Int J App Pharm, Vol 14, Issue 2, 2022, 10-22Review Article

OVERVIEW OF MITOXANTRONE-A POTENTIAL CANDIDATE FOR TREATMENT OF BREAST CANCER

PREETHI S.1, HITESH KUMAR1, VISHAL B. RAWAL1, RAMKISHAN AJMEER2, VIKAS JAIN1*

1Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India, 2Central Drugs Standard Control Organisation East zone Kolkata- 20
Email: vikasjain@jssuni.edu.in

Received: 29 Oct 2021, Revised and Accepted: 13 Dec 2021


ABSTRACT

Anthraquinones are one of the popular classes of aromatic compounds which possess potential anticancer properties by suppressing the nucleic acid formation and proteins essential to the survival of cancerous cells. Mitoxantrone (MT) is an antibiotic and antineoplastic agent belonging to the anthracycline class of compounds which exhibit minimal incident of drug resistance. It is a synthetic anticancer drug, bound to enzyme topoisomerase IIα inhibitor, and intercalates DNA topoisomerase IIα, preventing re-ligations in DNA strands fragmentation and disruption of DNA repair. The expression of this enzyme was used tumor cells marker because of its key function in cell proliferation. The cleavable complex of topoisomerase IIα is hypothesized to damage the DNA and may enhance apoptosis in tumor cell proliferation. The susceptibility of cells to mitoxantrone is associated with cell topoisomerase II α protein and lowered resistance in breast cancer line cell lines to topoisomerase IIα inhibitors. MT is an ABC-transporter in breast cancer, also designated to be associated with “Breast cancer resistance protein” (BCRP) and it is also a cell cycle non-specific anti-cancer drug and P-glycoprotein substrate. Multiple drug resistance is one of the major drawbacks of this drug which can be avoided by reducing the efflux of the drug from cancer cells by formulating drug by using lipophilic carriers. This manuscript discusses about MT's source, chemistry, physicochemical properties, anti-cancer effects of mitoxantrone and possible pathways, Mitoxantrone targeting topoisomerase II inhibitor for cancer therapy and its mechanism, Various Nano formulation development strategy, toxicity profile, and a few patents related information.

Keywords: Breast cancer, Anthraquinone, Mitoxantrone, Nano formulation, Multiple drug resistance


INTRODUCTION

Cancer is emerging as a major problem globally, both in developed and developing countries. Currently, every year 10 million new cancer cases are diagnosed across the globe [1]. By the end of 2021, cancer rates could rise by 50 % which may account for 15 million new cases and with a projected death fig. of 8.8 million. By 2030, over 21.7 million cancer cases and 13 million cancer deaths are predicted [2–4]. Every year breast cancer affects 2.1 million women and with being frequently reported in the female population [1, 5]). In India, 1.73 million cases were reported in the year 2020, which also contributes to the highest cancer-related mortalities among women [6–8]. Considering high morbidity and mortality associated with breast cancer treatment in the last few decades, cytostatic as well as cytotoxic drugs have been developed [9, 10] the anthraquinone class of antineoplastic agents exhibits better therapeutic efficiency to treat breast cancer compared to that of anthracyclines [11]. Anthraquinone derivatives exhibit antitumor activity by binding to DNA polymerase in tumor cells that results in inhibition of cell growth (cytostatic) or even cell necrosis (cytotoxicity) [12, 13].

Anthraquinone (C14H8O2) is an aromatic organic compound also known as dioxo anthracene or anthracene Dione [14]. Anthraquinones can be obtained from natural as well as synthetic sources; the compounds of this class have a rigid structure consisting of a flat tricyclic aromatic anthracene which contains two keto groups located at the 9th and 10th positions. The tricyclic anthraquinone core gets imgded in the DNA double helix of abnormal cells, which undergo a specific redox reaction that generates superoxide radical anion (O2-) [15]. The bioactive properties of 9,10-anthraquinone (IUPAC: 9,10-dioxoanthracene) are as shown in fig. 1 [16–24]. Anthraquinone compounds have potential anticancer properties as they can inhibit the synthesis of nucleic acid and proteins of cancer cells [25–27].

Fig. 1: Bioactive properties of 9, 10 anthracene dione

Anthraquinone derivatives bind to DNA of cancer cells and inhibit the topoisomerase II enzyme, making them as effective cancer cell growth inhibitors [28]. Binding of these derivatives with topoisomerase-II results in a cleavable complex, which induces the breakage of DNA strands, leading to cell death through apoptosis [29]. Food and Drug Administration (FDA) has approved four naturally and synthetic anthraquinone derivatives for cancer treatment which have the capability to interact with the DNA sequence and result in apoptosis [30]. Drugs derived from anthraquinone derivatives currently in use for cancer therapy are Daunorubicin, Doxorubicin, Mitoxantrone, and Amsacrine [31].

One such anthracenedione derivative is Mitoxantrone (MT), a synthetic anticancer agent, which was originally designed as a simplified analogue of the anthraquinone-containing anthracyclines [32, 33]. MT is an antibiotic with an antineoplastic activity that interferes with the growth and metastasis of cancer cells in the body [34, 35]. MT is a doxorubicin analogue and is generally used in combination with other chemotherapeutic agents to improve its antitumor activity, minimize dose-related side effects [36, 37]. Simultaneously Neidhart et al., In 1980, conducted a prospective comparison of antineoplastic agents for breast cancer, mitoxantrone and doxorubicin as therapy for minimally pretreated patients with breast cancer. So MT is the promising antitumor agent which improves the therapeutic efficacy and it is used in metastatic breast cancer treatment [38–41]. The clinical trial data on metastatic breast cancer treatment, MT monotherapy had shown average positive response in approx. 33 % of patients with no prior chemotherapy exposure [42]. The toxic effects related to MT such as cardiotoxicity and gastrointestinal effect like nausea, vomiting, fatigue were comparatively lower than other anthraquinone derivatives.

MT is used effectively in disease-modifying therapy (DMT), which can also be employed in the therapy of acute non-lymphocytic prostate cancer and leukemia. MT is a novel photosensitizer for photodynamic therapy for breast cancer and reported to cause MCF-7 cell death in vitro [43]. Recent advancements in Nano drug delivery technology and photodynamic therapy have the potential to become an effective alternative to surgery for advanced breast cancer, which reduce the total exposure of the drug to healthy tissues and organs [44, 45]. This review aims to summarize MT as a therapeutic molecule for breast cancer in case of both benign and malignant tumor, including its source, structural modification, physicochemical properties, mechanism, pharmacological action, its multiple drug resistance, pharmacokinetics and metabolism. A comprehensive survey of relevant various Nano formulations is provided, with innovations made in recent years to improve drug resistance and therapeutic effectiveness [46, 47].

Mitoxantrone-source, structural modification, physicochemical properties

MT (1, 4-bis-[[2-(dimethylamino) ethyl-amino]-9,10-anthracenedione) is being developed by the American Cyanamid Company and the Midwest Research Institute as a possible chemotherapeutic agent. Murdock et al. performed structural modifications of MT that included 5,8-dihydroxylation of the anthracenedione nucleus and replacement of both terminal dimethylamino groups with hydroxyethyl functions [48]. The MT compound was primarily developed from ballpoint pen ink ingredient, although it was discovered to have antitumor activity after a routine screening by the National Cancer Institute (NCI) [49–51]. Mitoxantrone (fig. 2) is a hydroxyquinone with amino functionalities attached to aliphatic side groups, but it lacks an amino sugar moiety at the C9 position [50–52]. The basis for the mitoxantrone structure was drew on discovery of ant leukemic agents had a distinct N–O–O triangular pharmacophore (fig. 3) which is previously there in anthracyclines and the amino group is removed, which was considered to have involved in anthracycline cardio toxic [51–53]. The physicochemical properties of MT are given in table 1 [54–60].

Fig. 2: Structure of mitoxantrone

Table 1: Physicochemical properties of mitoxantrone

Characteristics Physical properties
Occurrence The synthetic compound belongs to the class of organic compounds.
Chemical class Anthracenes
IUPAC 1,4-dihydroxy-5,8-bis[2-(2-hydroxyethylamino) ethyl amino] anthracene-9,10-dione and 1,4-dihydroxy-5,8-bis[[2-[(2-hydroxyethyl) amino]ethyl]amino]-9,10anthracenedione dihydrochloride
Molecular formula C22-H28-N4-O6 and C22H28N4O6 •2HCl
Molecular weight Mitoxantrone: 444.5g/mol, Mitoxantrone hydrochloride: 517.41g/mol
Melting point 203-205 °C
Purity >98.0%
Appearance Blue-black solid
Solubility Slightly soluble in methanol; sparingly soluble in water, practically insoluble in acetonitrile, acetone and chloroform.
Stability 15 ° to 25 °C (59 ° to 77 °F) under refrigeration.
Route of administration Intravenous, Intraperitoneal, Continuous and Intermittent infusion.

Fig. 3: Structure of mitoxantrone with N-O-O triangular pharmacophore

Anticancer effects of mitoxantrone and possible pathways

Mitoxantrone exhibits its anticancer activity by acting as an intercalating agent. It inhibits both RNA and DNA synthesis. It is six to seven times more potent than anthracycline derivatives in inhibiting the incorporation of 3H-thymidine and 3H-uridine into DNA and RNA thus disrupting their replication respectively. It also binds to an enzyme called topoisomerase II, which tends to breakage of the DNA strands and stops the synthesis of DNA, resulting in cell death via apoptosis as shown (fig. 4) [61–63]. In addition, mitoxantrone has been reported to exhibit anticancer activity by various mechanisms like autophagy [64–66], paraptosis [67], radiosensitization [68, 69], aberrant cell metabolism [70–72].

Fig. 4: Mechanism of action of mitoxantrone on cytotoxicity activity

There are multiple mechanisms by which MT shows its anticancer activity which is identified by several scientists through their research findings. Lown et al. [73] have demonstrated that, on increasing the concentration of MT progressively to already relaxed PM2-DNA, it shows an intercalative binding with isolated DNA plasmid, MT could also possibly show extensive inter DNA cross-linking with side arms of DNA, making a network of linked molecules. Similarly, Foye et al. [74] discuss that MT binds to isolated DNA at two sites, i.e. (1) Intercalation is seen between base pairs which are successive to each other and (2) Electrostatic interaction with DNA, occurring between amine side chains present in MT and phosphate groups present in DNA. The another study on the basis of in vivo study of interactions between MT and tumor cellular DNA demonstrated that, a) MT does not cause any changes in DNA supercoiling like other classical drugs with intercalating properties. b) It induces protein-related double and single strands DNA fragment and c) MT potentially disrupts single strands of non-protein-based DNA [75].

