↓ cell proliferation
-↑ expression of two cell cycle repressor genes, CDKN1A and CHES1
[97]
Chlorogenic acidIn vitroK562, Molt 4, U937, THP-1,
REH cell lines
-induced apoptosis of several Bcr-Abl–positive CML cell lines and primary cells from CML patients in vitro
-destroyed Bcr-Abl–positive K562 cells in vivo
-no effect on the growth and viability of Bcr-Abl–negative lymphocytic and myeloid cell lines and primary CML cells
-↓ viability of Bcr-Abl–positive cells in vitro and in vivo
-induced apoptosis of Bcr-Abl–positive cells
-inhibited autophosphorylation of p210Bcr-Abl fusion protein
-modulated MAP kinase pathways in K562 cells
[98]
In vivoNude female mice
Emodin In vitroK562 cells-inhibited the growth of K562 cells harboring BCR-ABL in vitro and in vivo
-induced apoptosis by inhibition of PETN/PI3K/Akt level and deletion of BCR-ABL
[99]
Gambogic acidIn vitroK562 cells-inhibited the viability of K562 cells
-induced the accumulation of autophagic vacuoles and up-regulation of two autophagy-related proteins (Beclin 1 and LC3)
↓ mRNA levels of BCR/ABL fusion genes and SQSTM1/sequestosome 1 (p62) protein levels
-induced cell death through autophagy and apoptosis pathways in CML K562 cells
[100]
Gambogic acidIn vitroKBM5, KBM5-T315I, and K562 cells-induced apoptosis and cell proliferation inhibition in CML cells
-induced caspase activation in CML cells
-inhibited the proteasome function in CML cells
-down-regulated Bcr-Abl protein and inhibited its downstream signaling
-inhibited the growth of imatinib-resistant Bcr-Abl-T315I xenografts in nude mice
[101]
CurcuminIn vitroK562 and LAMA84 cells↓ miR-21 levels in CML cells
-induced PTEN expression in CML cells
↓ AKT phosphorylation and VEGF expression and release
↓ CML cells migration
↓ Bcr-Abl expression in CML cells through the cellular increase of miR-196b
-curcumin-treated mice developed smaller tumors
[102]
In vivoSCID mice
Resveratrol In vitroK562 cells-induced apoptosis and phosphorylation of H2AX at Ser139
-stimulated p38 and JNK activation in K562 cells during apoptosis
-p38 and JNK regulated resveratrol-induced H2AX phosphorylation in K562 cells
↓ phosphorylation of histone H3 at Ser10
[103]
Resveratrol In vitroK562 cells↓ cell viability and triggered cell apoptosis in K562 cells
↑ Bax/Bcl-2 ratio and release of cytochrome c into the cytosol
-induced the activation of caspase-3
↑ cleaved PARP
[104]
Resveratrol In vitroK562 and K562/IMA-3 cells-inhibited cell growth
↑ in loss of mitochondrial membrane potential
↑ caspase-3 activity
-induced apoptosis in K562 and K562/IMA-3 cells
[105]
Phenethyl isothiocyanate (PEITC)In vitroK-562, KU812 cells↑ cytotoxic efficacy of IM
PEITC in combination with IM down-regulated the expression of p210bcr/abl in chronic myelogenous leukemia cell lines (K-562)
-inhibited the expressions of PKCα, PKCβII, and PKCζ (both phosphorylated and total form)
↓ expression of Raf1 and ERK1/2, two important target proteins in PKC signaling cascade
↓ expression of Raf1 and ERK1/2 through Bcr-Abl and PKC inhibition
[106]
PEITCIn vitroK562 cells-induced cell death through the induction of ROS stress and oxidative damage
-suppressed cell growth and caused apoptosis by promoting Fas and Fas ligand expression, increasing ROS generation and by the successive release of cytochrome c as well as the activation of caspase-9 and caspase-3
