Next Article in Journal
Patient Selection and Outcomes of Laparoscopic Microwave Ablation of Hepatocellular Carcinoma
Next Article in Special Issue
Hyperleukocytosis in Childhood Acute Leukemia: Early Complications and Survival Outcomes
Previous Article in Journal
Chondrosarcoma Resistance to Radiation Therapy: Origins and Potential Therapeutic Solutions
Previous Article in Special Issue
Pediatric Myeloid Sarcoma, More than Just a Chloroma: A Review of Clinical Presentations, Significance, and Biology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Battlefield of Chemotherapy in Pediatric Cancers

1
Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA
2
Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
3
Institute for Biotechnology, St. John’s University, Queens, NY 11439, USA
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(7), 1963; https://doi.org/10.3390/cancers15071963
Submission received: 10 February 2023 / Revised: 12 March 2023 / Accepted: 21 March 2023 / Published: 24 March 2023
(This article belongs to the Special Issue Pediatric Cancers)

Abstract

:

Simple Summary

The survival rate for pediatric cancers has improved significantly over the last decades. Conventional chemotherapies play a vital role in pediatric cancer treatment, especially in low- and middle-income countries, and the roster of chemo drugs for use in children has expanded. However, patients suffer from chemotherapy as a result of its countless side effects. Furthermore, multidrug resistance (MDR) continues to be an insurmountable obstacle that limits survival for a considerable number of patients. In this review, we discuss severe side effects in pediatric chemotherapies such as doxorubicin-induced cardiotoxicity (DIC) and vincristine-induced peripheral neuropathy (VIPN). Here, MDR mechanisms in chemotherapy are elucidated with the aim of improving survival, while also reducing the intensity and toxicity of chemotherapy. Furthermore, we focus on various drug transporters in common types of pediatric tumors, which could provide different therapeutic strategies.

Abstract

The survival rate for pediatric cancers has remarkably improved in recent years. Conventional chemotherapy plays a crucial role in treating pediatric cancers, especially in low- and middle-income countries where access to advanced treatments may be limited. The Food and Drug Administration (FDA) approved chemotherapy drugs that can be used in children have expanded, but patients still face numerous side effects from the treatment. In addition, multidrug resistance (MDR) continues to pose a major challenge in improving the survival rates for a significant number of patients. This review focuses on the severe side effects of pediatric chemotherapy, including doxorubicin-induced cardiotoxicity (DIC) and vincristine-induced peripheral neuropathy (VIPN). We also delve into the mechanisms of MDR in chemotherapy to the improve survival and reduce the toxicity of treatment. Additionally, the review focuses on various drug transporters found in common types of pediatric tumors, which could offer different therapeutic options.

1. Introduction

Pediatric cancer is relatively rare and has a high survival rate compared with adult cancer. Pediatric cancer includes 12 major types and over 100 subtypes [1]. In the United States, pediatric cancer therapies have made remarkable advances over the last 70 years, and the overall survival (OS) rate for pediatric cancer patients has increased to 80% [2,3]. However, cancer is still the top cause of death by disease in children. In 2023, diagnosed pediatric cancer patients in the USA will include ~9910 children (birth to age 14) and ~5280 adolescents (aged 15–19 years). In addition, approximately 1040 children and 550 adolescents in the USA will die from cancer in 2023 [4]. An improvement in these amounts can be achieved by an increased understanding of the molecular basis of cancer through international collaborative efforts.
Despite these successes, the incidences and mortalities between countries are greatly different. In 2017, the 5-year survival rate of childhood cancer was 80% in high-income countries. However, data suggest that far fewer children survive in most low- and middle-income countries—the 5-year survival rate of children is ~40%, while more than 90% of children at risk of potential pediatric cancer live in these countries [5,6]. Because of major advances in modern science over the past several decades, the improvement in pediatric cancer in high-income countries has not translated to most low- and middle-income countries. There are still gaps among these countries regarding recognition, diagnosis, and treatment [7].
Despite financial hardship or treatment results, conventional chemotherapy is still a standard option for childhood cancer treatments in low- and middle-income countries, where molecular-targeted drugs are broadly accessible [8,9,10]. Severe side effects and resistance mechanisms against cancer therapies remain the primary reasons limiting chemotherapy outcomes [11]. Here, we consider the advancement of chemotherapy in pediatric cancer, including drugs and specific considerations. Beyond the previous focus on adult cancer alone, in the current review, we discuss severe side effects such as neuropathy and myopathy, the effects of the MDR phenomenon, and the role of various drug transporters in common types of pediatric tumors that could lead to more efficient pediatric chemotherapy.

2. Current Chemotherapy in Pediatric Cancer

There are currently various cancer treatment approaches for cancer. The main four lines of treatment are surgical removal, immunotherapy, radiotherapy, and chemotherapy [12]. Surgical removal has the longest history in cancer treatment. It is an essential treatment option and often is often performed together with other cancer treatments, depending on the type of cancer [13]. Immunotherapy is generally considered to have been applied in 1890 by Dr. Coley, who injected a mixture of live and inactivated Streptococcus and Serratia to achieve responses [14]. It modulates the immune system to act better against cancer, and it includes immune checkpoint inhibitors, T-cell transfer therapy, monoclonal antibodies, and treatment vaccines. Radiotherapy precisely delivers radiation to kill cancer cells and shrink tumors, and was started in 1896 by Emile Grubbé, who treated a breast cancer patient using X-rays [15]. Chemotherapy was first introduced after the Second World War, and works by interfering with cell proliferation. Other cancer treatment approaches include stem cell transplantation, targeted therapy, photodynamic therapy, and hormone therapy.
The efficient treatment of pediatric cancer started after 1945. Sidney Farber treated a 3-year-old patient with acute lymphoblastic leukemia (ALL) with aminopterin [16]. As a conventional cancer therapy, chemotherapy has played an important role in pediatric cancer over the past several decades. In pediatric diagnosis, ALL is the most common type of cancer. The OS of ALL increased from 57% in the 1970s to 96% in recent years [17]. This enormous progress is partly attributed to advances in chemotherapy [3]. Currently, chemotherapy has reached its limit, as severe toxicities have been observed in pediatric patients during treatment. Modifications to chemotherapy are made in order to reduce multiple toxicities. For instance, studies have provided evidence that chronomodulated chemotherapy, based on the body’s intrinsic circadian clock, might minimize toxicity while maintaining an anticancer activity [18]. High-dose chemotherapy has been explored for treating childhood malignant glioma so as to pass the blood−brain barrier (BBB), reduce cell chemoresistance, and achieve a more comprehensive response [19].
Chemo drugs are grouped by function, chemical structure, and interaction with other medications. Types of chemo drugs include alkylating agents, antimetabolites, antibiotics, topoisomerase inhibitors, mitotic inhibitors, and corticosteroids [20,21]. In 1997, under the Food and Drug Administration Modernization Act2 (FDAMA), a pediatric exclusivity provision was enacted by the U.S. Congress. Few chemo drugs were explicitly approved for treating pediatric cancer. The FDAMA later reauthorized as the Best Pharmaceuticals Act for Children (BPCA), which encourages pediatric drug studies in companies by providing a financial incentive. A total of 16 chemo drugs have been approved by the FDA with pediatric indications in the U.S. Before the FDAMA, only nine chemo drugs were approved for pediatric indications, see Table 1. After the FDAMA (1997–2022), seven chemo drugs were approved to treat childhood cancer (see Table 2).
There are less drugs with pediatric indications worldwide compared with adult indications [22]. For some chemo drugs, the initial approval was only for adult indications. On 20 May 2022, Azacitidine was newly approved for childhood cancers, while it first approved on 19 May 2004 for treating all subtypes of myelodysplastic syndrome. Moreover, FDA approved these chemo drugs (Clofarabine, Nelarabine, Erwinia, and Mercaptopurine) for both adult and pediatric indications at the same time (Table 2). Although efforts have been made in pediatric chemotherapy, only a few chemo drugs have gained pediatric indications—there is a gap between pediatric and adult approval.
The therapy to treat pediatric cancers requires specific prerequisites and considerations, as patients are still growing and developing. Some adverse effects of cancer treatments are more severe for children than adults, as developing organs are more susceptible. Infection-related serious adverse events are more common in ALL patients receiving chemotherapy, which enhances the risk of death [23]. Therefore, it is necessary to identify drug resistance mechanisms to reduce chemotherapy intensity and toxicity. The counterstrategies against drug resistance are discussed in the following section.