Mitoxantrone–targeting topoisomerase II inhibitor for cancer therapy

Topoisomerase inhibitors (TI), inhibits cancer cell proliferation by avoiding DNA replication, arousing DNA damage, and thus provoking cell cycle arrest. The DNA topoisomerase enzyme is divided into two types: type I and type II [74]. The DNA topology is altered by Type I topoisomerases by passing a single DNA strand by breaking in the opposing single strand and cleaving it with an active site that is tyrosine residue, producing a phosphodiester link with the enzyme [76]. Type II A topoisomerases, which include eukaryotic topoisomerase II and II, have a three-domain structure that spans the A and B subunits that form the homodimer (or heterotetramer in prokaryotes) as shown in (fig. 5). The N-terminal domain consists an ATP-binding region (ATPase domain), a core domain containing a TOPRIM fold and a DNA-binding region, and a C-terminal domain of unknown function [76, 77]. Eukaryotic TOP IIA has three sections known as the N-gate, DNA gate, C-gate, and the catalytic Tyr805, which is responsible for cleavage present in the DNA-gate. Topoisomerase II alters the structure of DNA strands by breaking and reconnecting the phosphodiester backbone of DNA using identical active site tyrosine residues [78]. This is accomplished through the production of temporary, enzyme-bridged double-strand breaks. This enzyme is commonly utilized as a marker of cancer cells due to its function in cell proliferation [77]. TOP II plays a critical part throughout the replication of DNA and its main activities are chromosomal segregation. Topoisomerase I, IIα, and IIβ are the targets for commercialized anti-cancer agents [78]. Topoisomerase IIα (Topo IIα) is the target of multiple chemotherapeutics such as anthracyclines and other intercalators, such as mitoxantrones [79]. It plays a vital role in DNA replication and has been associated in breast cancer with the proliferation of cellular and HER2/new protein overexpression [80]. Mitoxantrone may act as the topoisomerase IIα catalytic cycle. But, most likely by interfering with the activities of the enzyme, all of it stimulates the creation of protein-linked DNA strand breaks[81]. The cleavable complex is hypothesized to damage the DNA, to cause toxicity, and causes apoptosis in tumor cells. The sensitivity of cells to mitoxantrone depends on cell topoisomerase II α protein and lowered topoisomerase II resistance in breast cancer line cell lines to topoisomerase IIα inhibitors [82].

Fig. V: Structure of topoisomerase IIα

(Topoisomerase IIα enzymes with bound DNA and Two sub-family kind domains (The DNA ribbon is dark blue; the chain A of topoisomerase IIα is red; the chain B is light blue)) [83].

Mechanism of topoisomerase II inhibitor for cancer therapy

Prevention of the enzyme-DNA topoisomerase takes place by some of the generally accepted mechanisms at molecular level that are mentioned below [84, 85].

Substrate competitive inhibition

To the topoisomerase active site, an inhibitor molecule is attached which blocks the DNA substrate from binding. There are no known inhibitors that are topoisomerase-specific which function in this way, however present research on DNA-competitive inhibitors of other DNA-binding proteins imply that this mechanism might potentially function in DNA topoisomerases II [86].

Topoisomerase poisons mechanism

The DNA re-ligation is blocked by the protein-DNA-drug complex which is ternary. It prevents the enzyme from forming a "cleavage complex." This drug inhibits enzyme turnover and accumulation of cytotoxic cleavage complex within the cell in excessive quantities [87].

Potent inhibition of the ATP

The ATP binding site has one competitive inhibition of the type II topoisomerases that prevent enzymatic activation by ATP-hydrolysis. Type II topoisomerase is necessary for ATP synthesis as well as for the domain which binds ATP is distinct from the DNA binding domain. Further, the dependency or absence of DNA topoisomerases is distinguishable from the Mg 2+for catalytic activity. Mg2+ appears to be required for Type I A and Type II topoisomerases, however, catalytic action is required for Type IB topoisomerases if Mg 2+is not present. [87].

The stabilization of the cleavage complex

It is the topoisomerase II poisons mechanism which encompasses the production of a ternary complex that is locked of cleaved DNA, protein, and antineoplastic medication, which grows and produces cytotoxicity [85]. Anthracyclines, such as mitoxantrones, are anthracyclines that target type II A topoisomerases in the treatment of breast cancer. These agents are bound to cleaved DNA/protein complexes, blocking DNA binding and seal the enzyme to the cleavage complex [88]. This cleavage complex develops and causes a breakdown of the DNA strand and finally cell death. The most commonly known topoisomerase poisons work is the interplay between the-1 and+1 DNA base pairs of the protein-DNA cleavage complex. Additional hydrophobic and electrostatic interactions with both DNA and protein increase the poison's binding and prevent DNA from being relegated to topoisomerase II [89, 90].

The redox-dependent

The covalent and redox-dependent creation of a complex of drug-enzyme and the complex of DNA cleavage, which results in a same build-up and eventual cell death, is the mechanism of the second topoisomerase poison. The increased interfacial topoisomerase poison in the active site between two DNA base pairs is caused by the interactions of the DNA bases with the connected, four-ring pharmacological system. Hydrogen bonding to surrounding protein residues and local DNA bases has been discovered in the surrounding ring systems of antineoplastic medications [89, 91].

Catalytic inhibition

The third ideal topoisomerase inhibitory mechanism encompasses the competitive binding of small molecules to the ATP binding site present in the N-terminal region of type II topoisomerases by catalytic inhibition [85, 92]. The torsional strain of the supercoiled DNA provides the energy required for the activity of type I topoisomerases, whereas ATP hydrolysis provides the energy required for the action of type II topoisomerases [84]. The passage of the DNA T segment is blocked by the competitive hydrolysis of ATP through smaller molecules from passing through the G-Gate to the C-terminal domain, which leads to catalytic topoisomerase inhibition, DNA transcript halting as well as cells apoptosis. This mechanism is not responsible for the DNA and cell damage induced by topoisomerase poisons. Despite the fact that no anticancer drugs with this mechanism of inhibition are currently on the market [93].

Nano formulation development strategy of mitoxantrone

With its beneficial safety profile, MT has recently gained more attention as the therapeutic compound in the treatment of cancer therapy [84]. Even though, MT is classified as a Class III drug according to the Biopharmaceutical Drug Deposition and Classification System (BDDCS), owing to its low permeability, low metabolism and bioavailability. MT has limited therapeutic responses because of its drug resistance in tumor cells which is a most concern in malignancy [96, 97]. To address the low permeability of the drug across the cell membrane, incorporation of the hydrophilic drug (MT) into lipophilic carrier system, like liposomes, polymeric mixed micelles, nanoparticles such as NLCs, SLNs, Nano-diamond NPs, gold NPs, Albumin NPs, graphene oxide NPs, iron NPs etc. have been extensively reported [98–101].

The incorporation of MT in lipophilic carrier system is employed to overcome many challenges such as modulation sustained drug release, reverse multidrug resistance (MDR), and improve its bioavailability [102, 103]. Several research studies shows that nanotechnology imparts stable formulation of MT, improve its bioavailability and therapeutic efficacy [104–107]. Nano drug delivery methods are advanced techniques for delivering drugs to tumor cells with minimal drug leakage to healthy cells [108, 109]. The development of MT’s Nano formulations can resolve the drug resistance in cancer cells and minimize drug efflux and enhance the retention of MT in malignant cells [33, 110]. Lu et al. have developed the solid lipid nanoparticles (SLNs) of MT, which improved the drug resistance in breast cancer and its lymph node metastases in mice. These MT-loaded SLNs depicted sustained release of the drug which have effectively inhibited the breast cancer and lymph node cancer in nude mice model with no toxicity in normal tissues and have reduced the lymph node cancer size up to 1.85±27.42 mm3 compared to MT alone (119.32±57.30 mm3) [111].

To improve the therapeutic targeting of MT, Though et al. developed Nanodiamonds (NDs) for the promising delivery of MT. NDs with diameters of about 5 nm and demonstrated their ability to enhance drug resistance while slowing tumor growth development, they evaluated the MT Nanodiamond complex, on the MDA-MB-231-Luc-D3H2LN in TNBC cell line that was virally transduced for resistance to MT, the comprehensive complex enhanced drug retention and efficacy. The results of the in vitro analysis suggested that MT Nano diamond could be a better drug delivery system for drug-resistant cancers [121]. Furthermore, ling et al. synthesized carrageenan hybrid nanostructured lipid carriers (NLCs) to improve the sustained drug release, bioavailability and anticancer efficacy for MT through the oral route. Results of the study depicted that the oral bioavailability of drug-loaded Nanocarriers was increased 3.5 times than free drug. The cytotoxicity investigation depicted that the MT Nanocarriers significantly enhances the anticancer efficacy compared to pure MT against MCF-7/MX cells and diminished the BCRP associated drug resistance [122].

Likewise, MT was utilized to reduce the MDR effect with photosensitive properties to improve anticancer efficacy. The MT was mixed with poly(ε-caprolactone)-pluronic F68 to prepare micelles. When it was subjected to close-infrared light and induced irritation, MDR's influence on MCF-7/ADR cells was reversed by photochemical interactions. This resulted in cytotoxicity of cancer cells. Usually, MT causes apoptosis in MCF-7/ADR cells by producing ROS and decreasing P-glycoprotein activity. These mixed micelles effectively reversed the MDR effect via photodynamic therapy [112]. MT possesses broad range of therapeutic effects to against advanced breast cancer [123]. It is found to be highly effective in ovarian cancer [124], colon cancer, non-Hodgkin's lymphoma, acute myeloid leukemia [125–127], bladder cancer [128], prostate cancer [129], and glioblastomas [113, 130]. The various Nano formulations loaded with MT for effective treatment in breast cancer therapy and various cancer treatment are compiled in (table 2).

Table II: Formulation development and strategies based on nanotechnology of mitoxantrone

Nano formulation Method of preparation Major component of the delivery system Purpose Key findings/conclusion

Multifunctional lipid-sodium

glycocholate Nano carriers

Emulsification ultra-sonication method Compritol 888, Cremophor RH40, Miglyol 812, lecithin Combination therapy by combining the "BCRP bypassing effect." Co-encapsulation of the hydrophilic anticancer drug, BCRP inhibitor, into TMLGNs is an effective platform for MDR reversal [39].

Hydrophobically Modified Pullulan

Nanoparticles

Nano Precipitation method Pullulan, cholesterol Prevent the growth of cancer cells in the bladder and the migration of MB49 cells with nanoparticles The release of Cholesterol-substituted pullulan polymer NPs is proportional to acidity. Also, larger NPs have shown better inhibition of cancer cells in the bladder due to migration than smaller NP [49].
Novel nanostructured lipid-dextran sulfate hybrid carriers (NLDCs)

Emulsification-ultra sonication

Method.

Lipid-based drug delivery system. Delivers water-soluble cytotoxic drug in cancer chemotherapy for MDR. NLDCs of small size show high efficacy of drug encapsulation, long-term release characteristics, desired pharmacokinetics, and cytotoxicity [98].
MT-PFP/PPP mixed micelles Solvent Evaporation method.

Poly(ε-caprolactone), poly (d, l-lactide-

co-glycoside), poly (ethylene glycol) pluronic F68.

To examine the photosensitizing properties of MT clinically used as a PDT. MT-PFP/PPP micelles were able to decrease P-gp activity, increase ROS levels and cell uptake, which reversed the MDR effect after irradiation and triggered cell apoptosis [112].