[107]
Indole-3-carbinolIn vitroK562 cells-promoted mitochondrial apoptosis of CML-derived K562 cells, as evidenced by the activation of caspases and PARP cleavage
↓ the cellular levels of phospho-Akt and phospho-signal transducer and activator of transcription 5
-activated the p38 mitogen-activated protein kinase
↓ expression of human telomerase and c-Myc
[108]
Emodin In vitroK562 cells-inhibited K562 cell viability in vitro
-caused K562 cell morphological changes in vitro
-induced K562 cell division cycle arrest at G0/G1 phase in vitro
-induced K562 cell apoptosis in vitro and in vivo
↓ Bcl-2 ↑ Bax
-induced the activation of caspase-3, -8, and -9 in vitro and in vivo
↓ the tumor volume and tumor weight in nude mice
[109]
In vivoBALB/c nude mice
6-ShogaolIn vitroK562S and K562R cells-inhibited cell viability, induced apoptosis in both K562S and K562R
↑ pro-apoptotic Bax gene and ↓ anti-apoptotic BCL-2 gene expression levels significantly in both treated K562S and K562R cells
↑ MDR-1 mRNA expression level in K562S and K562R cells
↓ MRP-1 mRNA expression level in K562S cells
[110]
Parthenolide and DMAPTIn vitroK562, Meg-01, and KCL-22, HL-60 cells↓ viability of CML bulk and progenitor cells
-induced cell death in CML cells
↑ ROS levels in CML cells
-inhibited NF-κB activation in CML cells
-inhibited cell proliferation and arrested cell cycle of CML cells in G0 and G2 phases, correlated with down-regulation of cyclin D1 and cyclin A
[111]
Legend:↑ increased/up-regulated; ↓ decreased/down-regulated; Bax—Bcl-2-associated X protein; Bcl-2—B-cell lymphoma-2; B-CLL—B-cell chronic lymphocytic leukemia; HSP70—70 kilodalton heat shock proteins; Bcl-xL—B-cell lymphoma-extra-large; FOXM1- Forkhead box protein M1; Gli1—Smoothened and Glioma1; mRNA—messenger ribonucleic acid; Hh—Hedgehog; CDKN1B—cyclin-dependent kinase inhibitor 1B; Akt 1—protein kinase B 1; IFN-γ—interferon-gamma; BTG2—BTG anti-proliferation factor 2; CDKN1A—cyclin-dependent kinase inhibitor 1A; FAS—Fas cell surface death receptor; CFU-Mix—colony-forming unit-mix; BFU-E—burst-forming unit-erythroid; CFU-GM—granulocyte-macrophage colony-forming unit; LTC-IC—long-term culture initiating cell; AIF—apoptosis inducing factor; JAK2—Janus kinase 2; STAT3—signal transducer and activator of transcription 3; AKT—protein kinase B; MAPK—mitogen-activated kinase; JNK—c-Jun N-terminal kinase; ASM—acid sphingomyelinase; cGMP—cyclic guanosine monophosphate; CHES1—checkpoint suppressor 1; PI3K—phosphatidylinositol 3-kinase/protein kinase B; SQSTM1—sequestosome 1; PTEN—tumor suppressor gene phosphatase and tensin homolog; VEGF—vascular endothelial growth factor; miR-196b—microRNA 196b; PARP—poly(ADP-ribose) polymerase; IM—imatinib; PEITC—phenethyl isothiocyanate; Raf-1—proto-oncogene, serine/threonine kinase; ERK1/2—extracellular signal-regulated kinase 1/2; PKC—protein kinase C; ROS—reactive oxygen species; MDR-1—multidrug resistance mutation; MRP-1—multidrug resistance-associated protein 1; NF-κB—nuclear factor-κB; DMAPT—dimethyl amino parthenolide.
Table 3. Pharmacological effects of natural antioxidants in acute lymphoblastic leukemia (ALL).
Table 3. Pharmacological effects of natural antioxidants in acute lymphoblastic leukemia (ALL).