3. Severe Neuropathy and Myopathy Side Effects in Chemotherapy

In cancer treatment, chemotherapeutic agents are considered a double edge sword, as it kills cancer cells and other healthy, fast-growing cells, without differentiation. As a matter of fact, patients suffer from various non-negligible side effects such as hair loss, fatigue, nausea, vomiting, and diarrhea. Currently, doxorubicin (DOX) and vincristine (VCR) are two commonly used chemotherapeutic agents in pediatric cancer treatment [25,26]. However, the potential harm of neuropathy and myopathy has become a considerate risk factor that limits the effect of DOX and VCR [27,28]. The pathway mechanism of neuropathy and myopathy induced by DOX and VCR in illustrated in Figure 1. Thus, it is necessary to discuss the opposing side and corresponding solutions for these two chemo drugs, mainly when applied to pediatric cancer patients.
DOX, an anthracycline drug to treat solid tumors in children, such as Wilm’s tumor, was approved by the FDA in 1974 to become the first clinical liposomal encapsulated anticancer drug [29]. DOX acts as an intercalation that inhibits topoisomerase II and further obstructs DNA replication [30]. The recommended dosage (2 mg/mL) is the same in both pediatric and adult patients. Symptoms of the side effects may include abnormality in electrocardiography, rhythm disturbances, and even left ventricular hypertrophy that leads to a reduction in ejection fraction [31].
DIC is a severe dose-dependent risk factor in cancer treatment [32]. It has been reported that SNP (rs2229774) in retinoic acid receptor-γ (RARG) has a significant impact on the increased occurrence of DIC. Further investigation found that a RARG agonist CD1530 showed a cardioprotective effect in an in vivo mouse model of DIC [33]. Other drug transporter genomic variants in the SLC28A3 locus were also identified. Based on that, a single dosage of 3 µmol/L desipramine per day before the administration of DOX is recommended to prevent DIC [34]. Moreover, circular RNA (circRNA)-based therapy is a novel approach for treating DIC [35]. The overexpression of insulin receptor encoded circRNA (Circ-INSR) successfully prevented and reversed DIC in an in vivo mouse model [36]. Han. et al. reported another promising therapeutic target for DIC, namely tumor-suppressive human circular RNA CircITCH [37]. Other recent potential strategies that possibly reverse DIC include atg7-based autophagy activation [38], meteorin-like (METRNL) protein [39], sirtuin 1 (SIRT1) [40], berberine [41], ADAR2 [42], elabela (ELA) [43], phenylalanine-butyramide (FBA) [44], gasdermin D [45], melatonin [46], levosimendan [47], paeonol [48], SNX17 [49], irisin [50], isorhapontigenin [51], liensinine [52], etc.
VCR, known initially as Leurocristine, is the first-line chemotherapeutic medication often administered in the combination chemotherapeutic treatment of pediatric hematologic malignancies and solid tumors [53]. VCR acts as mitotic inhibitors by binding to the β-tubulin subunit of αβ-tubulin heterodimers, thus functionally destabilizing microtubule fibers, which ultimately leads to the termination of cancer cell division [54]. Previous studies have shown a cumulative effect for its neurotoxicity and overdosage may cause very serious or fatal outcomes [55,56]. Until 6 June 2022, it has been approved by FDA in the treatment of pediatric ALL, neuroblastoma, non-Hodgkin lymphoma, rhabdomyosarcoma, Wilm’s tumor, and other childhood kidney cancers. USP recommended dose of VCR injection for pediatric patients is 1.5–2 mg/m2 compared to adults is 1.4 mg/m2. For neonate and infant patients weighing 10 kg or less, the first dose should be 0.05 mg/kg, administered once a week. The latest research showed in the treatment of ALL and Wilm’s tumors, compared with older children, neonates or infants have similar clearance in vincristine. Thus, doses less than 0.05 mg/kg should not be applied in neonate and infant patients due to inappropriate suboptimal VCR exposures [57].
The common side effect encountered with VCR use is VIPN [58]. Clinical patterns of VIPN can be classified as sensory neuropathy (e.g., numbness and paresthesia), motor neuropathy (e.g., extremity weakness and walking difficulties), and autonomic neuropathy (e.g., constipation and urinary retention) [59]. VIPN can be assessed by the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), the pediatric-modified Total Neuropathy Scale (ped-m TNS), and the Total Neuropathy Score-Pediatric version (TNS-PV) [28,60]. There are currently no effective strategies for reducing vincristine-induced neurotoxicity. Also, whether VIPN exists chronically in the survivors remains ambiguous [61]. Some studies showed that a significant proportion of patients receiving VCR would undergo a certain extent VIPN [62,63]. In 2017, Tay et al. reported that ~16% of pediatric ALL survivors suffer from VIPN [64]. Nevertheless, in 2020, an investigation among 150 pediatric patients with ALL and Wilm’s tumors showed that significant side effects of the vincristine regimen are mostly neurotoxic, which is at a mild to moderate level [65]. In recent years, more and more studies have revealed the deep connection between genetic polymorphisms and VIPN [66,67,68]. The CEP72 genetic variant is an optimistic VIPN signature marker since the CEP72 gene encodes centrosome proteins that participate in the development of microtubules. Patients that carry CEP72 TT genotype take potentially higher risk and severity of VIPN than CC or CT genotype patients [69]. Besides, the enzyme CYP3A5 contributes to hepatic clearance of VCR, which means CYP3A5 genetic polymorphisms, and its allelic variants are assumed to be associated with VCR neurotoxicity in different human populations. CYP3A5 genotyping analysis results showed over 70% of African Americans had been found to have one or more CYP3A5*1 alleles (such as CYP3A5 expresser), which is about five times higher compared to Caucasians [70]. Moreover, VCR is transported by some members of ATP-binding cassette (ABC) transporter superfamily such as ABCB1, ABCC1, ABCC2, and ABCB4 [71]. Lopez-Lopez et al. reported that ABCC1 is the critical mediator acts transporting VCR into the blood, while ABCB1 and ABCC2 are indispensable in the biliary excretion of VCR. The genotypes rs3740066 GG and rs12826 GG of ABCC2 were identified with significant associations with increased VIPN, which suggested that ABCC2 polymorphism could be used as a potential biomarker for VIPN in screening and diagnosis of pediatric ALL [72].
Recently, numerous updates have been related to the strategy to overcome VIPN. Zhou et al. reported levo-corydalmine ameliorates VIPN in mice by inhibiting Cx43 expression and NFκB-dependent CXCL1/CXCR2 signalling pathway [73,74]. Also, both VIPN and tumor growth were alleviated by the inhibition of histone deacetylase 6 (HDAC6) in mice. Other prospective targets that possibly prevent VIPN include mitoquinone [75], puerarin [76], nerve growth factor (NGF) monoclonal antibody DS002 [77], minocycline [78], bergapten [79], etc.

4. MDR: The Challenge in Pediatric Cancer Chemotherapy

Cancer cells can develop resistance to one chemotherapeutic drug, as well as other chemotherapeutic drugs that may have different chemical properties and mechanisms of action, which is called multi-drug resistance (MDR) [80]. MDR can occur in both adult and pediatric cancer chemotherapy. Although the mechanisms of MDR have been studied for a few decades, MDR is a very limiting factor to the success of cancer chemotherapies. MDR exists not only in chemotherapy and radiation therapy, but also in newly developed therapies such as targeted therapy and immunotherapy [81,82]. Drug resistance can be divided into intrinsic resistance or extrinsic resistance based on the cause of its occurrence. Cancer cells may have inherent drug resistance before receiving chemotherapy. However, various adaptive responses of cancer cells during the treatment cause extrinsic or acquired drug resistance of cancer cells. Several MDR mechanisms have been discovered, yet they are not yet fully understood. As shown in Figure 2, the factors of MDR include increased drug inactivation, reduction of influx and increased efflux of drugs, decreased activation of prodrugs, epigenetic dysregulation, changes in cell surface markers, tumor microenvironment (TME), epithelial−mesenchymal transition (EMT), altered miRNA, disruption of responses to DNA damage, inhibition of apoptotic pathways, tumor heterogeneity, and cancer stem cells [80,83,84].

5. MDR-Related Drug Transporters and Their Roles in Pediatric Cancers

Drug transporters are membrane proteins involved in the absorption, distribution, and excretion of drugs. Two transporter superfamilies have been identified in humans: the solute carrier (SLC) superfamily and the ABC superfamily [85]. ABC transporters are generally involved with the efflux of drugs, and SLC transporters have been chiefly described as influx transporters [86]. In addition to transporting therapeutic drugs across membranes, these transporters also mediate the transport of endogenous compounds. There is considerable interest in transporters from both families, as they are known to confer MDR to cancer cells.

5.1. SLC Family Transporters

SLC family transporters are a family that includes more than 300 membrane-bound proteins involved in the influx and efflux of a wide array of substrates, such as ions, metabolites, and drugs [87]. SLC transports substrates based on the electrochemical potential difference between the biological membrane or the ion gradient originally generated by the primary active transporters. Genetic variants of SLC transporters and clinical outcomes of methotrexate (MTX) have been studied in pediatric patients with ALL [88]. Although SLC transporter families are essential in human health, related studies focusing on pediatric cancer therapy are rare. We limited our discussion to ABC transporters.

5.2. ABC Transporters

The ABC transporter superfamily, one of the most prominent transporter families, is responsible for MDR by mediating drug efflux, which subsequently leads to a low intracellular concentration of antineoplastic agents in cancer cells and deteriorates therapeutic outcome [89]. In addition, more than efflux pumps, other critical roles in cancer development of this transporter superfamily have been revealed step by step [90,91,92,93,94]. So far, 7 subfamilies (ABC-A to ABC-G) and at least 48 additional subfamily members have been found and characterized depending on their structural differences and similarities [95,96]. Among these subfamily members, three members have been demonstrated that are closely related to MDR in chemotherapy, including P-glycoprotein (P-gp/ABCB1/MDR1), multidrug resistance protein 1 (MRP1/ABCC1), and breast cancer resistance protein (BCRP/ABCG2) [97,98].

5.2.1. MDR1

The ABCB subfamily involves four full transporters (ABCB1/4/5/11) and 7 half transporters (ABCB2/3/6/7/8/9/10) that can transport a vast variety of molecules, including peptides, drugs, and ions [99]. Half transporters include two polypeptides, each having a transmembrane binding domain (TMD) and (nucleotide-binding domains) NBD to form a homo- or hetero-dimer. Full transporters are characterized as all four domains reside on a single polypeptide [97].
P-gp (also named ABCB1 or MDR1) is the first discovered and well-studied ABCB subfamily transporter that mediates MDR in cancer cells [100]. It is most expressed in the blood−brain barrier, liver, placenta, gallbladder, and endocrine tissues. In 2020, Nosol et al. revealed the protein structure of P-gp and found that inhibitors are bound in pairs and interact with structural features to block the function of P-gp [101]. As P-gp is the substrate of a broad range of antineoplastic agents, overexpressed P-gp has been demonstrated to lead to the development of drug resistance, including chemotherapeutic agents Vinca alkaloids (vinblastine and vincristine), Taxanes (paclitaxel and docetaxel), Anthracyclines (doxorubicin, daunorubicin, and epirubicin), and imatinib mesylate [102,103]. Meanwhile, a high-level expression of P-gp that causes cancer drug resistance has also been found in some tyrosine kinase inhibitors (Imatinib [104], GSK1070916 [105], and WYE354 [106]).
Altered expression levels of MDR1 have been detected in various tumors, such as neuroblastoma, rhabdomyosarcoma, and Wilm’s tumor. Neuroblastoma is the most common pediatric solid tumor, which accounts for 7–8% of childhood malignancies and 15% of all pediatric cancer deaths [107]. A study showed that the mRNA expression of MDR1 was increased in neuroblastoma patients with previous chemotherapy [108]. In addition, Qiu et al. [109] found that MDR1 hypermethylation expression can be associated with the pathogenesis and progression of neuroblastoma. MDR1 expression was observed to increase after chemotherapy in rhabdomyosarcoma and Wilm’s tumor [110]. These findings may be helpful to understand the role of MDR1 in pediatric malignancies regarding drug resistance and allow researchers to come up with strategies for therapeutic intervention.

5.2.2. MRPs

Multidrug resistance proteins (MRPs) include 9 transporters from 13 members in the ABCC subfamily due to their ability to mediate cancer MDR [111].
Although each of the MRPs have slight differences in structures and amino acid compositions, the mechanism of transport driven by ATP hydrolysis is much the same. Unlike P-gp, which extrudes mostly xenobiotics, MRPs account for the extruding of both endo- and xenobiotics, thus showing its crucial role in regulating MDR processes in cancer development [112]. The structure of MRP1 has shown a novel substrate recruitment mechanism in that substrates are recruited straight from the cytoplasm, whereas P-gp attaches substrates from the inner leaflet of the lipid bilayer [113]. MRPs are distributed in the human body in various tissues, including the blood−brain barrier, brain, lung, kidney, liver, etc. [111].
The expression of MRPs has been investigated in several pediatric malignant tumors as they are a vital factor causing cytotoxic drug resistance and chemotherapy failure. Abnormal MDR expressions have been observed in pediatric malignancies, such as ALL, neuroblastoma, rhabdomyosarcoma, Wilm’s tumor, and retinoblastoma [114]. After chemotherapy, MRP1 expression has been observed to be upregulated in neuroblastoma, hepatoblastoma, and rhabdomyosarcoma patients [110]. Increased expression of MRP2-6 and decreased expression of MRP1 and MRP10 have been observed in ALL patients, which are associated with high doses of three chemotherapies [115]. Henderson et al. found that the inhibition of MPR1 is associated with reduced neuroblastoma development in transgenic mice [116]. These studies indicate their role in the chemotherapeutic drug efflux of MPRs and cancer prognosis.