Plant virus-based

Nanoparticles

Ultracentrifugation method. Plant virus-based nanoparticles drug delivery system. Delivery of MT prevents poor penetration of the blood-brain barrier Uptake of CPMV–MT in U87-MG glioblastoma cells and this encapsulated MT maintain its therapeutic potential [113].
Solid lipid nanoparticles Film dispersion–ultrasonication Lecithin and Compritol®888 A novel approach to active delivery of antitumor drugs against breast cancer and lymph node metastases, with a therapeutic effect that is both inspiring and low in side effects. P388 cell lymph node tumor model was effectively developed, and the suppression of MTO-SLN against the metastases was promising. The MTO-SLN was promising in the per cent inhibition of the tumor growth [114].
Nano diamond - Nano diamonds To improve drug tolerance Improved efficacy and drug retention in an MDA-MB-231-Luc-D3H2LN [115].
Nanostructured lipid-carrageenan hybrid carriers Emulsification-ultra sonication Compritol 888 ATO, miglyol 812, cremophor RH40 and lecithin To enhance oral bioavailability, encapsulation efficiency, and reduce cytotoxicity  Enhance the encapsulation of breast cancer cells, increased oral bioavailability, and anti-tumor activity [116].
Folate-conjugated Albumin Nanoparticles Chemical cross-linking with glutaraldehyde (Coacervation method) Folate, Bovine serum albumin BSANP targets to SKOV3 cells and enhance therapeutic potential for cancer chemotherapy MT-BSANP-folate NPs increased the intracellular uptake of trapped MT in SKOV3 cell to enhance anticancer activity by passive accumulation [117].

Hyaluronic acid/

polyethylene glycol nanoparticles

Nano precipitation and lyophilisation

Hyaluronic acid,

polyethylene glycol

To exhibit significant

Cytotoxic effects on CD44-positive cell line.

NPs attached effectively to the receptor binding site demonstrate considerable cytotoxic effects in CD44-positive cell lines on cell viability [118].
Phospholipid-amorphous calcium carbonate hybrid nanoparticles Facile solvent-diffusion method Ammonium carbonate, anhydrous calcium, Chloride, PL (S100), DSPE-PEG2000, DSPE-PEG-FA Enforce the delivery of active agents within cancer cells with increased drug penetration NPs with enhanced performance, site targeting, controlled drug release, and increased drug penetration [119].

PEGylated Gold

Nano complexes

Chemical reduction Gold chloride trihydrate, Methoxy polyethylene glycol thiol In vivo via passive targeting for cancer therapy, enhanced retention and permeability effect. AuNPs-PEGs-MT enhanced stability, loading efficiency of Mitoxantrone (1.9-fold), and cytotoxicity [120].

Metabolism and its metabolites

The liver is the primary site of MT metabolism [131]. The metabolism of MT involves the microsomal pathway and/or peroxidase enzymes, including neutrophil myeloperoxidase are involved [132]. Its metabolites have been discovered in the bone marrow, kidney, heart, spleen, and lungs, in addition to the liver [127]. Inhibition of the functioning of cytochrome P450 combined oxidase function in a human hepatoma-derived cell line reduced the inhibitory effect of MT on cell proliferation. A rat hepatocyte model and human breast cancer cells both produced similar results [128–131]. Furthermore, phase II metabolism, namely conjugation with glucuronic acid and reduced glutathione (GSH), plays an important part in the MT detoxification process[133, 134]. Numerous pre-clinical studies have been carried out in rats, rabbits and anaesthetized pigs to identify different metabolites. Smyth et al. reported the oxidative metabolism of terminal hydroxyl groups of parent molecule (MT), causing the formation of the metabolites A and B. They have identified metabolites of mitoxantrone in plasma and urine. The metabolism in humans and animals indicated that MT might conjugate with glucuronic acid and glutathione [135]. Richard et al. separated several metabolites of MT in rabbits were identified as the mono and dicarboxylic acid derivatives [136].

They also evaluated the excretion of mitoxantrone in bile and urine. Mitoxantrone is mostly eliminated by the bile route, with minor levels excreted through the urine. The metabolite naphtoquinoxaline brings cellular damage to newborn cardiomyocytes that are isolated from rats [135]. This metabolite has already been discovered to be an in vivo MT product of biotransformation in humans, rats and pigs. When examining the variability among interspecies, the metabolic distinction allying humans and rats is that the mono as well as dicarboxylic acid derivatives of MT were substantial bioproducts from human metabolism, whereas they are negligible in rats [136]. Except for the results reported by Shipp et al. in that no studies have been conducted that relate MT bioproducts to its most extreme undesirable impact i.e. late irreversible cardiotoxicity [137–139].

Pharmacokinetics

According to Biopharmaceutical Drug Disposition and Classification System, MT is a class III drug that is poorly absorbed when given orally [96, 140]. The pharmacokinetics properties of MT are being studied in cancer patients as well as in animals through different routes of administration (intravenous, intrapleural, intraperitoneal, and intra-arterial) and were examined by HPLC and total radioactivity method [141]. After intravenous injection, MT has a rapid dispersion followed by a longer clearance process distinguished by extensive accumulation in highly perfused organs, according to pharmacokinetic experiments in humans and laboratory animals. Intravenous administration of MT follows a three-phase method for elimination from the blood plasma, a fast initial (α) distribution phase with a half-life (0.1 h), an intermediate (β) half-life distribution phase (1.1 h), and a terminal gamma elimination phase (γ) (42.6 h.) with a half-life of 12 d [75, 95]. After five days of the urinary sample collection, Albert et al., in their study, stated that about the whole MT dose was retrieved in an unaltered form at a rate of 6.5 percent (range: 5.2-7.9 %) [75]. Despite the persistence of MT in the tissues, the pharmacokinetics of the drug did not appear to be affected by repeated daily administration for five days or at 3-week intervals for up to 12 courses, with no significant changes in the terminal half-life, urinary excretion, and volume of distribution [75, 142]. A steep dose-response curve, dose-limiting myelosuppression, and extensive tissue binding have been reported by systemic administration of MT. However, since MT is non-vesicant, regional drug administration can effectively solve these problems. Mitoxantrone is often delivered into a deep tissue compartment from which it is slowly retrieved, as illustrated by its prolonged plasma terminal phase half-life, extraordinarily large volume of distribution (Vd), and a relatively substantial quantity of mitoxantrone (>15 % of the administered dose) was found in necropsy tissues 35 d after dosing [82, 117]. These outcomes back up a pharmacologic justification for using mitoxantrone in an irregular dosage regimen.

Multiple drug resistance of mitoxantrone

Drug resistance tumor cells are considered to be a major concern of cancer chemotherapy. The principal aspect of clinical failure and death in cancer patients is due to resistance to chemotherapy. Patients who acquire resistance to cytotoxic treatment often develop resistance to several ant leukemic drugs, leading to multidrug resistance (MDR) phenotype in cancerous tissue. MT is a cell cycle non-specific anti-cancer drug and P-glycoprotein substrate used in the treatment of breast cancer [143]. Mitoxantrone transporter is identified as ABC-transporter in a breast cancer-derived cell line and is designated as “breast cancer resistance protein” (BCRP) and also as “mitoxantrone resistance-associated protein [144, 145]. The resistance occurs by possible mechanisms like decreased accumulation of intracellular drugs, often with over-expression of P-glycoprotein (MDR1) mediating increased drug efflux, enzymatic modifications that minimize susceptibility to DNA damage or improve DNA repair, altered drug metabolism, distribution, and binding site [146–149]. The inhibition of downstream death signaling pathways are all forms of drug resistance mechanisms generally observed with mitoxantrone [150–152]. In clinical studies, it is observed that efflux transport is carried out by ATP-binding cassette (ABC) transporters, which transport substrate drugs from the cell in an energy-dependent manner against a concentration gradient [150]. P glycoproteins (P-gp) and multidrug resistance proteins (MRPs) are two transmembrane xenobiotic transporter proteins that imparts a significant role in clinical drug resistance in drug-sensitive human breast cancer cells [152–156]. BCRP overexpresses several chemotherapeutic drugs, including mitoxantrone, adriamycin, and doxorubicin, out of the cell to several cancer cell lines [157–159].

Table 3: Toxicity of mitoxantrone

S. No. Toxicities Treatment Dose Drug delivery system Main findings
1 Cardiovascular toxicity, Congestive heart failure and AML. Advanced breast cancer

12 mg/m2every

3 mo

(140 mg/m2*)

MT intravenous infusion Cardiac disease has been found in cancer patients who received cumulative dosages either alone or in conjunction with other cytotoxic agents. Elevated risk of Leukaemia in 0.25% of patients (n=802) has been observed [168].
2 Hematological Effects ANLL 12 mg/m2for 5 d or 14 mg/m2for 3 d MT intravenous infusion

Granulocyte recovery times for refractory ANLL have been recorded to be 26 to 32 d, but this refers to a count of at least 1000/l.

MT is efficacious in previously untreated ANLL in conjunction with cytosine arabinoside. CR after 240 d of induction obtained 89% of patients treated with mitoxantrone [170].

3 Neutropenia Prostate cancer 2–5 mg/m2 MT intravenous infusion The maximum dose of MT that could be tolerated was 4 mg/m2. Patients receiving 2–4 mg/m2 had no dose-limiting toxicities, while those receiving 5 mg/m2 had none [171].
4 Gastrointestinal toxicity. Breast cancer 12 mg/m2 MT intravenous infusion 43% (n=100) of patients treated had nausea, vomiting, or both, but these effects were severe in less than 1% of patients [107,172].
5 Hematologic toxicities and Non-hematologic toxicities. Malignant lymphoma and Advanced solid tumor. 6-18 mg/m2

PEGylated liposomal mitoxantrone-

loaded into small unilamellar vesicles 60 nm plm60-s

Extreme leukopenia was found in only one patient (16 mg/m2). Some hematological toxicity symptoms include thrombocytopenia, erythropenia, and a drop-in hemoglobin level.
Dyspnea, nausea, skin rash, vomit, pruritus, and an increase in ALT were among the non-hematologic toxicities, but they were all treated [173].
6 Alopecia Breast cancer 12 mg/m2 MT intravenous infusion Dyspnea, nausea, skin rash, vomit, pruritus, and an increase in ALT were among the non-hematologic side effects, but they were all treated.
No clinical cardiotoxicity was seen with dosages of 24-144 mg/m2(mean 78 mg/m2) [174].

*= maximum cumulative lifetime dose; *1= MT in combination with methylprednisolone; MS: Multiple sclerosis; LVEF: Left ventricular ejection fraction; n=Number of patients; AML: Acute myelogenous leukemia; ANLL: Acute Non-Lymphoblastic Leukemia; CR: complete remission ALT: Alanine aminotransferase.