Bioactive CompoundIn Vitro/In Vivo/Clinical StudyCancer Cell Line and Animal ModelBioactive EffectReferences
QuercetinIn vivoC57BL/6J (CD45.2+) and B6.SJL-PtprcaPepcb/BoyJ mice-enhanced the cytotoxicity of Adriamycin to leukemic cells
-improved the survival of mice with T-ALL
-enhanced the SOD activity and reduced the MDA content in the heart
[122]
Antho 50In vitroJurkat cells-induced apoptosis in Jurkat cells
↑ ROS formation
↑ tumor suppressor p73 and cell cycle regulator p21 expression levels
-cleaved caspase-3 expression levels
↓ expression levels of p-Akt, survivin, PcG proteins, HDACs, DNMT1, and UHRF1
[117]
Delphinidin-3-O-glucoside and delphinidin-3-O-rutinosideIn vitroJurkat and Molt-4 cell lines-induced proapoptotic response in Jurkat cells[121]
DMFIn vitroYCUB series-induced G0/G1 cell cycle arrest
↓ the expression of phosphorylated retinoblastoma-associated protein 1
↑ induced apoptosis in ALL cell lines
↓ the intracellular levels of glutathione
-antagonized the cytotoxicity of 4-hydroperoxy-cyclophosphamide, cytarabine, vincristine, and L-asparaginase in all tested ALL cells
[123]
EGCGIn vitroJurkat cells-decreased viability of cells
-induced apoptosis of lymphoblastic leukemia cells
-enhanced Fas expression in Jurkat cells
-increased caspase-3 positive cells
[124]
CurcuminIn vitro697, REH, RS4;11, and SupB15 cells -suppressed the viability in B-Pre-ALL cell lines
-induced apoptosis in B-Pre-ALL cell lines via activation of caspase-8 and truncation of BID protein
↑ the ratio of Bax/Bcl-2
-induced the dephosphorylation of the constitutive phosphorylated AKT/PKB
↓ the expression of cIAP1, and XIAP
↑ ROS
[125]
CurcuminIn vitroB6p210 and B6T315I cells-inhibited proliferation
-induced apoptosis
↓ NF-κB levels
↑ p53 levels
↓ c-Abl levels in cells expressing the wild, but not the mutant, BCR-ABL oncogene
-improved survival in diseased mice and ↓ WBC and GFP cell counts
[126]
In vivoB6 mice
ResveratrolIn vitroGC-resistant CEM-C1-15, Jurkat, Molt-4, and GC-sensitive CEM-C7-14 cells-inhibited the proliferation and induced apoptosis and autophagy in T-ALL cells
-induced cell cycle arrest at G0/G1 phase via up regulating CDK inhibitors p21 and p27 and down-regulating cyclin A and cyclin D1
↓ the expression of antiapoptotic proteins (Mcl-1 and Bcl-2)
↑ the expression of proapoptotic proteins (Bax, Bim, and Bad)
[127]
PterostilbeneIn vitroJurkat and Molt-4 cells↓ cell viability with different extent in two ALL cell lines
-induced apoptosis in lymphoblastic cells
↑ Fas expression both in mRNA and surface levels that results in apoptosis signal transduction improvement, which sensitized cells to apoptosis by immune effector cells
[128]
Gambogic acidIn vitroJurkat and Molt-4 cells-inhibited proliferation, induced apoptosis, and activated autophagy in T-ALL cell lines
-antileukemic effect against peripheral blood lymphocyte cells in patients with ALL
-inhibited phospho-GSK3β S9 protein levels to inactivate Wnt signaling
-suppressed β-catenin protein levels
[112]
Gallic acidIn vitroJurkat cells↓ cell viability[129]
Parthenolide In vitroB- and T-ALL cells-effective against bulk B- and T-ALL cells
-prevented engraftment of multiple LIC populations in NOD/LtSz-scld IL-2Rγc-null mice
-restoration of normal murine hemopoiesis
[130]
In vivoNOD/LtSz-scld IL-2Rγc-null mice
ThymoquinoneIn vitroJurkat cells↓ cell viability of Jurkat cells
-induced apoptosis in Jurkat lymphoblastic cell line
-combination with doxorubicine lead to a synergistic cytotoxicity
[131]
ThymoquinoneIn vitroCEMss cells-induced apoptosis in CEMss cells
↑ in chromatin condensation in the cell nucleus
↑ number of cellular DNA breaks in treated cells
↑ apoptosis with cell death-transducing signals by a down-regulation of Bcl-2 and up-regulation of Bax
↑ generation of cellular ROS, HSP70, and activation of caspases -3 and -8
-the mitochondrial apoptosis was associated with the S phase cell cycle arrest
[132]