5.2.3. BCRP

The human BCRP has 665 amino acid residues with a molecular weight of 72 kDa. It is a half-transporter that is prominently expressed in various tissues, including, but not limited to, the brain, placenta, testis, liver, breast, and BBB [117]. A high overexpression of BCRP can be observed in different drug resistance cancer types, including in solid tumors and hematopoietic malignancies [118,119]. Although the clinical significance of BCRP-mediated drug resistance remains unclear, many studies have shown strong evidence to support that modulating the expression of BCRP could enhance drug sensitivity in chemotherapy [120,121].
BCRP was first identified in 1998, followed by expression studies to explore its potential role in chemoresistance [122]. The expression of the BCRP gene in childhood ALL has been observed at a low expression level [123]. However, compared with a diagnosis when the co-expression of BCRP and MDR1 was observed, a higher RNA level of BCRP was expressed at the relapsed/refractory state in acute myeloid leukemia (AML) [124]. Correlations between BCRP and MRPs have also been reported. The combined high expression of BCRP and MRP4 is correlated with reduced antileukemia drug methotrexate accumulation. Similarly, evaluated expression of the BCRP gene was found in primary neuroblastoma mitoxantrone-resistant cells [125]. These results underscore the potential value of BCRP as a predictor of chemoresistance drug efflux.
Essentially, MDR can arise because of altered targeted proteins or cell signaling pathways. Changes in cellular or non-cellular processes is also a significant factor of MDR. However, most encountered MDRs are related to drug efflux, which is mainly caused by ABC transporters. Once MDR transporters are overexpressed, the efflux of a chemo drug can increase. For example, the overexpression of P-gp, MRP1, and BCRP decreases the chemosensitivity of cancer cells by limiting exposure to anticancer drugs [126]. The overexpression of P-gp in cancer cells is associated with increased drug resistance to DOX and paclitaxel [127,128]. After anticancer agent treatment, the overexpression of P-gp has been found in acute myeloid leukemia [129]. Decreased chemosensitivity and a high expression of P-gp and BCRP were noticed in MDR patients with chronic lymphocytic leukemia, metastatic breast cancer, and multiple myeloma [130]. BCRP is regulated by proteins such as TGF-β1 and VEGFR-2 [131,132].

6. Tackling Strategies Regarding Drug Transporters

MDR is one of the barrier mechanisms against chemotherapy and thus is considered a key factor leading to the failure of chemotherapeutics. The overexpression of MDR1 that can enhance the efflux of cytotoxic agents is one of the targets to improve the effect of chemotherapeutics. One of the strategies is to reverse resistance mechanisms [133]. Several P MDR1 inhibitors have been investigated and shown successful MDR reversal on a drug-resistant prostate cancer cell line, without exhibiting a toxic potential [134]. Lei et al. [135] observed intracellular accumulation of paclitaxel and decreased drug efflux activity in the MDR1-knockout colorectal cancer cell line.
Furthermore, applying a nano-drug delivery system (NDDS) is a practical approach to enhance chemotherapy validity due to the targeted co-delivery, reduced sides affected, and long-time blood circulation achieved [136]. Curcumin has an antitumor activity and reverses the tumor MDR effect by regulating the MDR1 protein [137]. Degradable poly (lactic-co-glycolic acid) (PLGA) nanoparticles coloaded with curcumin and DOX could directly target cells or xenografted tumors and inhibit the growth of DOX-resistant esophageal carcinoma with a high biosafety [138]. NDDS-containing compounds have been developed that can inhibit MDR1, including tariquidar (XR9576), tetrandrine, verapamil, and cyclosporin A [139].
Therapy regimens can be different for treating children and adults as they have different drug-resistance profiles. The triple combination of fludarabine, ara-C, and G-CSF has been used in the treatment of childhood AML and caused an additive cell kill [140]. Tipifarnib can target the malignancies, such as leukemia, by activating RAS proteins (HRAS, KRAS, and NRAS) [141].

7. Special Considerations of Resistance in Pediatric Cancer Treatment

Pediatric malignancies have significant differences in their treatment compared with adult tumors, and thus require special considerations. Pan-cancer analyses have shown that pediatric cancers have comparatively lower mutation frequencies compared with adult cancers [142]. Epigenetic dysregulation, however, seems to be a particular factor in many types of pediatric cancers [143,144,145]. These genetic and non-genetic changes suggest therapeutic implications regarding chemotherapy.
Children with ALL have better prognoses and outcomes than adult patients with ALL. The 5-year OS rate is 87% for children aged 0–15 years, as opposed to 44% for adults aged 20–29 years [146]. A plethora of factors are responsible for the different outcomes, including socio-economic factors, resistance, disease heterogeneity, host responses, therapeutic treatment, etc. [147]. Resistance is one of the main factors leading to variables among ALL patients of different ages. For example, the activation of P-gp has a higher expression in adults [148]. In addition, the accumulated mutations in the p53 gene and lower methotrexate polyglutamate may also contribute to the differences in the responses of drug resistance mechanisms [148,149]. Genetic lesions of polycomb repressor complex 2 (PRC2) have been reported in pediatric T-cell ALL (T-ALL), which promotes mutations of the IL7R/JAK/STAT pathway. Poor prednisone response and persistent MRD have been connected to adult T-ALL patients with loss-of-function alterations of PRC2 [150]. Genescà et al. reported that a complex karyotype (≥3 cytogenetic alterations) in adult T-ALL was associated with a minimal residual disease (MRD) level, but no correlation regarding the prednisone response [151]. Protein tyrosine phosphatase nonreceptor type 2 (PTPN2) is a phosphatase suppressing a gene in T-cell ALL. Deletions of PTPN2 in pediatric patients were associated with a higher glucocorticoid response and improved survival in children, yet these trends were not found in adults [149]. Other studies have suggested that a single subclone with additional mutations confers resistance to therapy, although half of the leukemia patients had multiple subclonal mutations [152]. Because children are not simply small adults, the difference in outcomes across age categories should be considered in diagnosis and therapy.

8. Clinical Trials and Recommendations for Risk Surveillance

With typically more aggressive protocols being used in children than in adults, chemotherapy is the standard treatment in cancers such as ALL, AML, and Hodgkin lymphoma [153]. The number of clinical trials related to chemotherapy-induced side effects is on the rise.
Notably, VIPN is being studied in two clinical trials. A Phase 4 clinical trial (NCT02923388) is testing Vitamin B12 and vitamin B6 in ALL patients treated with VCR.
The results from a study of 102 patients showed that vitamin B6 and B12 significantly reduced the incidence, relative risk, and severity of VIPN. The amount needed to treat was encouragingly low, and vitamin B6 and B12 were recommended as promising neuroprotective agents against VIPN [154]. A clinical trial (NCT02796365) is currently underway to evaluate the effectiveness of exercise rehabilitation as a preventive measure to DIC. The studies are summarized in Table 3.
Significantly, four clinical trials are investigating or studying DIC. A Phase 2 clinical trial (NCT04166253) is testing vitamin D in breast cancer patients treated with DOX. A clinical trial (NCT02796365) is currently underway to evaluate the effectiveness of exercise rehabilitation as a preventive measure for DIC. The studies are summarized in Table 3.
PanCare is Pan-European Network for care of pediatric cancer survivors. The risk for significant and potentially life-threatening late effects can be identified by certain long-term follow-up projects such as PanCareSurPass and PanCareFollowUp. Based on the reports from 10 countries, ototoxicity following platinum-based chemotherapy has been evaluated regarding the quality of evidence. The ototoxicity surveillance recommendations for pediatric cancer survivors’ future studies should focus on the evaluation of otoprotectants and the identification of optimal threshold doses to prevent ototoxicity [155]. In addition, candidate genetic markers are useful for identifying childhood cancer patients at risk of severe late effects, such as SLC22A2, which detects those at risk of platinum-induced hearing loss [156]. Moreover, adequate knowledge of cancer history, subsequent treatment exposure, and potential risks of late effects are needed to enhance survivors’ health and self-management skills. Accessible and reliable information is essential to increase awareness about late effects, which is necessary for providing personal recommendations for surveillance and prevention [157].

9. Conclusions

Together with the significant advancements in target therapy and immunotherapy, chemotherapy remains the primary treatment option for childhood cancer patients in most low- and middle-income countries. Recognizing the absolute validity of chemo drugs is crucial as they have shown to be highly effective for obtaining long-term survival in childhood cancers. Until now, in total, 16 chemo drugs have been approved by the FDA for pediatric chemotherapy. Unlike adult cancer patients, children diagnosed with cancer are less tolerant of chemotherapy. The numerous adverse effects and occurrence of MDR have become a significant obstacle in pediatric chemotherapy. It is crucial to alleviate severe side effects such as VIPN and DIC. Meanwhile, a comprehensive understanding of MDR and its reversal mechanism is essential. To improve follow-up care quality, the electronic document summarizes the clinical history of childhood/adolescent cancer survivors, including treatments received, and provides personalized follow-up and screening recommendations. By overcoming the challenges of traditional chemotherapy, we will hopefully be able to improve both the survival rate and overall quality of life of pediatric cancer patients.

Author Contributions

L.B. and Y.W. wrote the original draft of the manuscript; Y.L. was responsible for preparing figures and tables; J.N.D.W. and Z.H. reviewed and edited the manuscript; Z.-S.C. supervised the paper. All authors have read and agreed to the published version of the manuscript.