Sri K et al. investigated in key role of multidrug resistance protein (MRP1) and ATP binding cassette Subfamily C Member 1 (ABCC1) in MT cross-resistance in the MCF7 cell line. The MCF7/VP resistant cell line exhibits elevated levels of MRP1 compared to the MCF7/WT parental cell line. MCF7/VP cells are 6–10 times relatively more resistant to MT than MCF7/WT cells. MT efflux is ATP-dependent and inhibited by cyclosporine A and sulfinpyrazone. With these agents, inhibition of MT efflux sensitizes cells to MT cytotoxicity and partially reverses MT resistance in MCF7/VP cells. It concluded that overexpression of MRP1 in MCF7/VP cells is expected to be enhance the MT efflux and resistance [160]

Potent inhibition of BCRP to minimize drug efflux is a promising approach to overcome drug resistance limitations [161–163]. However, because MT is a substrate of the BCRP efflux transporter, tumor cells are extremely resistant to it [156]. To improve MT efficacy, Nano carriers drug delivery that can minimize efflux, prolong drug retention in drug-resistant cancer cells, and induce complexity in tissues can be developed.

Toxicity of mitoxantrone

MT is a synthetic derivative with an antitumor activity similar to that of anthracyclines but with less toxicity [164]. The toxicity profile of MT is associated with the total dose administered. It is usually well-tolerated at standard doses [165]. Anaemia, cardiotoxicity, neutropenia, and liver toxicity were some of the most commonly reported unexpected side effects in Phase II/III clinical trials [166, 167]. Liver injury linked to MT is possibly due to hypersensitivity reaction [168]. The published data also indicate many other toxic effects (table 3). Most of these side effects were intermediate or mild, such as nausea, abdominal pain, vomiting, fever, or bone marrow suppression. The major long-term toxicity is dose-dependent and is a strict limiting factor for the duration of treatment [169]. To reduce the risk of cardiac events, the drug should be administered slowly and carefully through an intravenous route (over 30 min, as it may cause severe local tissue damage) and administration of MT has shown good tolerance at an acceptable level [109]. Analogues of MT with much lower cardiotoxicity are currently being investigated in experimental animal models [58, 138].

Patents on mitoxantrone and related formulations

The records were found using a variety of databases, including Google Patents, Espacenet, WIPO, and the USPTO search engines. Terminologies like mitoxantrone, mitoxantrone formulations for cancer, use of mitoxantrone in breast cancer, etc., were used to perform a search in different databases. Considering patents written in English, we concentrated on the relevant material, title, abstract, and study status. There are some patents on the MT-based drug delivery system that were considered for this review and are listed in (table 4).

Table 4: Patents on mitoxantrone based drug delivery systems

Nano-carrier system

Therapeutic

agent

Therapeutic indication Patent application number Patent proprietor Outcome
Liposome Mitoxantrone Non-Hodgkin's lymphoma, myeloma, advanced breast cancer, bladder, ovarian and hepatocellular carcinomas US5858397A·1999 Univ British Columbia [CA]

Liposomal formulations of mitoxantrone

[175]

Sustained-release implant Mitoxantrone Entity tumor/Solid tumors CN101176710A·2008 Jinan Shuihua Medical Science [CN] Mitoxantrone sustained-release agent for curing tumors [176]
Nanoparticles Mitoxantrone Prolongs the survival time of S180 mice, improves the efficacy, and reduces systemic toxicity CN107684627A·2018 Univ Capital Medical Sciences Mesoporous silicon dioxide-methotrexate-mitoxantrone nanoparticles [177]
Liposome Mitoxantrone High entrapment rate, strong stability and short half-life CN103622909A·2014 Univ Jilin Cardiolipin-liposome preparation and its application in antitumor drugs [178]
TRACER Mitoxantrone Pharmaceutical preparations and application of MT as a lymph tracer. CN102397561A·2012 Univ Shenyang Pharmaceutical Application of mitoxantrone as lymph tracer [179]

CONCLUSION

MT is an antibiotic with an antineoplastic activity that interferes with the growth and spread of cancer cells in the body. In spite of being potential anticancer drug, Mitoxantrone’s utility is limited due to low permeability, low metabolism, bioavailability, drug resistance, cardiotoxicity, and gastrointestinal disorders. Mitoxantrone targeting topoisomerase II inhibitor for cancer therapy. To address these drawbacks, MT is now being formulated as Nanocarrier systems to enhance permeability, bioavailability, efficient tumor targeting, controlled drug release and drug resistance. The reviewed literature in this article emphasis on the recent advances of MT-containing Nano-drug delivery systems, such as Nano-diamond, PEGylated Gold Nano-complexes, lipid-based nanoparticles such as SLNs, NLCCs, micelles, and other lipid-based nanoparticles, transdermal cubic phases, and photodynamic therapy, as a better option for increasing the effectiveness of DDS in tumor treatment. However, till date, these Nano formulations containing MT are used only in in vitro and in vivo cell line studies.

FUTURE DIRECTION

In the future, Nanotechnology-based approaches such as SEDDS, Quantum dots, Carbon nanotubes, can be used as a promising therapeutic solution for targeting drug resistance and its low permeability. MT appears to be metabolized in the liver and hence further studies must be carried out to determine the effect of liver dysfunction on the disposition and toxicity of MT. Hence it is concluded that this literature serves as a valuable resource for a comprehensive review of MT as the potential target moiety for developing Nano strategies for clinical use. To overcome the drug resistance and toxicity issues which are currently research progress needs to be focused and improved on nanotechnology-based approaches.

ABBREVIATIONS

BC: Breast cancer, TNBC: Triple-negative breast cancer, MT: Mitoxantrone, DNA: Deoxyribonucleic acid, RNA: Ribonucleic acid, ABC transporter: ATP-binding cassette transporter, BCRP: Breast cancer resistance protein, FDA: Food and Drug Administration, DMT: Disease-modifying therapy, TOP2A: Topoisomerase inhibitor type II, NCI: National Cancer Institute, PM2: Plaque morphology mutant, BDDCS: Biopharmaceutical Drug Deposition and Classification System, NPs: Nanoparticles, NLCs: Nano structured lipid carriers, SLNs: Solid lipid nanoparticles, MDR: Multidrug resistance, NDs: Nano diamonds, BSANPs: Bovine serum albumin nano particles, PFP: poly(ε-caprolactone)–pluronic F68–poly(ε-caprolactone), PLGA: Poly D, L-lactic-co-glycolic acid, PEG: Polyethylene glycol, NLCCs: Nanostructured lipid-carrageenan hybrid carriers, NLDCs: Novel nanostructured lipid-dextran sulfate hybrid carriers, PL/ACC: Phospholipid/amorphous calcium carbonate, CHP: Cholesterol-substituted pullulan polymers, TMLGNs: Three-in-one multifunctional lipid sodium glycocholate nanoparticles, PDT: Photodynamic therapy, HPLC: High performance liquid chromatography, Vd: volume of distribution, MRP: Multidrug resistance protein, MS: Multiple sclerosis, LVEF: Left ventricular ejection fraction, AML: Acute myelogenous leukemia, ANLL: Acute Non-lymphoblastic Leukemia, CR: complete remission, ALT: Alanine aminotransferase, SEDDS: Self-emulsifying drug delivery systems.

ACKNOWLEDGMENT

All the authors have contributed equally for the work done.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All authors have contributed equally.

CONFLICT OF INTERESTS

The authors declare that they have no conflict of interest.

REFERENCES

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7-30. doi: 10.3322/caac.21590, PMID 31912902.

  2. Eaton L. World cancer rates set to double by 2020. Br Med J. 2003;326(7392):728. doi: 10.1136/bmj.326.7392.728/a, PMID 12676827.

  3. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015 Mar 1;136(5):E359-86. doi: 10.1002/ijc.29210, PMID 25220842.

  4. Dobson JM, Hohenhaus AE, Peaston AE. Cancer chemotherapy. In: Small animal clinical pharmacology. Second. 1st ed. Elsevier; 2008. p. 330-66.

  5. Kumar A. Comprehensive review on etiopathogenesis, treatment and emerging therapies of breast cancer. Asian J Pharm Clin Res. 2021;14(8):20-33. doi: 10.22159/ajpcr.2021.v14i8.41974.

  6. Mathew A, George PS, Arjunan A, Augustine P, Kalavathy MC, Padmakumari G, Mathew BS. Temporal trends and future prediction of breast cancer incidence across age groups in Trivandrum, South India. Asian Pac J Cancer Prev. 2016;17(6):2895-9. PMID 27356709.

  7. Labrèche F, Goldberg MS, Hashim D, Weiderpass E. Breast cancer. Occup Cancers. 2020. p. 417-38.

  8. Cancer control opportunities in low- and middle-income countries [internet]. Washington, DC: National Academies Press; 2007.

  9. Schirrmacher V. From chemotherapy to biological therapy: a review of novel concepts to reduce the side effects of systemic cancer treatment. Int J Oncol. 2019;54(2):407-19. doi: 10.3892/ijo.2018.4661, PMID 30570109.

  10. Kesharwani SS, Mallya P, Kumar VA, Jain V, Sharma S, Dey S. Nobiletin as a molecule for formulation development: an overview of advanced formulation and nanotechnology-based strategies of nobiletin. AAPS PharmSciTech. 2020 Aug 5;21(6):226. doi: 10.1208/s12249-020-01767-0, PMID 32761293.

  11. Kharasch ED, Wendel NK, Novak RF. Anthracenedione antineoplastic agent effects on drug metabolism in vitro and in vivo: relationship between structure and mechanism of inhibition. Fundam Appl Toxicol. 1987;9(1):18-25. doi: 10.1016/0272-0590(87)90149-7, PMID 3114031.

  12. Al-Otaibi JS, Teesdale Spittle P, El Gogary TM. Interaction of anthraquinone anti-cancer drugs with DNA: experimental and computational quantum chemical study. J Mol Struct. 2017;1127:751-60. doi: 10.1016/j.molstruc.2016.08.007.

  13. Kreft D, Wang Y, Rattay M, Toensing K, Anselmetti D. Binding mechanism of anti-cancer chemotherapeutic drug mitoxantrone to DNA characterized by magnetic tweezers. J Nanobiotechnology. 2018;16(1):56. doi: 10.1186/s12951-018-0381-y, PMID 30005668.

  14. Singh RS, SM, Chauhan SM. 9,10-Anthraquinones and other biologically active compounds from the genus Rubia. Chem Biodivers. 2004;1(9):1241-64. doi: 10.1002/cbdv.200490088, PMID 17191903.

  15. Tian W, Wang C, Li D, Hou H. Novel anthraquinone compounds as anticancer agents and their potential mechanism. Future Med Chem. 2020;12(7):627-44. doi: 10.4155/fmc-2019-0322, PMID 32175770.

  16. Chien SC, Wu YC, Chen ZW, Yang WC. Naturally occurring anthraquinones: chemistry and therapeutic potential in autoimmune diabetes. Evid Based Complement Alternat Med. 2015;2015:357357. doi: 10.1155/2015/357357, PMID 25866536.

  17. Huang Q, Lu G, Shen HM, Chung MC, Ong CN. Anti‐cancer properties of anthraquinones from rhubarb. Med Res Rev. 2007;27(5):609-30. doi: 10.1002/med.20094, PMID 17022020.

  18. Chien SC, Wu YC, Chen ZW, Yang WC. Naturally occurring anthraquinones: chemistry and therapeutic potential in autoimmune diabetes. Evid Based Complement Alternat Med. 2015;2015:357357. doi: 10.1155/2015/357357, PMID 25866536.