Indole-3-carbinol In vitroNALM-6 cells-induced cell-growth inhibition, G1 cell-cycle arrest, and apoptosis in NALM-6 cells
↑ the expression of p53, p21, and Bax proteins
-induced p53 accumulation and expression of pro-apoptotic p53 target genes
↑ PUMA, NOXA, and Apaf-1
-suppressed NF-κB activation and inhibited the protein expression of NF-κB-regulated antiapoptotic (IAP1, Bcl-xL, Bcl-2, XIAP) and proliferative (c-Myc) gene products
-repressed antiapoptotic NF-κB target genes
-potentiated doxorubicin-induced apoptosis through caspase activation and PARP cleavage
-inhibited doxorubicin-induced NF-κB activation in NALM-6 cells
[133]
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; T-ALL—T cell acute lymphoblastic leukemia; SOD—superoxide dismutase; MDA—malondialdehyde; ROS—reactive oxygen species; p-Akt—Akt phosphoration; PcG—polycomb group; HDACs—histone deacetylases; DNMT1—DNA methyl transferase 1; UHRF1—ubiquitin like with PHD and ring finger domains 1; DMF- 5,7-dimethoxyflavone; EGCG—epigallocatechin-3-gallate; B-Pre-ALL—B-precursor ALL; Bax—Bcl-2-associated X protein; Bcl-2—B-cell lymphoma-2; Akt—protein kinase B; cIAP1—cellular inhibitor of apoptosis protein-1; XIAP—X-linked inhibitor of apoptosis protein; NF-κB—nuclear factor-κB; c-Abl—Abelson tyrosine kinase; WBC—white blood cell; CEM—human acute T-lymphoblastic leukemia cell line; T-ALL—T-cell acute lymphoblastic leukemia; GFP—green fluorescent protein; cyclin-dependent kinase (CDK); Mcl-1—myeloid cell leukemia 1; Bad—Bcl-2-associated death promoter; LICs—leukemia initiating cells; Hsp70—70 kilodalton heat shock protein; NF-κB—nuclear factor-κB; Apaf-1—apoptotic protease activating factor 1; Bcl-xL—B-cell lymphoma-extra-large; PARP—poly(ADP-ribose) polymerase.
Table 4. Pharmacological effects of natural compounds in chronic lymphocytic leukemia (CLL).
Table 4. Pharmacological effects of natural compounds in chronic lymphocytic leukemia (CLL).
Bioactive CompoundIn Vitro/In Vivo/Clinical StudyCancer Cell Line and Animal ModelBioactive EffectReferences
Antho 50In vitro -induced apoptosis in B CLL cells
-induced an early caspase-3 activation and UHRF1 down-regulation in B CLL cells independently of the status of tumor suppressor genes p53 and p73
↓ Bcl-2 associated with Bad dephosphorylation
-induced PEG-catalase-sensitive formation of ROS in B CLL cells
[144]
LuteolinIn vitroHG-3 and EHEB cells-↑ the apoptotic cell population in both CLL cells lines by increasing the activities of caspase-3 and -9 and triggering the intrinsic apoptotic pathway [145]
Apigenin In vitroEheb cells-induced apoptosis in human lymphoma B cells in vitro
-prevented the reverted mutations
[146]
EGCGIn vitroCLL B cells
-induced CLL B-cell apoptosis
-suppressed Bcl-2, XIAP, and Mcl-1
-down-regulated the phosphorylation of VEGF-R1 and VEGF-R2
[147]
ChrysinIn vitroCLL and healthy B-lymphocytes↑ cytotoxicity, intracellular ROS, mitochondrial membrane potential collapse, ADP/ATP ratio, caspase-3 activation and apoptosis
-inhibited complex II and ATPases in cancerous mitochondria
-promoted apoptosis in CLL B-lymphocytes by selectively targeting of mitochondria
[148]
ChrysinIn vitroMOLT-4 and JVM-13 cell lines, B-CLL cells derived from 28 patients-induced the activation of proapoptotic Bax
↓ the expression of antiapoptotic Bcl-2 protein
-released cytochrome c from mitochondria into cytosol
-activated caspase-3
-induced apoptosis of peripheral blood lymphocytes isolated from human CLL patients
[90]
ResveratrolIn vitroWSU-CLL and ESKOL cells-inhibited proliferation in leukemic B-cell lines
-induced apoptosis in the two cell lines as well as in B-CLL patients’ cells, as evidenced by the increase in annexin V binding, caspase activation, DNA fragmentation, and decrease of the mitochondrial transmembrane potential
-inhibited in situ NO release in WSU-CLL, ESKOL, and B-CLL patients’ cells
-down-regulation of the two anti-apoptotic proteins iNOS and Bcl-2
[149]