Funding

Science and Technology Plan of Jiangxi Provincial Health Commission (no: 202210918); the Guiding Science and Technology Programme of Ganzhou city (no: GZ2022ZSF231).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ward, E.; DeSantis, C.; Robbins, A.; Kohler, B.; Jemal, A. Childhood and adolescent cancer statistics, 2014. CA Cancer J. Clin. 2014, 64, 83–103. [Google Scholar] [CrossRef] [PubMed]
  2. McEachron, T.A.; Helman, L.J. Recent Advances in Pediatric Cancer Research. Cancer Res. 2021, 81, 5783–5799. [Google Scholar] [CrossRef]
  3. Shah, N. Dodging the bullet: Therapeutic resistance mechanisms in pediatric cancers. Cancer Drug Resist. 2019, 2, 428–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  5. GBD 2017 Childhood Cancer Collaborators. The global burden of childhood and adolescent cancer in 2017: An analysis of the Global Burden of Disease Study 2017. Lancet Oncol. 2019, 20, 1211–1225. [Google Scholar] [CrossRef] [Green Version]
  6. Rodriguez-Galindo, C.; Friedrich, P.; Alcasabas, P.; Antillon, F.; Banavali, S.; Castillo, L.; Israels, T.; Jeha, S.; Harif, M.; Sullivan, M.J.; et al. Toward the Cure of All Children with Cancer Through Collaborative Efforts: Pediatric Oncology as a Global Challenge. J. Clin. Oncol. 2015, 33, 3065–3073. [Google Scholar] [CrossRef]
  7. Pramesh, C.S.; Badwe, R.A.; Bhoo-Pathy, N.; Booth, C.M.; Chinnaswamy, G.; Dare, A.J.; de Andrade, V.P.; Hunter, D.J.; Gopal, S.; Gospodarowicz, M.; et al. Priorities for cancer research in low- and middle-income countries: A global perspective. Nat. Med. 2022, 28, 649–657. [Google Scholar] [CrossRef]
  8. Selmouni, F.; Zidouh, A.; Belakhel, L.; Sauvaget, C.; Bennani, M.; Khazraji, Y.C.; Benider, A.; Wild, C.P.; Bekkali, R.; Fadhil, I.; et al. Tackling cancer burden in low-income and middle-income countries: Morocco as an exemplar. Lancet Oncol. 2018, 19, e93–e101. [Google Scholar] [CrossRef]
  9. Patel, A.; Goldstein, D.A.; Tannock, I.F. Improving access to immunotherapy in low- and middle-income countries. Ann. Oncol. 2022, 33, 360–361. [Google Scholar] [CrossRef]
  10. Chantada, G.; Lam, C.G.; Howard, S.C. Optimizing outcomes for children with non-Hodgkin lymphoma in low- and middle-income countries by early correct diagnosis, reducing toxic death and preventing abandonment. Br. J. Haematol. 2019, 185, 1125–1135. [Google Scholar] [CrossRef]
  11. Yalcin-Ozkat, G. Molecular Modeling Strategies of Cancer Multidrug Resistance. Drug Resist. Updat. 2021, 59, 100789. [Google Scholar] [CrossRef] [PubMed]
  12. Kattner, P.; Strobel, H.; Khoshnevis, N.; Grunert, M.; Bartholomae, S.; Pruss, M.; Fitzel, R.; Halatsch, M.E.; Schilberg, K.; Siegelin, M.D.; et al. Compare and contrast: Pediatric cancer versus adult malignancies. Cancer Metastasis Rev. 2019, 38, 673–682. [Google Scholar] [CrossRef] [PubMed]
  13. Nonnenmacher, L.; Hasslacher, S.; Zimmermann, J.; Karpel-Massler, G.; La Ferla-Bruhl, K.; Barry, S.E.; Burster, T.; Siegelin, M.D.; Bruhl, O.; Halatsch, M.E.; et al. Cell Death Induction in Cancer Therapy—Past, Present, and Future. Crit. Rev. Oncog. 2016, 21, 253–267. [Google Scholar] [CrossRef] [PubMed]
  14. Abbott, M.; Ustoyev, Y. Cancer and the Immune System: The History and Background of Immunotherapy. Semin. Oncol. Nurs. 2019, 35, 150923. [Google Scholar] [CrossRef]
  15. Citrin, D.E. Recent Developments in Radiotherapy. N. Engl. J. Med. 2017, 377, 1065–1075. [Google Scholar] [CrossRef]
  16. Fletcher, J.I.; Ziegler, D.S.; Trahair, T.N.; Marshall, G.M.; Haber, M.; Norris, M.D. Too many targets, not enough patients: Rethinking neuroblastoma clinical trials. Nat. Rev. Cancer 2018, 18, 389–400. [Google Scholar] [CrossRef]
  17. Malczewska, M.; Kosmider, K.; Bednarz, K.; Ostapinska, K.; Lejman, M.; Zawitkowska, J. Recent Advances in Treatment Options for Childhood Acute Lymphoblastic Leukemia. Cancers 2022, 14, 2021. [Google Scholar] [CrossRef]
  18. Printezi, M.I.; Kilgallen, A.B.; Bond, M.J.G.; Stibler, U.; Putker, M.; Teske, A.J.; Cramer, M.J.; Punt, C.J.A.; Sluijter, J.P.G.; Huitema, A.D.R.; et al. Toxicity and efficacy of chronomodulated chemotherapy: A systematic review. Lancet Oncol. 2022, 23, e129–e143. [Google Scholar] [CrossRef]
  19. Massimino, M.; Biassoni, V. Use of high-dose chemotherapy in front-line therapy of childhood malignant glioma. Expert Rev. Anticancer. Ther. 2006, 6, 709–717. [Google Scholar] [CrossRef]
  20. Mir, M.A.; Sofi, S.; Qayoom, H. Chapter 4—Conventional adjuvant chemotherapy in combination with surgery, radiotherapy, and other specific targets. In Combinational Therapy in Triple Negative Breast Cancer; Mir, M., Ed.; Academic Press: Cambridge, MA, USA, 2022; pp. 95–120. [Google Scholar]
  21. Principe, D.R.; Kamath, S.D.; Korc, M.; Munshi, H.G. The immune modifying effects of chemotherapy and advances in chemo-immunotherapy. Pharmacol. Ther. 2022, 236, 108111. [Google Scholar] [CrossRef]
  22. Nishiwaki, S.; Ando, Y. Gap between pediatric and adult approvals of molecular targeted drugs. Sci. Rep. 2020, 10, 17145. [Google Scholar] [CrossRef]
  23. Xu, F.L.; Guan, X.M.; Wen, X.H.; Shen, Y.L.; Xiao, J.W.; Guo, Y.X.; Deng, M.Y.; Yu, J. Serious adverse events associated with chemotherapy in children with acute lymphoblastic leukemia. Zhongguo Dang Dai Er Ke Za Zhi 2020, 22, 828–833. [Google Scholar] [PubMed]
  24. Barone, A.; Casey, D.; McKee, A.E.; Reaman, G. Cancer drugs approved for use in children: Impact of legislative initiatives and future opportunities. Pediatr. Blood Cancer 2019, 66, e27809. [Google Scholar] [CrossRef] [PubMed]
  25. Wu, C.Y.; Li, G.T.; Chu, C.C.; Guo, H.L.; Fang, W.R.; Li, T.; Wang, Y.R.; Xu, J.; Hu, Y.H.; Zhou, L.; et al. Proactive therapeutic drug monitoring of vincristine in pediatric and adult cancer patients: Current supporting evidence and future efforts. Arch. Toxicol. 2022, 97, 377–392. [Google Scholar] [CrossRef] [PubMed]
  26. Bhagat, A.; Kleinerman, E.S. Anthracycline-Induced Cardiotoxicity: Causes, Mechanisms, and Prevention. Adv. Exp. Med. Biol. 2020, 1257, 181–192. [Google Scholar] [PubMed]
  27. Mancilla, T.R.; Iskra, B.; Aune, G.J. Doxorubicin-Induced Cardiomyopathy in Children. Compr. Physiol. 2019, 9, 905–931. [Google Scholar] [PubMed]
  28. van de Velde, M.E.; Kaspers, G.L.; Abbink, F.C.H.; Wilhelm, A.J.; Ket, J.C.F.; van den Berg, M.H. Vincristine-induced peripheral neuropathy in children with cancer: A systematic review. Crit. Rev. Oncol. Hematol. 2017, 114, 114–130. [Google Scholar] [CrossRef]
  29. Rivankar, S. An overview of doxorubicin formulations in cancer therapy. J. Cancer Res. Ther. 2014, 10, 853–858. [Google Scholar] [CrossRef]
  30. Garbayo, E.; Pascual-Gil, S.; Rodriguez-Nogales, C.; Saludas, L.; Estella-Hermoso de Mendoza, A.; Blanco-Prieto, M.J. Nanomedicine and drug delivery systems in cancer and regenerative medicine. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 12, e1637. [Google Scholar] [CrossRef]
  31. Liu, C.; Ma, X.; Zhuang, J.; Liu, L.; Sun, C. Cardiotoxicity of doxorubicin-based cancer treatment: What is the protective cognition that phytochemicals provide us? Pharmacol. Res. 2020, 160, 105062. [Google Scholar] [CrossRef]
  32. Kalyanaraman, B. Teaching the basics of the mechanism of doxorubicin-induced cardiotoxicity: Have we been barking up the wrong tree? Redox Biol. 2020, 29, 101394. [Google Scholar] [CrossRef] [PubMed]
  33. Magdy, T.; Jiang, Z.; Jouni, M.; Fonoudi, H.; Lyra-Leite, D.; Jung, G.; Romero-Tejeda, M.; Kuo, H.H.; Fetterman, K.A.; Gharib, M.; et al. RARG variant predictive of doxorubicin-induced cardiotoxicity identifies a cardioprotective therapy. Cell Stem Cell 2021, 28, 2076–2089.e7. [Google Scholar] [CrossRef] [PubMed]
  34. Magdy, T.; Jouni, M.; Kuo, H.H.; Weddle, C.J.; Lyra-Leite, D.; Fonoudi, H.; Romero-Tejeda, M.; Gharib, M.; Javed, H.; Fajardo, G.; et al. Identification of Drug Transporter Genomic Variants and Inhibitors That Protect Against Doxorubicin-Induced Cardiotoxicity. Circulation 2022, 145, 279–294. [Google Scholar] [CrossRef] [PubMed]
  35. Lim, G.B. Circular RNA prevents doxorubicin-induced cardiotoxicity. Nat. Rev. Cardiol. 2022, 19, 574. [Google Scholar] [CrossRef]
  36. Lu, D.; Chatterjee, S.; Xiao, K.; Riedel, I.; Huang, C.K.; Costa, A.; Cushman, S.; Neufeldt, D.; Rode, L.; Schmidt, A.; et al. A circular RNA derived from the insulin receptor locus protects against doxorubicin-induced cardiotoxicity. Eur. Heart J. 2022, 43, 4496–4511. [Google Scholar] [CrossRef]
  37. Han, D.; Wang, Y.; Wang, Y.; Dai, X.; Zhou, T.; Chen, J.; Tao, B.; Zhang, J.; Cao, F. The Tumor-Suppressive Human Circular RNA CircITCH Sponges miR-330-5p to Ameliorate Doxorubicin-Induced Cardiotoxicity Through Upregulating SIRT6, Survivin, and SERCA2a. Circ. Res. 2020, 127, e108–e125. [Google Scholar] [CrossRef]
  38. Wang, Y.; Lu, X.; Wang, X.; Qiu, Q.; Zhu, P.; Ma, L.; Ma, X.; Herrmann, J.; Lin, X.; Wang, W.; et al. atg7-Based Autophagy Activation Reverses Doxorubicin-Induced Cardiotoxicity. Circ. Res. 2021, 129, e166–e182. [Google Scholar] [CrossRef]