  19. Winter RW, Cornell KA, Johnson LL, Ignatushchenko M, Hinrichs DJ, Riscoe MK. Potentiation of the antimalarial agent rufigallol. Antimicrob Agents Chemother. 1996;40(6):1408-11. doi: 10.1128/AAC.40.6.1408, PMID 8726010.

  20. Fosso MY, Chan KY, Gregory R, Chang CW. Library synthesis and antibacterial investigation of cationic anthraquinone analogs. ACS Comb Sci. 2012;14(3):231-5. doi: 10.1021/co2002075, PMID 22324350.

  21. Friedman M, Xu A, Lee R, Nguyen DN, Phan TA, Hamada SM, Panchel R, Tam CC, Kim JH, Cheng LW, Land KM. The inhibitory activity of anthraquinones against pathogenic protozoa, bacteria, and fungi and the relationship to structure. Molecules. 2020 Jul 7;25(13):3101. doi: 10.3390/molecules25133101, PMID 32646028.

  22. Kshirsagar AD, Panchal PV, Harle UN, Nanda RK, Shaikh HM. Anti-inflammatory and antiarthritic activity of anthraquinone derivatives in rodents. Int J Inflam. 2014;2014:690596. doi: 10.1155/2014/690596, PMID 25610704.

  23. Mellado M, Madrid A, PenA-CorteS H, LoPez R, Jara C, Espinoza L. Antioxidant activity of anthraquinones isolated from leaves of muehlenbeckia hastulata (JE SM.) Johnst. (polygonaceae). (polygonaceae). J Chil Chem Soc. 2013;58(2):1767-70. doi: 10.4067/S0717-97072013000200028.

  24. Huang Q, Lu G, Shen HM, Chung MCM, Ong CN. Anti-cancer properties of anthraquinones from rhubarb. Med Res Rev. 2007 Sep;27(5):609-30. doi: 10.1002/med.20094, PMID 17022020.

  25. Siddamurthi S, Gutti G, Jana S, Kumar A, Singh SK. Anthraquinone: A promising scaffold for the discovery and development of therapeutic agents in cancer therapy. Future Med Chem. 2020;12(11):1037-69. doi: 10.4155/fmc-2019-0198, PMID 32349522.

  26. Ritter JK, Chen F, Sheen YY, Tran HM, Kimura S, Yeatman MT, Owens IS. A novel complex locus UGT1 encodes human bilirubin, phenol, and other UDP-glucuronosyltransferase isozymes with identical carboxyl termini. J Biol Chem. 1992;267(5):3257-61. doi: 10.1016/S0021-9258(19)50724-4, PMID 1339448.

  27. Colbow K, Dunyluk RP. Energy transfer in photosynthesis. Int J Quantum Chem. 2009 Jun 18;10(S3);Suppl 3:151-9. doi: 10.1002/qua.560100718. PMID 4751233

  28. El-Gogary TM, El-Gendy EM. Noncovalent attachment of psoralen derivatives with DNA: Hartree–Fock and density functional studies on the probes. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy. 2003;59(11):2635-44. doi: 10.1016/S1386-1425(03)00038-6.

  29. Hande KR. Topoisomerase II inhibitors. Update on Cancer Therapeutics. 2008;3(1):13-26. doi: 10.1016/j.uct.2008.02.001. PMID 11686011

  30. Zheng Y, Zhu L, Fan L, Zhao W, Wang J, Hao X, Zhu Y, Hu X, Yuan Y, Shao J, Wang W. Synthesis, SAR and pharmacological characterization of novel anthraquinone cation compounds as potential anticancer agents. Eur J Med Chem. 2017;125:902-13. doi: 10.1016/j.ejmech.2016.10.012, PMID 27769031.

  31. Diaz Munoz G, Miranda IL, Sartori SK, de Rezende DC, Diaz MAN. Anthraquinones: an overview. Stud Nat Prod Chem. 2018;58:313-38. doi: 10.1016/B978-0-444-64056-7.00011-8.

  32. DeVita Jr V, Rosenberg SA, DeVita-Cancer HS. Principles and practice of oncology (Jul); 2001.

  33. Feofanov A, Sharonov S, Fleury F, Kudelina I, Nabiev I. Quantitative confocal spectral imaging analysis of mitoxantrone within living K562 cells: intracellular accumulation and distribution of monomers, aggregates, naphtoquinoxaline metabolite, and drug-target complexes. Biophys J. 1997;73(6):3328-36. doi: 10.1016/S0006-3495(97)78357-7, PMID 9414243.

  34. Lorna De Leoz MA, Chua MT, Ann Endoma-Arias MA, Concepcion GP, Cruz LJ, De Leoz MLA. A modified procedure for the preparation of mitoxantrone. Philipp J Sci. 2006;135(2):83-92.

  35. Von Hoff DD, Coltman CA, Forseth B. Activity of mitoxantrone in a human tumor cloning system. Cancer Res. 1981;41(5):1853-5. PMID 7214352.

  36. Neidhart JA, Gochnour D, Roach R, Hoth D, Young D. A comparison of mitoxantrone and doxorubicin in breast cancer. J Clin Oncol. 1986;4(5):672-7. doi: 10.1200/JCO.1986.4.5.672, PMID 3517241.

  37. Zee-Cheng RK, Cheng CC. Antineoplastic agents. Structure–activity relationship study of bis (substituted aminoalkylamino) anthraquinones. J Med Chem. 1978;21(3):291-4. doi: 10.1021/jm00201a012, PMID 628005.

  38. YAP HY, Yap ITY, Blumenshcin GR, Schell FC, Buzdar A, Valdivieso M BG. Dihydroxyanthracenedione: A promising new drug in the treatment of metastatic breast cancer. Ann Intern Med. 1981 Dec 1;95(6):694.

  39. Ling G, Zhang T, Zhang P, Sun J, He Z. Synergistic and complete reversal of the multidrug resistance of mitoxantrone hydrochloride by three-in-one multifunctional lipid-sodium glycocholate nanocarriers based on simultaneous BCRP and Bcl-2 inhibition. Int J Nanomedicine. 2016;11:4077-91. doi: 10.2147/IJN.S95767, PMID 27601896.

  40. Smith IE, Stuart-Harris R, Pavlidis N, Bozek T. Mitoxantrone (Novantrone) as single agent and in combination chemotherapy in the treatment of advanced breast cancer. Cancer Treat Rev. 1983;10;Suppl B:37-40. doi: 10.1016/0305-7372(83)90020-8, PMID 6661733.

  41. Neidhart JA, Gochnour D, Roach RW, Steinberg JA, Young D. Mitoxantrone versus doxorubicin in advanced breast cancer: A randomized cross-over trial. Cancer Treat Rev. 1983;10;Suppl B:41-6. doi: 10.1016/0305-7372(83)90021-x, PMID 6362877.

  42. Brufman G, Haim N, Ben-Baruch N, Sulkes A. Second-line chemotherapy with mitoxantrone as a single agent in metastatic breast cancer. J Chemother. 1993;5(1):43-6. doi: 10.1080/1120009x.1993.11739208, PMID 8459264.

  43. Montazerabadi A-RR, Sazgarnia A, Bahreyni-Toosi MH, Ahmadi A, Shakeri-Zadeh A, Aledavood A. Mitoxantrone as a prospective photosensitizer for photodynamic therapy of breast cancer. Photodiagn Photodyn Ther. 2012 Mar;9(1):46-51. doi: 10.1016/j.pdpdt.2011.08.004, PMID 22369728.

  44. Carter KA, Wang S, Geng J, Luo D, Shao S, Lovell JF. Metal chelation modulates phototherapeutic properties of mitoxantrone-loaded porphyrin–phospholipid liposomes. Mol Pharm. 2016 Feb;13(2):420-7. doi: 10.1021/acs.molpharmaceut.5b00653, PMID 26691879.

  45. Jain V, Kumar H, Anod HV, Chand P, Gupta NV, Dey S, Kesharwani SS. A review of nanotechnology-based approaches for breast cancer and triple-negative breast cancer. J Control Release. 2020;326(Apr):628-47. doi: 10.1016/j.jconrel.2020.07.003, PMID 32653502.

  46. Bowden GT, Peng YM AD. Comparative molecular pharmacology of the anthracene anticancer drugs bisantrene and mitoxantrone. Proc Am Assoc Cancer Res. 1984;25(Mar):296.

  47. Chand P, Kumar H, Badduri N, Gupta NV, Bettada VG, Madhunapantula SV, Kesharwani SS, Dey S, Jain V. Design and evaluation of cabazitaxel loaded NLCs against breast cancer cell lines. Colloids Surf B Biointerfaces. 2021 Mar;199:111535. doi: 10.1016/j.colsurfb.2020.111535.

  48. Cornbleet MA, Stuart Harris RC, Smith IE, Coleman RE, Rubens RD, McDonald M, Mouridsen HT, Rainer H, van Oosterom AT, Smyth JF SJ. Mitoxantrone for the treatment of advanced breast cancer: single-agent therapy in previously untreated patients. Eur J Cancer Clin Oncol. 1984;20(9):1141-6. doi: 10.1016/0277-5379(84)90122-6, PMID 6541135.

  49. Tao X, Tao T, Wen Y, Yi J, He L, Huang Z, Nie Y, Yao X, Wang Y, He C, Yang X. Novel delivery of mitoxantrone with hydrophobically modified pullulan nanoparticles to inhibit bladder cancer cell and the effect of nano-drug size on inhibition efficiency. Nanoscale Res Lett. 2018;13(1):345. doi: 10.1186/s11671-018-2769-x, PMID 30377872.

  50. Carmen Avendano JCC, Avendano C, Menendez JC, Carmen Avendano JCC, Avendano C, Menendez JC. Anticancer drugs acting via radical species. Med Chem Anticancer Drugs. 2015;20:133-95.

  51. Cheng CC. The design, synthesis and development of a new class of potent antineoplastic anthraquinones. Prog Med Chem. 1983;20:83-118. doi: 10.1016/s0079-6468(08)70217-0.

  52. Adamson RH. Letter: Daunomycin (NSC-82151) and adriamycin (NSC-123127): a hypothesis concerning antitumor activity and cardiotoxicity. Cancer Chemother Rep. 1974;58(3):293. PMID 4841712.

  53. Durr FE. Biochemical pharmacology and tumor biology of mitoxantrone and ametantrone. In: Lown JW, Edseditor Anthracycline and Aanthracenedione-Bbased Aanticancer Aagents. The Netherlands: Amsterdam, The Netherlands; 1988. p. 163-200.

  54. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004;56(2):185-229. doi: 10.1124/pr.56.2.6, PMID 15169927.

  55. Mitoxantrone injection product monograph. Novopharm Limited. Rose BD editor. Mitoxantrone. 15.3 ed. Waltham, MA; 2008.