In vitroleukemic lymphocytes from patients with B-CLL
Resveratrol and quercetinIn vitrohuman 232B4 CLL cells↓ proliferation of human 232B4 CLL cells
-induced apoptosis in 232B4 CLL cells through induction of caspase-3 activity
-inhibited cell cycle progression
-arrested cell cycle mainly in G0/G1
[140]
Curcumin Clinical studyTwenty-one patients with stage 0/1 CLL↓ ALC at four patients (20%)
↓ in ALC was accompanied by an ↑ in CD4, CD8, and NK cells
[150]
Curcumin and rapamycin PBMCs from patients with B-CLL-induced apoptosis in B-CLL cells obtained from patients with CLL
↑ caspase-9, -3, and -7 activity
↓ anti-apoptotic Bcl-2 levels,
↑ the pro-apoptotic protein Bax
[151]
Allanxanthone C and macluraxanthoneIn vivoxenograft murine model of human CLL-prolongation of the survival in mice injected with the two xanthones
[152]
PEITCIn vitroPrimary leukemia cells -killed CLL cells with 17p-deletion
-cytotoxic effect against p53-/-leukemia cells from mice in vitro and in vivo
↑ ROS accumulation and GSH depletion in p53-deficient CLL cells
↓ Mcl-1 protein in CLL cells
-induced leukemia cell death in mice
-prolonged the median survival time of the animals
[153]
In vivoTCL1-Tg:p53+ mice
ParthenolideIn vitrocells isolated
from CLL patients
-induced apoptosis in CLL cells
-activated the mitochondrial pathway of apoptosis
-induced a proapoptotic Bax conformational change, release of mitochondrial cytochrome c, and caspase activation
↓ nuclear levels of the antiapoptotic transcription factor NF-κB and diminished phosphorylation of its negative regulator IκB
[154]
ParthenolideIn vitroPBMCs from
B-CLL patients
-displayed potent cytotoxic and apoptotic effects on B-CLL cells in vitro
↓ in the cell viability of B-CLL cells
[155]
Allicin In vitroPBMC cells
CD20+ cells
-induced in vitro apoptosis
-killed the CD20+ tumor B cells via apoptosis
-exhibited tumoricidal effect in vivo
[156]
In vivoBALB/c mice
Indole-3-carbinol In vitroPBMCs cells
hMSC-TERT cells
-induced cytotoxicity in CLL cells but not in normal lymphocytes
↓ XIAP and cIAP1/2 and induced caspase 9-dependent apoptosis of CLL cells
-sinergic activity with fludarabine in CLL cells and overcame stroma-mediated drug-resistance
-mechanism of cell death involved p53-dependent and independent apoptosis
-sinergic activity with F-ara-A in all types of CLL cells and restored F-ara-A sensitivity in fludarabine-resistant CLL cells
[157]
In vivoC57bl/6 mice
Legend: ↑ increased/up-regulated; ↓ decreased/down-regulated; Antho 50—anthocyanin-rich dietary bilberry extract; B-CLL—B-cell chronic lymphocytic leukemia; UHRF1—ubiquitin like with PHD and ring finger domains 1; Bcl-2—B-cell lymphoma-2; Bad—Bcl-2-associated death promoter; PEG-catalase—membrane permeant analog of catalase; ROS—Reactive oxygen species; XIAP—X-linked inhibitor of apoptosis protein; Mcl-1—myeloid cell leukemia-1; VEGF-R1 and VEGF-R2—VEGF membrane receptors; ADP—adenosine diphosphate; ATP—adenosine triphosphate; NO—nitric oxide; iNOS—inducible nitric oxide synthase; ALC—absolute lymphocyte count; PBMCs—peripheral blood mononuclear cells; GSH—reduced glutathione; NF-κB—nuclear factor-κB; hMSC-TERT—human telomerase reverse transcriptase catalytic subunit; cIAP1—cellular inhibitor of apoptosis protein-1.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cotoraci, C.; Ciceu, A.; Sasu, A.; Miutescu, E.; Hermenean, A. The Anti-Leukemic Activity of Natural Compounds. Molecules 2021, 26, 2709. https://doi.org/10.3390/molecules26092709

AMA Style

Cotoraci C, Ciceu A, Sasu A, Miutescu E, Hermenean A. The Anti-Leukemic Activity of Natural Compounds. Molecules. 2021; 26(9):2709. https://doi.org/10.3390/molecules26092709

Chicago/Turabian Style

Cotoraci, Coralia, Alina Ciceu, Alciona Sasu, Eftimie Miutescu, and Anca Hermenean. 2021. "The Anti-Leukemic Activity of Natural Compounds" Molecules 26, no. 9: 2709. https://doi.org/10.3390/molecules26092709

Article Metrics

Back to TopTop