  39. Hu, C.; Zhang, X.; Song, P.; Yuan, Y.P.; Kong, C.Y.; Wu, H.M.; Xu, S.C.; Ma, Z.G.; Tang, Q.Z. Meteorin-like protein attenuates doxorubicin-induced cardiotoxicity via activating cAMP/PKA/SIRT1 pathway. Redox Biol. 2020, 37, 101747. [Google Scholar] [CrossRef]
  40. Wang, A.J.; Tang, Y.; Zhang, J.; Wang, B.J.; Xiao, M.; Lu, G.; Li, J.; Liu, Q.; Guo, Y.; Gu, J. Cardiac SIRT1 ameliorates doxorubicin-induced cardiotoxicity by targeting sestrin 2. Redox Biol. 2022, 52, 102310. [Google Scholar] [CrossRef]
  41. Wu, Y.Z.; Zhang, L.; Wu, Z.X.; Shan, T.T.; Xiong, C. Berberine Ameliorates Doxorubicin-Induced Cardiotoxicity via a SIRT1/p66Shc-Mediated Pathway. Oxid. Med. Cell. Longev. 2019, 2019, 2150394. [Google Scholar] [CrossRef] [Green Version]
  42. Wu, X.; Wang, L.; Wang, K.; Li, J.; Chen, R.; Wu, X.; Ni, G.; Liu, C.; Das, S.; Sluijter, J.P.G.; et al. ADAR2 increases in exercised heart and protects against myocardial infarction and doxorubicin-induced cardiotoxicity. Mol. Ther. 2022, 30, 400–414. [Google Scholar] [CrossRef]
  43. Chen, D.; Yu, W.; Zhong, C.; Hong, Q.; Huang, G.; Que, D.; Wang, Y.; Yang, Y.; Rui, B.; Zhuang, Z.; et al. Elabela ameliorates doxorubicin-induced cardiotoxicity by promoting autophagic flux through TFEB pathway. Pharmacol. Res. 2022, 178, 106186. [Google Scholar] [CrossRef] [PubMed]
  44. Russo, M.; Guida, F.; Paparo, L.; Trinchese, G.; Aitoro, R.; Avagliano, C.; Fiordelisi, A.; Napolitano, F.; Mercurio, V.; Sala, V.; et al. The novel butyrate derivative phenylalanine-butyramide protects from doxorubicin-induced cardiotoxicity. Eur. J. Heart Fail. 2019, 21, 519–528. [Google Scholar] [CrossRef] [PubMed]
  45. Ye, B.; Shi, X.; Xu, J.; Dai, S.; Xu, J.; Fan, X.; Han, B.; Han, J. Gasdermin D mediates doxorubicin-induced cardiomyocyte pyroptosis and cardiotoxicity via directly binding to doxorubicin and changes in mitochondrial damage. Transl. Res. 2022, 248, 36–50. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, G.; Song, M.; Hoang, D.H.; Tran, Q.H.; Choe, W.; Kang, I.; Kim, S.S.; Ha, J. Melatonin prevents doxorubicin-induced cardiotoxicity through suppression of AMPKalpha2-dependent mitochondrial damage. Exp. Mol. Med. 2020, 52, 2055–2068. [Google Scholar] [CrossRef]
  47. Efentakis, P.; Varela, A.; Chavdoula, E.; Sigala, F.; Sanoudou, D.; Tenta, R.; Gioti, K.; Kostomitsopoulos, N.; Papapetropoulos, A.; Tasouli, A.; et al. Levosimendan prevents doxorubicin-induced cardiotoxicity in time- and dose-dependent manner: Implications for inotropy. Cardiovasc. Res. 2020, 116, 576–591. [Google Scholar] [CrossRef]
  48. Ding, M.; Shi, R.; Fu, F.; Li, M.; De, D.; Du, Y.; Li, Z. Paeonol protects against doxorubicin-induced cardiotoxicity by promoting Mfn2-mediated mitochondrial fusion through activating the PKCepsilon-Stat3 pathway. J. Adv. Res. 2022; in press. [Google Scholar]
  49. Zhang, Y.; Ni, L.; Lin, B.; Hu, L.; Lin, Z.; Yang, J.; Wang, J.; Ma, H.; Liu, Y.; Yang, J.; et al. SNX17 protects the heart from doxorubicin-induced cardiotoxicity by modulating LMOD2 degradation. Pharmacol. Res. 2021, 169, 105642. [Google Scholar] [CrossRef]
  50. Pan, J.A.; Zhang, H.; Lin, H.; Gao, L.; Zhang, H.L.; Zhang, J.F.; Wang, C.Q.; Gu, J. Irisin ameliorates doxorubicin-induced cardiac perivascular fibrosis through inhibiting endothelial-to-mesenchymal transition by regulating ROS accumulation and autophagy disorder in endothelial cells. Redox Biol. 2021, 46, 102120. [Google Scholar] [CrossRef]
  51. Wang, P.; Wang, M.; Hu, Y.; Chen, J.; Cao, Y.; Liu, C.; Wu, Z.; Shen, J.; Lu, J.; Liu, P. Isorhapontigenin protects against doxorubicin-induced cardiotoxicity via increasing YAP1 expression. Acta Pharm. Sin. B 2021, 11, 680–693. [Google Scholar] [CrossRef]
  52. Liang, X.; Wang, S.; Wang, L.; Ceylan, A.F.; Ren, J.; Zhang, Y. Mitophagy inhibitor liensinine suppresses doxorubicin-induced cardiotoxicity through inhibition of Drp1-mediated maladaptive mitochondrial fission. Pharmacol. Res. 2020, 157, 104846. [Google Scholar] [CrossRef]
  53. Martino, E.; Casamassima, G.; Castiglione, S.; Cellupica, E.; Pantalone, S.; Papagni, F.; Rui, M.; Siciliano, A.M.; Collina, S. Vinca alkaloids and analogues as anti-cancer agents: Looking back, peering ahead. Bioorg. Med. Chem. Lett. 2018, 28, 2816–2826. [Google Scholar] [CrossRef] [PubMed]
  54. Keenan, B.; Finol-Urdaneta, R.K.; Hope, A.; Bremner, J.B.; Kavallaris, M.; Lucena-Agell, D.; Oliva, M.A.; Diaz, J.F.; Vine, K.L. N-alkylisatin-based microtubule destabilizers bind to the colchicine site on tubulin and retain efficacy in drug resistant acute lymphoblastic leukemia cell lines with less in vitro neurotoxicity. Cancer Cell Int. 2020, 20, 170. [Google Scholar] [CrossRef] [PubMed]
  55. Harris, C.M.; Blanchaert, R.H. Bilateral recurrent laryngeal nerve palsy resulting from treatment with vincristine. J. Oral Maxillofac. Surg. 2006, 64, 738–739. [Google Scholar] [CrossRef] [PubMed]
  56. Nama, N.; Barker, M.K.; Kwan, C.; Sabarre, C.; Solimano, V.; Rankin, A.; Raabe, J.; Ross, C.J.; Carleton, B.; Zwicker, J.G.; et al. Vincristine-induced peripheral neurotoxicity: A prospective cohort. Pediatr. Hematol. Oncol. 2020, 37, 15–28. [Google Scholar] [CrossRef] [PubMed]
  57. Barnett, S.; Hellmann, F.; Parke, E.; Makin, G.; Tweddle, D.A.; Osborne, C.; Hempel, G.; Veal, G.J. Vincristine dosing, drug exposure and therapeutic drug monitoring in neonate and infant cancer patients. Eur. J. Cancer 2022, 164, 127–136. [Google Scholar] [CrossRef]
  58. Uittenboogaard, A.; Neutel, C.L.G.; Ket, J.C.F.; Njuguna, F.; Huitema, A.D.R.; Kaspers, G.J.L.; van de Velde, M.E. Pharmacogenomics of Vincristine-Induced Peripheral Neuropathy in Children with Cancer: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 612. [Google Scholar] [CrossRef]
  59. Triarico, S.; Romano, A.; Attina, G.; Capozza, M.A.; Maurizi, P.; Mastrangelo, S.; Ruggiero, A. Vincristine-Induced Peripheral Neuropathy (VIPN) in Pediatric Tumors: Mechanisms, Risk Factors, Strategies of Prevention and Treatment. Int. J. Mol. Sci. 2021, 22, 4112. [Google Scholar] [CrossRef]
  60. Smolik, S.; Arland, L.; Hensley, M.A.; Schissel, D.; Shepperd, B.; Thomas, K.; Rodgers, C. Assessment Tools for Peripheral Neuropathy in Pediatric Oncology: A Systematic Review from the Children’s Oncology Group. J. Pediatr. Oncol. Nurs. 2018, 35, 267–275. [Google Scholar] [CrossRef]
  61. Al-Mahayri, Z.N.; AlAhmad, M.M.; Ali, B.R. Long-Term Effects of Pediatric Acute Lymphoblastic Leukemia Chemotherapy: Can Recent Findings Inform Old Strategies? Front. Oncol. 2021, 11, 710163. [Google Scholar] [CrossRef]
  62. Smith, E.M.L.; Kuisell, C.; Cho, Y.; Kanzawa-Lee, G.A.; Gilchrist, L.S.; Park, S.B.; Scott, M.R.; Alberti, P.; Toxic Neuropathy Consortium of the Peripheral Nerve Society. Characteristics and patterns of pediatric chemotherapy-induced peripheral neuropathy: A systematic review. Cancer Treat. Res. Commun. 2021, 28, 100420. [Google Scholar] [CrossRef]
  63. Madsen, M.L.; Due, H.; Ejskjaer, N.; Jensen, P.; Madsen, J.; Dybkaer, K. Aspects of vincristine-induced neuropathy in hematologic malignancies: A systematic review. Cancer Chemother. Pharmacol. 2019, 84, 471–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Tay, C.G.; Lee, V.W.M.; Ong, L.C.; Goh, K.J.; Ariffin, H.; Fong, C.Y. Vincristine-induced peripheral neuropathy in survivors of childhood acute lymphoblastic leukaemia. Pediatr. Blood Cancer 2017, 64, e26471. [Google Scholar] [CrossRef] [PubMed]
  65. Adil, M.K.; Ali, Z.; Arshad, U.; Fawad, U. Vincristine induced neurotoxicity in children who underwent chemotherapy for acute lymphoblastic leukemia and Wilm’s tumor. Pak. J. Med. Sci. 2021, 37, 1331–1334. [Google Scholar] [CrossRef] [PubMed]
  66. Li, G.Z.; Hu, Y.H.; Li, D.Y.; Zhang, Y.; Guo, H.L.; Li, Y.M.; Chen, F.; Xu, J. Vincristine-induced peripheral neuropathy: A mini-review. Neurotoxicology 2020, 81, 161–171. [Google Scholar] [CrossRef] [PubMed]
  67. Yang, Q.Y.; Hu, Y.H.; Guo, H.L.; Xia, Y.; Zhang, Y.; Fang, W.R.; Li, Y.M.; Xu, J.; Chen, F.; Wang, Y.R.; et al. Vincristine-Induced Peripheral Neuropathy in Childhood Acute Lymphoblastic Leukemia: Genetic Variation as a Potential Risk Factor. Front. Pharmacol. 2021, 12, 771487. [Google Scholar] [CrossRef]
  68. Pozzi, E.; Fumagalli, G.; Chiorazzi, A.; Canta, A.; Cavaletti, G. Genetic factors influencing the development of vincristine-induced neurotoxicity. Expert Opin. Drug Metab. Toxicol. 2021, 17, 215–226. [Google Scholar] [CrossRef]
  69. Stock, W.; Diouf, B.; Crews, K.R.; Pei, D.; Cheng, C.; Laumann, K.; Mandrekar, S.J.; Luger, S.; Advani, A.; Stone, R.M.; et al. An Inherited Genetic Variant in CEP72 Promoter Predisposes to Vincristine-Induced Peripheral Neuropathy in Adults with Acute Lymphoblastic Leukemia. Clin. Pharmacol. Ther. 2017, 101, 391–395. [Google Scholar] [CrossRef] [Green Version]