  56. Richmond Hill O. Mitoxantrone injection package insert. Pharmaceutical partners of Canada; 2021.

  57. Et CM. Mitoxantrone (Novantrone) Drugs.com. 2021.

  58. Saint Laurent Q. Mitoxantrone injection, USP product monograph. Hospira Healthcare Corporation; 2014.

  59. Stuart Harris RC, Smith IE. Mitoxantrone: a phase II study in the treatment of patients with advanced breast carcinoma and other solid tumours. Cancer Chemother Pharmacol. 1982;8(2):179-82. doi: 10.1007/BF00255480, PMID 7105382.

  60. Durr Fe, Wallace Re Citarella Rv. Molecular and biochemical pharmacology of mitoxantrone. Cancer Treat Rev. 1983;10:3-11.

  61. Niang M, Soukup T, Bukac J, Siman P, Stoklasova A, Cerman J. Chemotherapy: open access biochemical and pharmacological effects of mitoxantrone and acetyl-L-carnitine in mice with a solid form of Ehrlich tumour. 2015;5(1):1-2.

  62. Traganos F, Evenson DP, Staiano-Coico L, Darzynkiewicz Z, Melamed MR, Evenson DP, Staiano-coico L, Darzynkiewicz Z Melamed MR. Action of dihydroxyanthraquinone on cell cycle progression and survival of a variety of cultured mammalian cells. Cancer Res. 1980;40(3):671-81. PMID 6162553.

  63. AM Huang LKL, Huang AM, Lin KW LW, Huang AM LKL, KL. 1-Hydroxy-3-[(E)-4-(piperazine-diium)but-2-enyloxy]-9,10-anthraquinone ditrifluoroactate induced autophagic cell death in human PC3 cells. Chem Biol Interact. 2018;281:60-8.

  64. Liu Y, Liu Y ZYT. An autophagy-dependent cell death of MDA-MB-231 cells triggered by a novel Rhein derivative 4F. Anticancer Drugs. 2019;30(10):1038-47. doi: 10.1097/CAD.0000000000000820, PMID 31274517.

  65. Chen H, Zhao C, He R, Zhou M, Liu Y, Guo X, Wang M, Zhu F, Qin R, Li X ZCH, Chen H, Zhao C HR. Danthron suppresses autophagy and sensitizes pancreatic cancer cells to doxorubicin. Toxicol Vitr. 2019;54:345-53. doi: 10.1016/j.tiv.2018.10.019.

  66. Tian W, LJS JS, Tian W, Li J SZ, Tian W LJS. Novel anthraquinone compounds induce cancer cell death through paraptosis. ACS Med Chem Lett. 2019;10(5):732–6.

  67. Wang D, Wang S, Liu Q, Wang M, Wang C, Yang H. SZ-685C exhibits potent anticancer activity in both radiosensitive and radioresistant NPC cells through the miR-205-PTEN-Akt pathway. Oncol Rep. 2013;29(6):2341-7. doi: 10.3892/or.2013.2376, PMID 23564023.

  68. Su Z, Li Z, Wang C, Tian W, Lan F, Liang D, Li J, Li D, Hou H. A novel rhein derivative: activation of Rac1/NADPH pathway enhances sensitivity of nasopharyngeal carcinoma cells to radiotherapy. Cell Signal. 2019;54:35-45. doi: 10.1016/j.cellsig.2018.11.015. PMID 30463023.

  69. Koerner SK, Hanai JI, Bai S, Jernigan FE, Oki M, Komaba C, Shuto E, Sukhatme VP, Sun L BS, Koerner SK HJB. Design and synthesis of emodin derivatives as novel inhibitors of ATP-citrate lyase. Eur J Med Chem. 2017;126:920-8. doi: 10.1016/j.ejmech.2016.12.018, PMID 27997879.

  70. YYZ WQ, Wang Q, Yan Y ZJ. Physcion 8-O-β-glucopyranoside inhibits clear-cell renal cell carcinoma bydownregulating hexokinase II and inhibiting glycolysis. Biomed Pharmacother. 2018;104:28-35.

  71. Huang K, Jiang L LH, Huang K JLL. Development of anthraquinone analogs as phosphoglycerate mutase 1 inhibitors. Molecules. 2019;24(5):845.

  72. Lown JW, Hanstock CC, Bradley RD SD, Lown JW, Hanstock CC, Bradley BD, Scraba DG, Lown JW, Hanstock CC, Bradley RD SD. Interactions of the antitumor agents mitoxantrone and bisantrene with deoxyribonucleic acids studied by electron microscopy. Mol Pharmacol. 1984;25(1):178–84.

  73. Foye WO, Vajragupta O, Sengupta SK. DNA-binding specificity and RNA polymerase inhibitory activity of bis(aminoalkyl)anthraquinones and bis(methylthio)vinylquinolinium iodides. J Pharm Sci. 1982;71(2):253-7. doi: 10.1002/jps.2600710228, PMID 7038093.

  74. Alberts DS, Yei L, Peng M, Bowden GT, Dalton WS, Mackel C. Pharmacology of mitoxantrone: mode of action and pharmacokinetics x section. Hematology/Oncology. 1985;107:101-7.

  75. Garnier F, Debat H, Nadal M. Type IA DNA topoisomerases: A universal core and multiple activities. Methods Mol Biol. 2018;1703:1-20. doi: 10.1007/978-1-4939-7459-7_1, PMID 29177730.

  76. Fry AM, Chresta CM, Davies SM, Claire Walker MC, Harris AL, Hartley JA, Masters JR, Hickson ID. Relationship between topoisomerase II level and chemosensitivity in human tumor cell lines. Cancer Res. 1991;51(24):6592-5. PMID 1660343.

  77. Sissi C, Palumbo M. Effects of magnesium and related divalent metal ions in topoisomerase structure and function. Nucleic Acids Res. 2009 Feb 1;37(3):702-11. doi: 10.1093/nar/gkp024, PMID 19188255.

  78. Hsiang YH, Liu LF. DNA topoisomerase poisons as antitumor drugs. Cancer chemother challenges futur. Proceedings of the fourth Nagoya international symposium cancer treat ICS904; 1989. p. 305-11.

  79. Depowski PL, Rosenthal SI, Brien TP, Stylos S, Johnson RL, Ross JS. Topoisomerase IIα expression in breast cancer: correlation with outcome variables. Mod Pathol. 2000;13(5):542-7. doi: 10.1038/modpathol.3880094, PMID 10824926.

  80. Hevener K, Verstak TA, Lutat KE, Riggsbee DL, Mooney JW. Recent developments in topoisomerase-targeted cancer chemotherapy. Acta Pharm Sin B. 2018 Oct;8(6):844-61. doi: 10.1016/j.apsb.2018.07.008, PMID 30505655.

  81. Abu Saleh M, Solayman M, Hoque MM, Khan MAK, Sarwar MG, Halim MA. Inhibition of DNA topoisomerase Type II α (TOP2A) by mitoxantrone and its halogenated derivatives: A combined density functional and molecular docking study. BioMed Res Int. 2016;2016:1-12. doi: 10.1155/2016/6817502.

  82. Hevener KE, Verstak TA, Lutat KE, Riggsbee DL, Mooney JW. Recent developments in topoisomerase-targeted cancer chemotherapy. Acta Pharm Sin B. 2018;8(6):844-61. doi: 10.1016/j.apsb.2018.07.008, PMID 30505655.

  83. Nitiss JL. Targeting DNA topoisomerase II in cancer chemotherapy. Nat Rev Cancer. 2009 May 20;9(5):338-50. doi: 10.1038/nrc2607, PMID 19377506.

  84. Pommier Y, Leo E, Zhang H, Marchand C. DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem Biol. 2010 May;17(5):421-33. doi: 10.1016/j.chembiol.2010.04.012, PMID 20534341.

  85. Ali JA, Jackson AP, Howells AJ, Maxwell A. The 43-kilodalton N-terminal fragment of the DNA gyrase B protein hydrolyzes ATP and binds coumarin drugs. Biochemistry. 1993 Mar 16;32(10):2717-24. doi: 10.1021/bi00061a033, PMID 8383523.

  86. Bisacchi GS, Manchester JI. A new-class antibacterial-almost. Lessons in drug discovery and development: a critical analysis of more than 50 years of effort toward ATPase inhibitors of DNA gyrase and topoisomerase IV. ACS Infect Dis. 2015 Jan 9;1(1):4-41. doi: 10.1021/id500013t, PMID 27620144.

  87. Lindsey RH, Pendleton M, Ashley RE, Mercer SL, Deweese JE, Osheroff N. Catalytic core of human topoisomerase IIα: insights into enzyme–DNA interactions and drug mechanism. Biochemistry. 2014 Oct 21;53(41):6595-602. doi: 10.1021/bi5010816, PMID 25280269.

  88. Gibson EG, Deweese JE. Covalent poisons of topoisomerase II. Curr Top Pharmacol. 2013;17(1):1-12.

  89. Deweese JE, Osheroff N. The DNA cleavage reaction of topoisomerase II: Wolf in sheep’s clothing. Nucleic Acids Res. 2009 Feb 1;37(3):738-48. doi: 10.1093/nar/gkn937, PMID 19042970.

  90. Roca J, Wang JC. DNA transport by a type II DNA topoisomerase: evidence in favor of a two-gate mechanism. Cell. 1994 May;77(4):609-16. doi: 10.1016/0092-8674(94)90222-4, PMID 8187179.

  91. Maxwell A, Lawson DM. The ATP-binding site of type II topoisomerases as a target for antibacterial drugs. Curr Top Med Chem. 2003 Jan 1;3(3):283-303. doi: 10.2174/1568026033452500, PMID 12570764.

  92. Chene P, Rudloff J, Schoepfer J, Furet P, Meier P, Qian Z, Schlaeppi JM, Schmitz R, Radimerski T. Catalytic inhibition of topoisomerase II by a novel rationally designed ATP-competitive purine analogue. BMC Chem Biol. 2009 Dec 7;9(1):1. doi: 10.1186/1472-6769-9-1, PMID 19128485.

  93. Roboz J, Richardson CL, Holland JF. Comparison of the interaction of antineoplastic aminoanthraquinone analogs with DNA using competitive fluorescence polarization. Life Sci. 1982;31(1):25-30. doi: 10.1016/0024-3205(82)90396-4, PMID 7109851.

  94. Faulds D, Balfour JA, Chrisp P, Langtry HD. Mitoxantrone. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in the chemotherapy of cancer. Drugs. 1991;41(3):400-49. doi: 10.2165/00003495-199141030-00007, PMID 1711446.

  95. Benet LZ, Broccatelli F, Oprea TI. BDDCS applied to over 900 drugs. AAPS J. 2011;13(4):519-47. doi: 10.1208/s12248-011-9290-9, PMID 21818695.

  96. Wu CY, Benet LZ. Predicting drug disposition via application of BCS: transport/absorption/ elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res. 2005;22(1):11-23. doi: 10.1007/s11095-004-9004-4, PMID 15771225.

  97. Zhang P, Ling G, Pan X, Zhang P, Ling G, Pan X, Sun J, Zhang T. Novel nanostructured lipid-dextran sulfate hybrid carriers overcome tumor multidrug resistance of mitoxantrone hydrochloride. Nanomedicine Nanotechnology, Biol Med. 2012;8(2):185–93.