  70. Egbelakin, A.; Ferguson, M.J.; MacGill, E.A.; Lehmann, A.S.; Topletz, A.R.; Quinney, S.K.; Li, L.; McCammack, K.C.; Hall, S.D.; Renbarger, J.L. Increased risk of vincristine neurotoxicity associated with low CYP3A5 expression genotype in children with acute lymphoblastic leukemia. Pediatr. Blood Cancer 2011, 56, 361–367. [Google Scholar] [CrossRef] [Green Version]
  71. Zgheib, N.K.; Ghanem, K.M.; Tamim, H.; Aridi, C.; Shahine, R.; Tarek, N.; Saab, R.; Abboud, M.R.; El-Solh, H.; Muwakkit, S.A. Genetic polymorphisms in candidate genes are not associated with increased vincristine-related peripheral neuropathy in Arab children treated for acute childhood leukemia: A single institution study. Pharmacogenet. Genom. 2018, 28, 189–195. [Google Scholar] [CrossRef] [PubMed]
  72. Lopez-Lopez, E.; Gutierrez-Camino, A.; Astigarraga, I.; Navajas, A.; Echebarria-Barona, A.; Garcia-Miguel, P.; Garcia de Andoin, N.; Lobo, C.; Guerra-Merino, I.; Martin-Guerrero, I.; et al. Vincristine pharmacokinetics pathway and neurotoxicity during early phases of treatment in pediatric acute lymphoblastic leukemia. Pharmacogenomics 2016, 17, 731–741. [Google Scholar] [CrossRef]
  73. Zhou, L.; Ao, L.; Yan, Y.; Li, C.; Li, W.; Ye, A.; Liu, J.; Hu, Y.; Fang, W.; Li, Y. Levo-corydalmine Attenuates Vincristine-Induced Neuropathic Pain in Mice by Upregulating the Nrf2/HO-1/CO Pathway to Inhibit Connexin 43 Expression. Neurotherapeutics 2020, 17, 340–355. [Google Scholar] [CrossRef]
  74. Zhou, L.; Hu, Y.; Li, C.; Yan, Y.; Ao, L.; Yu, B.; Fang, W.; Liu, J.; Li, Y. Levo-corydalmine alleviates vincristine-induced neuropathic pain in mice by inhibiting an NF-kappa B-dependent CXCL1/CXCR2 signaling pathway. Neuropharmacology 2018, 135, 34–47. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, X.J.; Wang, L.; Song, X.Y. Mitoquinone alleviates vincristine-induced neuropathic pain through inhibiting oxidative stress and apoptosis via the improvement of mitochondrial dysfunction. Biomed. Pharmacother. 2020, 125, 110003. [Google Scholar] [CrossRef]
  76. Xie, H.; Chen, Y.; Du, K.; Wu, W.; Feng, X. Puerarin alleviates vincristine-induced neuropathic pain and neuroinflammation via inhibition of nuclear factor-kappaB and activation of the TGF-beta/Smad pathway in rats. Int. Immunopharmacol. 2020, 89, 107060. [Google Scholar] [CrossRef] [PubMed]
  77. Liang, Z.J.; Tan, J.; Tang, L.; Xie, Z.B.; Chen, G.J.; Liu, G.J.; Yuan, L.; Wang, K.X.; Ding, H.P.; Qiu, H.; et al. NGF monoclonal antibody DS002 alleviates chemotherapy-induced peripheral neuropathy in rats. Acta Pharmacol. Sin. 2022, 43, 2841–2847. [Google Scholar] [CrossRef] [PubMed]
  78. Starobova, H.; Mueller, A.; Allavena, R.; Lohman, R.J.; Sweet, M.J.; Vetter, I. Minocycline Prevents the Development of Mechanical Allodynia in Mouse Models of Vincristine-Induced Peripheral Neuropathy. Front. Neurosci. 2019, 13, 653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Singh, G.; Singh, A.; Singh, P.; Bhatti, R. Bergapten Ameliorates Vincristine-Induced Peripheral Neuropathy by Inhibition of Inflammatory Cytokines and NFkappaB Signaling. ACS Chem. Neurosci. 2019, 10, 3008–3017. [Google Scholar] [CrossRef]
  80. Vasan, N.; Baselga, J.; Hyman, D.M. A view on drug resistance in cancer. Nature 2019, 575, 299–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
  82. Zhang, C.; Liu, X.; Jin, S.; Chen, Y.; Guo, R. Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol. Cancer 2022, 21, 47. [Google Scholar] [CrossRef]
  83. Singh, M.S.; Tammam, S.N.; Shetab Boushehri, M.A.; Lamprecht, A. MDR in cancer: Addressing the underlying cellular alterations with the use of nanocarriers. Pharmacol. Res. 2017, 126, 2–30. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, Q.; Yang, Z.; Nie, Y.; Shi, Y.; Fan, D. Multi-drug resistance in cancer chemotherapeutics: Mechanisms and lab approaches. Cancer Lett. 2014, 347, 159–166. [Google Scholar] [CrossRef]
  85. Lin, L.; Yee, S.W.; Kim, R.B.; Giacomini, K.M. SLC transporters as therapeutic targets: Emerging opportunities. Nat. Rev. Drug Discov. 2015, 14, 543–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Girardi, E.; Cesar-Razquin, A.; Lindinger, S.; Papakostas, K.; Konecka, J.; Hemmerich, J.; Kickinger, S.; Kartnig, F.; Gurtl, B.; Klavins, K.; et al. A widespread role for SLC transmembrane transporters in resistance to cytotoxic drugs. Nat. Chem. Biol. 2020, 16, 469–478. [Google Scholar] [CrossRef] [PubMed]
  87. Fang, X.; Liu, Y.; Xiao, W.; Zhao, N.; Zhu, C.; Yu, D.; Zhao, Y. Prognostic SLC family genes promote cell proliferation, migration, and invasion in hepatocellular carcinoma. Acta Biochim. Biophys. Sin. 2021, 53, 1065–1075. [Google Scholar] [CrossRef] [PubMed]
  88. Hu, Y.H.; Zhou, L.; Wang, S.S.; Jing, X.; Guo, H.L.; Sun, F.; Zhang, Y.; Chen, F.; Xu, J.; Ji, X. Methotrexate Disposition in Pediatric Patients with Acute Lymphoblastic Leukemia: What Have We Learnt from the Genetic Variants of Drug Transporters. Curr. Pharm. Des. 2019, 25, 627–634. [Google Scholar] [CrossRef] [PubMed]
  89. Robey, R.W.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer 2018, 18, 452–464. [Google Scholar] [CrossRef] [PubMed]
  90. Fletcher, J.I.; Haber, M.; Henderson, M.J.; Norris, M.D. ABC transporters in cancer: More than just drug efflux pumps. Nat. Rev. Cancer 2010, 10, 147–156. [Google Scholar] [CrossRef]
  91. Muriithi, W.; Macharia, L.W.; Heming, C.P.; Echevarria, J.L.; Nyachieo, A.; Filho, P.N.; Neto, V.M. ABC transporters and the hallmarks of cancer: Roles in cancer aggressiveness beyond multidrug resistance. Cancer Biol. Med. 2020, 17, 253–269. [Google Scholar] [CrossRef]
  92. Domenichini, A.; Adamska, A.; Falasca, M. ABC transporters as cancer drivers: Potential functions in cancer development. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 52–60. [Google Scholar] [CrossRef]
  93. Pasello, M.; Giudice, A.M.; Scotlandi, K. The ABC subfamily A transporters: Multifaceted players with incipient potentialities in cancer. Semin. Cancer Biol. 2020, 60, 57–71. [Google Scholar] [PubMed]
  94. Cui, Q.; Yang, Y.; Ji, N.; Wang, J.Q.; Ren, L.; Yang, D.H.; Chen, Z.S. Gaseous signaling molecules and their application in resistant cancer treatment: From invisible to visible. Future Med. Chem. 2019, 11, 323–336. [Google Scholar] [CrossRef] [PubMed]
  95. Nobili, S.; Lapucci, A.; Landini, I.; Coronnello, M.; Roviello, G.; Mini, E. Role of ATP-binding cassette transporters in cancer initiation and progression. Semin. Cancer Biol. 2020, 60, 72–95. [Google Scholar] [PubMed]
  96. Juan-Carlos, P.M.; Perla-Lidia, P.P.; Stephanie-Talia, M.M.; Monica-Griselda, A.M.; Luz-Maria, T.E. ABC transporter superfamily. An updated overview, relevance in cancer multidrug resistance and perspectives with personalized medicine. Mol. Biol. Rep. 2021, 48, 1883–1901. [Google Scholar]
  97. Wang, J.Q.; Wu, Z.X.; Yang, Y.; Teng, Q.X.; Li, Y.D.; Lei, Z.N.; Jani, K.A.; Kaushal, N.; Chen, Z.S. ATP-binding cassette (ABC) transporters in cancer: A review of recent updates. J. Evid. Based Med. 2021, 14, 232–256. [Google Scholar]
  98. Kadioglu, O.; Saeed, M.E.M.; Munder, M.; Spuller, A.; Greten, H.J.; Efferth, T. Effect of ABC transporter expression and mutational status on survival rates of cancer patients. Biomed. Pharmacother. 2020, 131, 110718. [Google Scholar]
  99. Szollosi, D.; Rose-Sperling, D.; Hellmich, U.A.; Stockner, T. Comparison of mechanistic transport cycle models of ABC exporters. Biochim. Biophys. Acta Biomembr. 2018, 1860, 818–832. [Google Scholar]
  100. Sarkadi, B.; Homolya, L.; Szakacs, G.; Varadi, A. Human multidrug resistance ABCB and ABCG transporters: Participation in a chemoimmunity defense system. Physiol. Rev. 2006, 86, 1179–1236. [Google Scholar] [CrossRef]
  101. Nosol, K.; Romane, K.; Irobalieva, R.N.; Alam, A.; Kowal, J.; Fujita, N.; Locher, K.P. Cryo-EM structures reveal distinct mechanisms of inhibition of the human multidrug transporter ABCB1. Proc. Natl. Acad. Sci. USA 2020, 117, 26245–26253. [Google Scholar] [CrossRef]
  102. Szakacs, G.; Paterson, J.K.; Ludwig, J.A.; Booth-Genthe, C.; Gottesman, M.M. Targeting multidrug resistance in cancer. Nat. Rev. Drug Discov. 2006, 5, 219–234. [Google Scholar]
  103. Choudhuri, S.; Klaassen, C.D. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP) efflux transporters. Int. J. Toxicol. 2006, 25, 231–259. [Google Scholar] [PubMed]
  104. Rajamani, B.M.; Benjamin, E.S.B.; Abraham, A.; Ganesan, S.; Lakshmi, K.M.; Anandan, S.; Karathedath, S.; Varatharajan, S.; Mohanan, E.; Janet, N.B.; et al. Plasma imatinib levels and ABCB1 polymorphism influences early molecular response and failure-free survival in newly diagnosed chronic phase CML patients. Sci. Rep. 2020, 10, 20640. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, Z.X.; Yang, Y.; Wang, J.Q.; Zhou, W.M.; Chen, J.; Fu, Y.G.; Patel, K.; Chen, Z.S.; Zhang, J.Y. Elevated ABCB1 Expression Confers Acquired Resistance to Aurora Kinase Inhibitor GSK-1070916 in Cancer Cells. Front. Pharmacol. 2020, 11, 615824. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, J.; Yang, D.H.; Yang, Y.; Wang, J.Q.; Cai, C.Y.; Lei, Z.N.; Teng, Q.X.; Wu, Z.X.; Zhao, L.; Chen, Z.S. Overexpression of ABCB1 Transporter Confers Resistance to mTOR Inhibitor WYE-354 in Cancer Cells. Int. J. Mol. Sci. 2020, 21, 1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Zafar, A.; Wang, W.; Liu, G.; Wang, X.; Xian, W.; McKeon, F.; Foster, J.; Zhou, J.; Zhang, R. Molecular targeting therapies for neuroblastoma: Progress and challenges. Med. Res. Rev. 2021, 41, 961–1021. [Google Scholar] [PubMed]
  108. Bourhis, J.; Benard, J.; Hartmann, O.; Boccon-Gibod, L.; Lemerle, J.; Riou, G. Correlation of MDR1 gene expression with chemotherapy in neuroblastoma. J. Natl. Cancer Inst. 1989, 81, 1401–1405. [Google Scholar] [CrossRef]
  109. Qiu, Y.Y.; Mirkin, B.L.; Dwivedi, R.S. MDR1 hypermethylation contributes to the progression of neuroblastoma. Mol. Cell. Biochem. 2007, 301, 131–135. [Google Scholar] [CrossRef]
  110. Oue, T.; Yoneda, A.; Uehara, S.; Yamanaka, H.; Fukuzawa, M. Increased expression of multidrug resistance-associated genes after chemotherapy in pediatric solid malignancies. J. Pediatr. Surg. 2009, 44, 377–380. [Google Scholar] [CrossRef]
  111. Wang, J.Q.; Yang, Y.; Cai, C.Y.; Teng, Q.X.; Cui, Q.; Lin, J.; Assaraf, Y.G.; Chen, Z.S. Multidrug resistance proteins (MRPs): Structure, function and the overcoming of cancer multidrug resistance. Drug Resist. Updat. 2021, 54, 100743. [Google Scholar]
  112. Cole, S.P. Targeting multidrug resistance protein 1 (MRP1, ABCC1): Past, present, and future. Annu. Rev. Pharmacol. Toxicol. 2014, 54, 95–117. [Google Scholar] [CrossRef]
  113. Arana, M.R.; Altenberg, G.A. ATP-binding Cassette Exporters: Structure and Mechanism with a Focus on P-glycoprotein and MRP1. Curr. Med. Chem. 2019, 26, 1062–1078. [Google Scholar] [PubMed]
  114. Fruci, D.; Cho, W.C.; Nobili, V.; Locatelli, F.; Alisi, A. Drug Transporters and Multiple Drug Resistance in Pediatric Solid Tumors. Curr. Drug Metab. 2016, 17, 308–316. [Google Scholar] [CrossRef] [PubMed]
  115. Mehrvar, N.; Abolghasemi, H.; Rezvany, M.R.; Esmaeil Akbari, M.; Saberynejad, J.; Mehrvar, A.; Ehsani, M.A.; Nourian, M.; Qaddoumi, I.; Movafagh, A. Pattern of ABCC Transporter Gene Expression in Pediatric Patients with Relapsed Acute Lymphoblastic Leukemia. Rep. Biochem. Mol. Biol. 2019, 8, 184–193. [Google Scholar] [PubMed]
  116. Henderson, M.J.; Haber, M.; Porro, A.; Munoz, M.A.; Iraci, N.; Xue, C.; Murray, J.; Flemming, C.L.; Smith, J.; Fletcher, J.I.; et al. ABCC multidrug transporters in childhood neuroblastoma: Clinical and biological effects independent of cytotoxic drug efflux. J. Natl. Cancer Inst. 2011, 103, 1236–1251. [Google Scholar] [PubMed] [Green Version]
  117. Pena-Solorzano, D.; Stark, S.A.; Konig, B.; Sierra, C.A.; Ochoa-Puentes, C. ABCG2/BCRP: Specific and Nonspecific Modulators. Med. Res. Rev. 2017, 37, 987–1050. [Google Scholar]
  118. Khot, M.I.; Downey, C.L.; Armstrong, G.; Svavarsdottir, H.S.; Jarral, F.; Andrew, H.; Jayne, D.G. The role of ABCG2 in modulating responses to anti-cancer photodynamic therapy. Photodiagnosis Photodyn. Ther. 2020, 29, 101579. [Google Scholar]
  119. Toyoda, Y.; Takada, T.; Suzuki, H. Inhibitors of Human ABCG2: From Technical Background to Recent Updates with Clinical Implications. Front. Pharmacol. 2019, 10, 208. [Google Scholar] [CrossRef] [Green Version]
  120. Jain, H.D.; Zhang, C.; Zhou, S.; Zhou, H.; Ma, J.; Liu, X.; Liao, X.; Deveau, A.M.; Dieckhaus, C.M.; Johnson, M.A.; et al. Synthesis and structure-activity relationship studies on tryprostatin A, an inhibitor of breast cancer resistance protein. Bioorg. Med. Chem. 2008, 16, 4626–4651. [Google Scholar]
  121. Zattoni, I.F.; Delabio, L.C.; Dutra, J.P.; Kita, D.H.; Scheiffer, G.; Hembecker, M.; Pereira, G.D.S.; Moure, V.R.; Valdameri, G. Targeting breast cancer resistance protein (BCRP/ABCG2): Functional inhibitors and expression modulators. Eur. J. Med. Chem. 2022, 237, 114346. [Google Scholar]
  122. Doyle, L.A.; Yang, W.; Abruzzo, L.V.; Krogmann, T.; Gao, Y.; Rishi, A.K.; Ross, D.D. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc. Natl. Acad. Sci. USA 1998, 95, 15665–15670. [Google Scholar] [CrossRef] [Green Version]
  123. Sauerbrey, A.; Sell, W.; Steinbach, D.; Voigt, A.; Zintl, F. Expression of the BCRP gene (ABCG2/MXR/ABCP) in childhood acute lymphoblastic leukaemia. Br. J. Haematol. 2002, 118, 147–150. [Google Scholar] [CrossRef] [PubMed]
  124. van den Heuvel-Eibrink, M.M.; Wiemer, E.A.; Prins, A.; Meijerink, J.P.; Vossebeld, P.J.; van der Holt, B.; Pieters, R.; Sonneveld, P. Increased expression of the breast cancer resistance protein (BCRP) in relapsed or refractory acute myeloid leukemia (AML). Leukemia 2002, 16, 833–839. [Google Scholar] [PubMed] [Green Version]
  125. Hirschmann-Jax, C.; Foster, A.E.; Wulf, G.G.; Nuchtern, J.G.; Jax, T.W.; Gobel, U.; Goodell, M.A.; Brenner, M.K. A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proc. Natl. Acad. Sci. USA 2004, 101, 14228–14233. [Google Scholar] [PubMed] [Green Version]
  126. Choi, Y.H.; Yu, A.M. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Curr. Pharm. Des. 2014, 20, 793–807. [Google Scholar] [PubMed]
  127. Vaidyanathan, A.; Sawers, L.; Gannon, A.L.; Chakravarty, P.; Scott, A.L.; Bray, S.E.; Ferguson, M.J.; Smith, G. ABCB1 (MDR1) induction defines a common resistance mechanism in paclitaxel- and olaparib-resistant ovarian cancer cells. Br. J. Cancer 2016, 115, 431–441. [Google Scholar] [CrossRef] [Green Version]
  128. Lal, S.; Wong, Z.W.; Sandanaraj, E.; Xiang, X.; Ang, P.C.; Lee, E.J.; Chowbay, B. Influence of ABCB1 and ABCG2 polymorphisms on doxorubicin disposition in Asian breast cancer patients. Cancer Sci. 2008, 99, 816–823. [Google Scholar]
  129. Wang, X.; Zhang, H.; Chen, X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019, 2, 141–160. [Google Scholar] [CrossRef] [Green Version]
  130. Wu, C.P.; Hsiao, S.H.; Huang, Y.H.; Hung, L.C.; Yu, Y.J.; Chang, Y.T.; Hung, T.H.; Wu, Y.S. Sitravatinib Sensitizes ABCB1- and ABCG2-Overexpressing Multidrug-Resistant Cancer Cells to Chemotherapeutic Drugs. Cancers 2020, 12, 195. [Google Scholar]
  131. Ahmed, F.; Haass, N.K. Microenvironment-Driven Dynamic Heterogeneity and Phenotypic Plasticity as a Mechanism of Melanoma Therapy Resistance. Front. Oncol. 2018, 8, 173. [Google Scholar] [CrossRef] [Green Version]
  132. Allen, J.D.; Brinkhuis, R.F.; Wijnholds, J.; Schinkel, A.H. The mouse Bcrp1/Mxr/Abcp gene: Amplification and overexpression in cell lines selected for resistance to topotecan, mitoxantrone, or doxorubicin. Cancer Res. 1999, 59, 4237–4241. [Google Scholar]
  133. Wei, G.; Wang, Y.; Yang, G.; Wang, Y.; Ju, R. Recent progress in nanomedicine for enhanced cancer chemotherapy. Theranostics 2021, 11, 6370–6392. [Google Scholar] [CrossRef] [PubMed]
  134. Nanayakkara, A.K.; Follit, C.A.; Chen, G.; Williams, N.S.; Vogel, P.D.; Wise, J.G. Targeted inhibitors of P-glycoprotein increase chemotherapeutic-induced mortality of multidrug resistant tumor cells. Sci. Rep. 2018, 8, 967. [Google Scholar]
  135. Lei, Z.N.; Teng, Q.X.; Wu, Z.X.; Ping, F.F.; Song, P.; Wurpel, J.N.D.; Chen, Z.S. Overcoming multidrug resistance by knockout of ABCB1 gene using CRISPR/Cas9 system in SW620/Ad300 colorectal cancer cells. MedComm 2021, 2, 765–777. [Google Scholar] [CrossRef] [PubMed]
  136. Yee Kuen, C.; Masarudin, M.J. Chitosan Nanoparticle-Based System: A New Insight into the Promising Controlled Release System for Lung Cancer Treatment. Molecules 2022, 27, 473. [Google Scholar] [CrossRef] [PubMed]
  137. Xu, T.; Guo, P.; He, Y.; Pi, C.; Wang, Y.; Feng, X.; Hou, Y.; Jiang, Q.; Zhao, L.; Wei, Y. Application of curcumin and its derivatives in tumor multidrug resistance. Phytother. Res. 2020, 34, 2438–2458. [Google Scholar] [PubMed]
  138. Gao, Y.; Zhu, Y.; Xu, X.; Wang, F.; Shen, W.; Leng, X.; Zhao, J.; Liu, B.; Wang, Y.; Liu, P. Surface PEGylated Cancer Cell Membrane-Coated Nanoparticles for Codelivery of Curcumin and Doxorubicin for the Treatment of Multidrug Resistant Esophageal Carcinoma. Front. Cell Dev. Biol. 2021, 9, 688070. [Google Scholar]
  139. Das, T.; Anand, U.; Pandey, S.K.; Ashby, C.R., Jr.; Assaraf, Y.G.; Chen, Z.S.; Dey, A. Therapeutic strategies to overcome taxane resistance in cancer. Drug Resist. Updat. 2021, 55, 100754. [Google Scholar]
  140. Styczynski, J. Drug resistance in childhood acute myeloid leukemia. Curr. Pharm. Biotechnol. 2007, 8, 59–75. [Google Scholar] [CrossRef]
  141. Roskoski, R., Jr. Blockade of mutant RAS oncogenic signaling with a special emphasis on KRAS. Pharmacol. Res. 2021, 172, 105806. [Google Scholar]
  142. Grobner, S.N.; Worst, B.C.; Weischenfeldt, J.; Buchhalter, I.; Kleinheinz, K.; Rudneva, V.A.; Johann, P.D.; Balasubramanian, G.P.; Segura-Wang, M.; Brabetz, S.; et al. The landscape of genomic alterations across childhood cancers. Nature 2018, 555, 321–327. [Google Scholar] [CrossRef] [Green Version]