  98. Mussi SV, Silva RC, Oliveira MC, Lucci CM, Azevedo RB, Ferreira LA. New approach to improve encapsulation and antitumor activity of doxorubicin loaded in solid lipid nanoparticles. Eur J Pharm Sci. 2013;48(1-2):282-90. doi: 10.1016/j.ejps.2012.10.025, PMID 23178339.

  99. Singh R, Mehra NK, Jain V, Jain NK. Gemcitabine-loaded smart carbon nanotubes for effective targeting to cancer cells. J Drug Target. 2013 Jul 14;21(6):581-92. doi: 10.3109/1061186X.2013.778264, PMID 23484494.

  100. Khan I, Kumar H, Mishra G, Gothwal A, Kesharwani P, Gupta U. Polymeric nanocarriers: A new horizon for the effective management of breast cancer. Curr Pharm Des. 2017;23(35):5315-26. doi: 10.2174/1381612823666170829164828, PMID 28875848.

  101. Muller RH, Radtke M, Wissing SA. Nanostructured lipid matrices for improved microencapsulation of drugs. Int J Pharm. 2002;242(1-2):121-8. doi: 10.1016/s0378-5173(02)00180-1, PMID 12176234.

  102. Jain V, Kumar H, Chand P, Jain S, SP. Lipid‐based nanocarriers as drug delivery system and its applications. In: Nanopharmaceutical advanced delivery systems. Wiley; 2021. p. 1-29.

  103. Doughty JC, Kane E, Cooke TG, McArdle CS. Mitoxantrone and methotrexate chemotherapy with and without mitomycin C in the regional treatment of locally advanced breast cancer. Breast. 2002;11(1):97-9. doi: 10.1054/brst.2001.0316, PMID 14965654.

  104. Hagemeister F, Cabanillas F, Coleman M, Gregory SA, Zinzani PL. The role of mitoxantrone in the treatment of indolent lymphomas. Oncologist. 2005;10(2):150-9. doi: 10.1634/theoncologist.10-2-150, PMID 15709217.

  105. Von Hoff DD, Pollard E, Kuhn J, Murray E, Coltman CA. Phase I clinical investigation of 1,4-dihydroxy-5,8-bis (((2-[(2-hydroxyethyl) amino] ethyl) amino))-9,10-anthracenedione dihydrochloride (NSC 301739), a new anthracenedione. Cancer Res. 1980 May;40(5):1516-8. PMID 7370989.

  106. Shenkenberg TD, Von Hoff DD. Mitoxantrone: A new anticancer drug with significant clinical activity. Ann Intern Med. 1986 Jul 1;105(1):67-81. doi: 10.7326/0003-4819-105-1-67, PMID 3521429.

  107. Lalhlenmawia H. formulation and in vitro evaluation of poly(d, l-lactide-co-glycolide) (plga) nanoparticles of ellagic acid and its effect on human breast cancer, mcf-7 cell line. Int J Curr Pharm Res 2021;13(5):56-62. doi: 10.22159/ijcpr.2021v13i5

  108. Kesharwani SS, Jain V, Dey S, Sharma S, Mallya P, Kumar VA. An overview of advanced formulation and nanotechnology-based approaches for solubility and bioavailability enhancement of silymarin. J Drug Deliv Sci Technol. 2020 Dec;60. doi: 10.1016/j.jddst.2020.102021, PMID 102021.

  109. Du Q, Chen H. The methoxyflavones in Citrus reticulata Blanco cv. ponkan and their antiproliferative activity against cancer cells. Food Chem. 2010;119(2):567-72. doi: 10.1016/j.foodchem.2009.06.059.

  110. Lu B, Xiong SB, Yang H, Yin XD, Chao RB. Solid lipid nanoparticles of mitoxantrone for local injection against breast cancer and its lymph node metastases. Eur J Pharm Sci. 2006;28(1-2):86-95. doi: 10.1016/j.ejps.2006.01.001, PMID 16472996.

  111. Li Z, Cai Y, Zhao Y, Yu H, Zhou H, Chen M. Polymeric mixed micelles loaded mitoxantrone for overcoming multidrug resistance in breast cancer via photodynamic therapy. Int J Nanomedicine. 2017;12:6595-604. doi: 10.2147/IJN.S138235, PMID 28919756.

  112. Lam P, Lin RD, Steinmetz NF. Delivery of mitoxantrone using a plant virus-based nanoparticle for the treatment of glioblastomas. J Mater Chem B. 2018;6(37):5888-95. doi: 10.1039/C8TB01191E, PMID 30923616.

  113. Lu B, Xiong SB, Yang H, Yin XD, Chao RB. Solid lipid nanoparticles of mitoxantrone for local injection against breast cancer and its lymph node metastases. Eur J Pharm Sci. 2006;28(1-2):86-95. doi: 10.1016/j.ejps.2006.01.001, PMID 16472996.

  114. Toh TB, Lee DK, Hou W, Abdullah LN, Nguyen J, Ho D, Chow EK. Nanodiamond − mitoxantrone complexes enhance drug retention in chemoresistant breast cancer cells. Mol Pharm. 2014;11(8):2683-91. doi: 10.1021/mp5001108, PMID 24867631.

  115. Ling G, Zhang T, Zhang P, Sun J, He Z. Nanostructured lipid-carrageenan hybrid carriers (NLCCs) for controlled delivery of mitoxantrone hydrochloride to enhance anticancer activity bypassing the BCRP-mediated efflux. Drug Dev Ind Pharm. 2016;42(8):1351-9. doi: 10.3109/03639045.2015.1135937, PMID 26754913.

  116. Zhang LK, Hou SX, Zhang JQ, Hu WJ, Wang CY. Preparation, characterization, and in vivo evaluation of mitoxantrone-loaded, folate-conjugated albumin nanoparticles. Arch Pharm Res. 2010;33(8):1193-8. doi: 10.1007/s12272-010-0809-x, PMID 20803122.

  117. Sargazi A, Kamali N, Shiri F, Heidari Majd M. Hyaluronic acid/polyethylene glycol nanoparticles for controlled delivery of mitoxantrone. Artif Cells Nanomed Biotechnol. 2018;46(3):500-9. doi: 10.1080/21691401.2017.1324462, PMID 28503952.

  118. Wang C, Han M, Liu X, Chen S, Hu F, Sun J, Yuan H. Mitoxantrone-preloaded water-responsive phospholipid-amorphous calcium carbonate hybrid nanoparticles for targeted and effective cancer therapy. Int J Nanomedicine. 2019;14:1503-17. doi: 10.2147/IJN.S193976, PMID 30880961.

  119. Yoon JH, Cho HJ, Jin HE, Maeng HJ. Mitoxantrone-loaded pegylated gold nanocomplexes for cancer therapy. J Nanosci Nanotechnol. 2019;19(2):687-90. doi: 10.1166/jnn.2019.15902, PMID 30360142.

  120. Toh TB, Lee DK, Hou W, Abdullah LN, Nguyen J, Ho D, Chow EK. Nanodiamond-mitoxantrone complexes enhance drug retention in chemoresistant breast cancer cells. Mol Pharm. 2014;11(8):2683-91. doi: 10.1021/mp5001108, PMID 24867631.

  121. Development D, Pharmacy I. Nanostructured lipid-carrageenan hybrid carriers (NLCCs) for controlled delivery of mitoxantrone hydro-chloride to enhance anticancer activity bypassing the BCRP-mediated efflux. 2015.

  122. Stuart-Harris RC, Bozek T, Pavlidis NA, Smith IE Mitoxantrone: an active new agent in the treatment of advanced breast cancer. Cancer Chemother Pharmacol. 1984;12(1):1-4. doi: 10.1007/BF00255899. PMID 6690066.

  123. Hendrick AM, Harris AL, Cantwell BMJ. Verapamil with mitoxantrone for advanced ovarian cancer: A negative phase II trial. Ann Oncol. 1991;2(1):71-2. doi: 10.1093/oxfordjournals.annonc.a057830.

  124. Dunn CJ, Goa KL. Mitoxantrone: a review of its pharmacological properties and use in acute nonlymphoblastic leukaemia. Drugs Aging. 1996;9(2):122-47. doi: 10.2165/00002512-199609020-00007, PMID 8820798.

  125. Ma S, Au K, Wan T, Chan L. Translocation in blastic transformation of atypical chronic myeloid leukemia. Leukemia. 1997;11(4):612-3. doi: 10.1038/sj.leu.2400612.

  126. Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. Blood. 2005;106(4):1154-63. doi: 10.1182/blood-2005-01-0178, PMID 15870183.

  127. Comella G, Casaretti R, Comella P, Antonio Daponte AP, Parziale A, Iervolino V, Santillo G, Zarrilli D. Treatment of advanced colorectal cancer with mitoxantrone, high dose folinic acid and fluorouracil. Tumori. 1991;77(5):445-6. doi: 10.1177/030089169107700515, PMID 1781041.

  128. Fusi A, Procopio G, Della Torre S, Ricotta R, Bianchini G, Salvioni R, Ferrari L, Martinetti A, Savelli G, Villa S, Bajetta E. Treatment options in hormone-refractory metastatic prostate carcinoma. Tumori. 2004;90(6):535-46. doi: 10.1177/030089160409000601, PMID 15762353.

  129. Hu OYP, Chang SP, Song YB, Chen KY, Law CK. Novel assay method for mitoxantrone in plasma, and its application in cancer patients. J Chromatogr B Biomed Sci Appl. 1990 Jan;532(2):337-50. doi: 10.1016/s0378-4347(00)83783-4, PMID 2084130.

  130. Patel KJ, Tredan O, Tannock IF. Distribution of the anticancer drugs doxorubicin, mitoxantrone and topotecan in tumors and normal tissues. Cancer Chemother Pharmacol. 2013;72(1):127-38. doi: 10.1007/s00280-013-2176-z, PMID 23680920.

  131. Panousis C, Kettle AJ, Phillips DR. Neutrophil-mediated activation of mitoxantrone to metabolites which form adducts with DNA. Cancer Lett. 1997;113(1-2):173-8. doi: 10.1016/s0304-3835(97)04611-9, PMID 9065819.

  132. Stuart Harris RC, Smith IE. Mitoxantrone: A phase II study in the treatment of patients with advanced breast carcinoma and other solid tumours. Cancer Chemother Pharmacol. 1982;8(2):179-82. doi: 10.1007/BF00255480, PMID 7105382.

  133. Ehlllllger G, Schiller C, Proksch B, Zeller K, Blaf J. Dnoa Connect; 1990.

  134. Smyth JF, Macpherson JS, Warrington PS, Leonard RCF, Wolf CR. The clinical pharmacology of mitozantrone. Cancer Chemother Pharmacol. 1986;17(2):149-52. doi: 10.1007/BF00306744, PMID 3719894.

  135. Richard B, Fabre G, De Sousa G, Fabre I, Rahmani R, Cano JP. Interspecies variability in mitoxantrone metabolism using primary cultures of hepatocytes isolated from rat, rabbit and humans. Biochem Pharmacol. 1991;41(2):255-62. doi: 10.1016/0006-2952(91)90484-m, PMID 1989635.

  136. Alberts DS, Peng YM, Leigh S, Davis TP, Woodward DL. Disposition of mitoxantrone in patients. Cancer Treal Rev Cancer Treat Rev. 1983;10Suppl B:23-7. doi: 10.1016/0305-7372(83)90018-x, PMID 6661732.

  137. Chiccarelli FS, Morrison JA, Cosulich DB, Perkinson NA, Ridge DN, Sum FW, Murdock KC, Woodward DL, Arnold ET. Identification of human urinary mitoxantrone metabolites. Cancer Res. 1986;46(9):4858-61. PMID 3731132.

  138. Richard B, Fabre G, Desousa G, Cano JP. Metabolism of mitoxantrone by hepatocytes in primary culture isolated from different species including man. Proc Am Assoc Cancer Res. 1987;28:1674.

  139. Benet LZ, Broccatelli F, Oprea TI. BDDCS applied to over 900 drugs. AAPS J. 2011;13(4):519-47. doi: 10.1208/s12248-011-9290-9, PMID 21818695.

  140. Batra VK, Morrison JA, Woodward DL, Siverd NS, Yacobi A. Pharmacokinetics of mitoxantrone in man and laboratory animals. Drug Metab Rev. 1986;17(3-4):311-29. doi: 10.3109/03602538608998294, PMID 3552542.

  141. Alberts DS, Peng Y, Leigh S, Davis TP, Woodward DL. Disposition of Mitoxantronein cancer patients. Cancer Res. 1985;45(4):1879-84.

  142. Kilmer PD. Review aarticle: rreview aarticle. Journal Theory, Pract Crit Journalism. 2010;11(3):369-73. doi: 10.1177/1461444810365020.

  143. Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK. A multidrug resistance transporter from human MCF-7 breast cancer cells (mitoxantrone͞ anthracyclines͞ transporter proteins). Med Sci. 1998;95(Dec):15665-70.

  144. Miyake K, Mickley L, Litman T, Zhan Z, Robey R, Cristensen B, Brangi M, Greenberger L, Dean M, Fojo T, Bates SE. Molecular cloning of cDNAs which are highly overexpressed in mitoxantrone-resistant cells: demonstration of homology to ABC transport genes. Cancer Res. 1999;59(1):8-13. PMID 9892175.

  145. Zhu X, Wong ILK, Chan KF, Cui J, Law MC, Chong TC, Hu X, Chow LMC, Chan TH. Triazole bridged flavonoid dimers as potent, nontoxic, and highly selective breast cancer resistance protein (BCRP/ABCG2) inhibitors. J Med Chem. 2019 Sep 26;62(18):8578-608. doi: 10.1021/acs.jmedchem.9b00963, PMID 31465686.

  146. Gillet JP, Gottesman MM. Mechanisms of multidrug resistance in cancer. In. Methods Mol Biol. 2010;596:47-76. doi: 10.1007/978-1-60761-416-6_4, PMID 19949920.

  147. Gerlach JH, Kartner N, Bell DR, Ling V. Multidrug resistance. Cancer Surv. 1986;5(1):25-46. PMID 2885085.

  148. Roninson IB, Chin JE, Choi KG, Gros P, Housman DE, Fojo A, Shen DW, Gottesman MM, Pastan I. Isolation of human mdr DNA sequences amplified in multidrug-resistant KB carcinoma cells. Proc Natl Acad Sci. USA. 1986 Jun 1;83(12):4538-42. doi: 10.1073/pnas.83.12.4538, PMID 3459187.

  149. Endicott JA, Ling V. The biochemistry of P-glycoprotein-mediated multidrug resistance. Annu Rev Biochem. 1989 Jun;58(1):137-71. doi: 10.1146/annurev.bi.58.070189.001033, PMID 2570548.

  150. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP–dependent transporters. Nat Rev Cancer. 2002 Jan;2(1):48-58. doi: 10.1038/nrc706, PMID 11902585.

  151. Gottesman MM, Pastan IH. The role of multidrug resistance efflux pumps in cancer: revisiting a JNCI publication exploring expression of the MDR1 (P-glycoprotein) gene. J Natl Cancer Inst. 2015 Sep 18;107(9):djv222. doi: 10.1093/jnci/djv222, PMID 26286731.

  152. Schabel FM, Skipper HE, Trader MW, Laster WR, Griswold DP, Corbett TH. Establishment of cross-resistance profiles for new agents. Cancer Treat Rep. 1983;67(10):905-22. PMID 6354439.

  153. Biedler JL, Spengler BA. Reverse transformation of multidrug-resistant cells. Cancer Metastasis Rev. 1994;13(2):191-207. doi: 10.1007/BF00689636, PMID 7923550.

  154. Mansoori B, Mohammadi A, Davudian S, Shirjang S, Baradaran B. The different mechanisms of cancer drug resistance: A brief review. Adv Pharm Bull. 2017;7(3):339-48. doi: 10.15171/apb.2017.041, PMID 29071215.

  155. Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, Ross DD. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci USA. 1998 Dec 22;95(26):15665-70. doi: 10.1073/pnas.95.26.15665, PMID 9861027.

  156. Yuan JH, Cheng JQ, Jiang LY, JI WD, Guo LF, Liu JJ, Xu XY, He JS, Wang XM, Zhuang ZX. Breast cancer resistance protein expression and 5-fluorouracil resistance. Biomed Environ Sci. 2008 Aug;21(4):290-5. doi: 10.1016/S0895-3988(08)60044-6, PMID 18837291.

  157. Austin Doyle L, Ross DD. Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2). Oncogene. 2003 Oct 23;22(47):7340-58. doi: 10.1038/sj.onc.1206938, PMID 14576842.

  158. Natarajan K, Xie Y, Baer MR, Ross DD. Role of breast cancer resistance protein (BCRP/ABCG2) in cancer drug resistance. Biochem Pharmacol. 2012 Apr;83(8):1084-103. doi: 10.1016/j.bcp.2012.01.002, PMID 22248732.

  159. Diah SK, Smitherman PK, Aldridge J, Volk EL, Schneider E, Townsend AJ, Morrow CS. Resistance to mitoxantrone in multidrug-resistant MCF7 breast cancer cells: evaluation of mitoxantrone transport and the role of multidrug resistance protein family proteins. Cancer Res. 2001;61(14):5461-7. PMID 11454692.

  160. Hu J, Zhang H, Liu L, Han B, Zhou G, Su P. TRPS1 confers multidrug resistance of breast cancer cells by regulating BCRP expression. Front Oncol. 2020 Jun 30;10:934. doi: 10.3389/fonc.2020.00934, PMID 32695669.

  161. Bolhuis H, van Veen HW, Poolman B, Driessen AJM, Konings WN. Mechanisms of multidrug transporters. FEMS Microbiol Rev. 1997 Aug;21(1):55-84. doi: 10.1111/j.1574-6976.1997.tb00345.x, PMID 9299702.

  162. Dean M, Allikmets R. Complete characterization of the human ABC gene family. J Bioenerg Biomembr. 2001;33(6):475-9. doi: 10.1023/a:1012823120935, PMID 11804189.

  163. Henderson BM, Dougherty WJ, James VC, Tilley LP, Noble JF. Safety assessment of a new anticancer compound, mitoxantrone, in beagle dogs: comparison with doxorubicin. I. Clinical observations. Cancer Treat Rep. 1982;66(5):1139-43. PMID 7083216.

  164. Each I, Agent AI, Code IATC, Saint-laurent TH. Revision P, no SC. Product monograph; 2014. p. 1-38. PMID 180582.

  165. Van de Wyngaert FA, Beguin C, D’Hooghe MB, Dooms G, Lissoir F, Carton H, Sindic CJ. A double-blind clinical trial of mitoxantrone versus methylprednisolone in relapsing, secondary progressive multiple sclerosis. Acta Neurol Belg. 2001;101(4):210-6. PMID 11851027.

  166. Millefiorini E, Gasperini C, Pozzilli C, D’Andrea F, Bastianello S, Trojano M, Morino S, Morra VB, Bozzao A, Calo’ A, Bernini ML, Gambi D, Prencipe M. Randomized placebo-controlled trial of mitoxantrone in relapsing-remitting multiple sclerosis: 24-month clinical and MRI outcome. J Neurol. 1997;244(3):153-9. doi: 10.1007/s004150050066, PMID 9050955.

  167. Dihydroxyanthracenedione SS. Drug name mitoxantrone. BC Cancer Drug Manual. 2019(May):1-9.

  168. Posner LE, Dukart G, Goldberg J, Bernstein T, Cartwright K. Mitoxantrone: an overview of safety and toxicity. Invest New Drugs. 1985;3(2):123-32. doi: 10.1007/BF00174159, PMID 3894276.

  169. Arlin Z, Case DC Jr, Moore J, Wiernik P, Feldman E, Saletan S, Desai P, Sia L, Cartwright K. Randomized multicenter trial of cytosine arabinoside with mitoxantrone or daunorubicin in previously untreated adult patients with acute nonlymphocytic leukemia (ANLL). Lederle Cooperative Group. Leukemia. 1990;4(3):177-83. PMID 2179638.

  170. Garzotto M, Myrthue A, Higano CS, Beer TM. Neoadjuvant mitoxantrone and docetaxel for high-risk localized prostate cancer. Urol Oncol Semin Orig Investig. 2006;24(3):254-9. doi: 10.1016/j.urolonc.2005.11.034, PMID 16678060.

  171. Smith IE, Stuart Harris R, Pavlidis N, Bozek TBT. Mitoxantrone (Novantrone) as single agent and in combination chemotherapy in the treatment of advanced breast cancer. Cancer Treat Rev. 1983:37-40. doi: 10.1016/0305-7372(83)90020-8, PMID 6661733.

  172. Yang J, Shi Y, Li C, Gui L, Zhao X, Liu P, Han X, Song Y, Li N, Du P, Zhang S. Phase I clinical trial of pegylated liposomal mitoxantrone plm60-s: pharmacokinetics, toxicity and preliminary efficacy. Cancer Chemother Pharmacol. 2014;74(3):637–-46. doi: 10.1007/s00280-014-2523-8, PMID 25034977.

  173. Bezwoda WR, Dansey R, Seymour L. First-line chemotherapy of advanced breast cancer with mitoxantrone, cyclophosphamide and vincristine 1. Oncology. 1989;46(4):208-11. doi: 10.1159/000226717, PMID 2740063.

  174. Bally Marcel B, Barber Lana W, Chang Charmaine W, Lim Howard J, Madden Thomas D. Liposomal Formulations Mitoxantrone; 1999.

  175. Wei J Sun. Mingxing, [Mingxing St Juan; wei]. Mitoxantrone sustained-release implantation agent curing entity tumour; 2008.

  176. Ming Pssnwyz, Classifications. Mesoporous silicon dioxide-methotrexate-mitoxantrone nanoparticles as well as preparation, activity and application thereof; 2018.

  177. Qiang Lh, Pei Jin. Cardiolipin-containing new liposome preparation, and its application in antitumor drugs; 2014.

  178. Cheng XF, Min Wang, Shujun Wang. Application of mitoxantrone as lymph tracer; 2012.