  143. Drozak, P.; Brylinski, L.; Zawitkowska, J. A Comprehensive Overview of Recent Advances in Epigenetics in Pediatric Acute Lymphoblastic Leukemia. Cancers 2022, 14, 5384. [Google Scholar] [CrossRef] [PubMed]
  144. Filbin, M.; Monje, M. Developmental origins and emerging therapeutic opportunities for childhood cancer. Nat. Med. 2019, 25, 367–376. [Google Scholar] [CrossRef] [PubMed]
  145. Nordlund, J.; Syvanen, A.C. Epigenetics in pediatric acute lymphoblastic leukemia. Semin. Cancer Biol. 2018, 51, 129–138. [Google Scholar] [PubMed]
  146. Kenderian, S.S.; Adults, Y. Acute lymphoblastic leukemia in adolescents and young adults—From genomics to the clinics. Clin. Oncol. Adolesc. Young Adults 2013, 3, 49–62. [Google Scholar]
  147. Neaga, A.; Jimbu, L.; Mesaros, O.; Bota, M.; Lazar, D.; Cainap, S.; Blag, C.; Zdrenghea, M. Why Do Children with Acute Lymphoblastic Leukemia Fare Better Than Adults? Cancers 2021, 13, 3886. [Google Scholar] [CrossRef]
  148. Styczynski, J.; Wysocki, M. In vitro drug resistance profiles of adult acute lymphoblastic leukemia: Possible explanation for difference in outcome to similar therapeutic regimens. Leuk. Lymphoma 2002, 43, 301–307. [Google Scholar] [CrossRef]
  149. Alcantara, M.; Simonin, M.; Lhermitte, L.; Touzart, A.; Dourthe, M.E.; Latiri, M.; Grardel, N.; Cayuela, J.M.; Chalandon, Y.; Graux, C.; et al. Clinical and biological features of PTPN2-deleted adult and pediatric T-cell acute lymphoblastic leukemia. Blood Adv. 2019, 3, 1981–1988. [Google Scholar] [CrossRef]
  150. Andrieu, G.P.; Kohn, M.; Simonin, M.; Smith, C.L.; Cieslak, A.; Dourthe, M.E.; Charbonnier, G.; Graux, C.; Huguet, F.; Lheritier, V.; et al. PRC2 loss of function confers a targetable vulnerability to BET proteins in T-ALL. Blood 2021, 138, 1855–1869. [Google Scholar]
  151. Genesca, E.; Morgades, M.; Gonzalez-Gil, C.; Fuster-Tormo, F.; Haferlach, C.; Meggendorfer, M.; Montesinos, P.; Barba, P.; Gil, C.; Coll, R.; et al. Adverse prognostic impact of complex karyotype (>/=3 cytogenetic alterations) in adult T-cell acute lymphoblastic leukemia (T-ALL). Leuk. Res. 2021, 109, 106612. [Google Scholar] [CrossRef]
  152. Ma, X.; Edmonson, M.; Yergeau, D.; Muzny, D.M.; Hampton, O.A.; Rusch, M.; Song, G.; Easton, J.; Harvey, R.C.; Wheeler, D.A.; et al. Rise and fall of subclones from diagnosis to relapse in pediatric B-acute lymphoblastic leukaemia. Nat. Commun. 2015, 6, 6604. [Google Scholar] [CrossRef] [Green Version]
  153. Miller, K.D.; Nogueira, L.; Devasia, T.; Mariotto, A.B.; Yabroff, K.R.; Jemal, A.; Kramer, J.; Siegel, R.L. Cancer treatment and survivorship statistics, 2022. CA Cancer J. Clin 2022, 72, 409–436. [Google Scholar] [CrossRef] [PubMed]
  154. Jeenia, F.T.; Sojib, F.A.; Rahman, M.S.; Ara, T.; Khan, R.; Tanin, M.J.U.J.M. Neuroprotective effect of vitamin B6 and vitamin B12 against vincristine-induced peripheral neuropathy: A randomized, double-blind, placebo controlled, multicenter trial. medRxiv 2021. [CrossRef]
  155. Clemens, E.; van den Heuvel-Eibrink, M.M.; Mulder, R.L.; Kremer, L.C.M.; Hudson, M.M.; Skinner, R.; Constine, L.S.; Bass, J.K.; Kuehni, C.E.; Langer, T.; et al. Recommendations for ototoxicity surveillance for childhood, adolescent, and young adult cancer survivors: A report from the International Late Effects of Childhood Cancer Guideline Harmonization Group in collaboration with the PanCare Consortium. Lancet Oncol. 2019, 20, e29–e41. [Google Scholar] [CrossRef] [PubMed]
  156. Langer, T.; Clemens, E.; Broer, L.; Maier, L.; Uitterlinden, A.G.; de Vries, A.C.H.; van Grotel, M.; Pluijm, S.F.M.; Binder, H.; Mayer, B.; et al. Usefulness of current candidate genetic markers to identify childhood cancer patients at risk for platinum-induced ototoxicity: Results of the European PanCareLIFE cohort study. Eur. J. Cancer 2020, 138, 212–224. [Google Scholar] [CrossRef] [PubMed]
  157. van Kalsbeek, R.J.; Mulder, R.L.; Haupt, R.; Muraca, M.; Hjorth, L.; Follin, C.; Kepak, T.; Kepakova, K.; Uyttebroeck, A.; Mangelschots, M.; et al. The PanCareFollowUp Care Intervention: A European harmonised approach to person-centred guideline-based survivorship care after childhood, adolescent and young adult cancer. Eur. J. Cancer 2022, 162, 34–44. [Google Scholar] [CrossRef]
Figure 1. Neuropathy and myopathy induced by two commonly used chemotherapeutic agents (DOX and VCR) in pediatric cancer treatment.
Figure 1. Neuropathy and myopathy induced by two commonly used chemotherapeutic agents (DOX and VCR) in pediatric cancer treatment.
Cancers 15 01963 g001
Figure 2. Mechanisms of MDR in cancer treatment. MDR occurs as a result of various causes, which ultimately lead to tumor recurrence.
Figure 2. Mechanisms of MDR in cancer treatment. MDR occurs as a result of various causes, which ultimately lead to tumor recurrence.
Cancers 15 01963 g002
Table 1. Chemo drugs approved prior to FDAMA, for which labeling includes pediatric indications [24].
Table 1. Chemo drugs approved prior to FDAMA, for which labeling includes pediatric indications [24].
DrugOriginal
Approval
Indications
Doxorubicin Hydrochloride7 August 1974Wilm’s Tumor and Other Childhood
Kidney Cancers
Vincristine Sulfate10 July 1963ALL, Neuroblastoma, Non-Hodgkin Lyphoma, Rhabdomyosarcoma, Wilm’s tumor and other childhood kidney
cancers
Cytarabine17 June 1969Acute Nonlymphocytic Leukemia
Cyclophosphamide16 November 1959ALL
Methotrexate Sodium (Trexall)10 August 1959ALL
Mercaptopurine (Purinethol, Purixan)11 September 1953ALL
Daunorubicin Hydrochloride (Rubidomycin)19 December 1979ALL
Procarbazine Hydrochloride (Matulane)22 July 1969Hodgkin Lymphoma
Dactinomycin (Cosmegen)10 December 1964Ewing sarcoma, gestational
trophoblastic disease
Data provided by National Cancer Institute (https://www.cancer.gov/), accessed on 30 January 2023, updated: 20 December 2022, and U.S. FDA (https://www.fda.gov/). The drugs and drug combinations are not listed here.
Table 2. Chemo drugs approved post FDAMA with pediatric specific indications (1997–2022) [24].
Table 2. Chemo drugs approved post FDAMA with pediatric specific indications (1997–2022) [24].
DrugsOriginal ApprovalPediatric
Approval
Indications for
Pediatric
Cancer
(Drugs approved post FDAMA with pediatric specific indications (1997–2022))
Azacitidine (Vidaza)19 May 200420 May 2022JMML
Calaspargase Pegol-mknl (Asparlas)same20 December 2018ALL
Everolimus1 November 201029 August 2012Giant Cell
Astrocytoma
Asparaginase Erwinia Chrysanthemi
(Erwinaze)
same18 November 2011ALL
Clofarabine (Clolar)same28 December 2004ALL
Pegaspargase (Oncaspar)same24 July 2006ALL
Nelarabine (Arranon)same28 October 2005Non-Hodgkin
Lymphoma
Data provided by National Cancer Institute (https://www.cancer.gov/), updated: 20 December 2022, and U.S. FDA (https://www.fda.gov/). JMML: juvenile myelomonocytic leukemia. The drugs and drug combinations are not listed here.
Table 3. Clinical trials of DIC and VIPN.
Table 3. Clinical trials of DIC and VIPN.
ToxicityStudy TitleNCT IdentifierPhasePatient NumberDiseaseStatusTreatment/Method
DIC *Protective Role of Vitamin D in Breast Cancer Patients Treated with DoxorubicinNCT04166253Phase 2100Breast cancerCompletedVitamin D
DIC99mTc-rhAnnexin V-128 Imaging and Cardiotoxicity in Patients with Early Breast CancerNCT02677714Phase 214Breast cancerTerminatedRadiation: 99mTc-rhAnnexin V-128
DICPrevention Using Exercise Rehabilitation to Offset Cardiac Toxicities Induced Via Chemotherapy (HF-PROACTIVE)NCT02796365Not Applicable29Breast cancer,
Gastric cancer,
Leukemia
CompletedExercise
DICEvaluation of Myocardial Injury After Anthracycline Chemotherapy in Osteosarcoma Patients Using CMRNCT04461223Not Applicable55Osteosarcoma
Myocardial Injury
UnknownContrast-enhanced cardiac magnetic resonance imaging, observational Study
VIPN #Neuroprotective Effect of Vitamin B12 and Vitamin B6 Against Vincristine Induced Peripheral NeuropathyNCT02923388Phase 488Acute Lymphoblastic Leukemia (ALL)CompletedVitamin B12 and vitamin B6
VIPNPhysiologic Measure of VIPNNCT04786977Not Applicable40Chemotherapy-induced Peripheral NeuropathyRecruitingNo Intervention, observational Study
Data provided by Clinical Trials (https://clinicaltrials.gov). * DIC-doxorubicin-induced cardiotoxicity; # Vincristine Induced Peripheral Neuropathy.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bo, L.; Wang, Y.; Li, Y.; Wurpel, J.N.D.; Huang, Z.; Chen, Z.-S. The Battlefield of Chemotherapy in Pediatric Cancers. Cancers 2023, 15, 1963. https://doi.org/10.3390/cancers15071963

AMA Style

Bo L, Wang Y, Li Y, Wurpel JND, Huang Z, Chen Z-S. The Battlefield of Chemotherapy in Pediatric Cancers. Cancers. 2023; 15(7):1963. https://doi.org/10.3390/cancers15071963

Chicago/Turabian Style

Bo, Letao, Youyou Wang, Yidong Li, John N. D. Wurpel, Zoufang Huang, and Zhe-Sheng Chen. 2023. "The Battlefield of Chemotherapy in Pediatric Cancers" Cancers 15, no. 7: 1963. https://doi.org/10.3390/cancers15071963

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop