Skip to main content

Nanotechnology in leukemia: diagnosis, efficient-targeted drug delivery, and clinical trials

Abstract

Leukemia is a group of malignant disorders which affect the blood and blood-forming tissues in the bone marrow, lymphatic system, and spleen. Many types of leukemia exist; thus, their diagnosis and treatment are somewhat complicated. The use of conventional strategies for treatment such as chemotherapy and radiotherapy may develop many side effects and toxicity. Hence, modern research is concerned with the development of specific nano-formulations for targeted delivery of anti-leukemic drugs avoiding toxic effects on normal cells. Nanostructures can be applied not only in treatment but also in diagnosis. In this article, types of leukemia, its causes, diagnosis as well as conventional treatment of leukemia shall be reviewed. Then, the use of nanoparticles in diagnosis of leukemia and synthesis of nanocarriers for efficient delivery of anti-leukemia drugs being investigated in in vivo and clinical studies. Therefore, it may contribute to the discovery of novel and emerging nanoparticles for targeted treatment of leukemia with less side effects and toxicities.

Introduction

Leukemia is a class of cancerous diseases that impact the bone marrow, lymphatic system, spleen, and blood-forming organs. It causes one type of leukocyte, i.e., white blood cell or WBC, to proliferate tremendously resulting in leukocytosis. Leukocytosis is a typical response to infection; but, when it persists over an extended period of time or increases gradually without an apparent reason, it may be a sign of cancer [1]. Leukemia comes in a variety of forms, some of which are more prevalent in young patients, while cases of other types of leukemia affect adults [2]. The most crucial thing is to promptly identify and diagnose affected patients; failing to do so could result in death [3]. Although the precise etiology of leukemia is unknown, a number of variables, including genetic predisposition, chromosomal abnormalities, chemical agents (such as benzene), chemotherapeutic drugs, radiation, immunocompromise, and viruses, may contribute to the disease's development [2].

Over the past few years, malignant cells, including leukemia, have gained resistance against cell division inhibitors and commonly used chemotherapeutic drugs [4]. This resistance suggests that cancer cells may be able to change their genetic makeup to initiate angiogenesis and combat the hypoxia brought on by these agents [5]. Because of this, the latest treatment approaches are made to specifically target the suppression of tumor angiogenesis or alter signaling pathways, which are responsible for the development of chemotherapy resistance and tolerance [6]. Adjuvant and neo-adjuvant surgery, radiation, and combinations of chemotherapeutic drugs are all part of conventional cancer treatment regimens. The side effects and toxicities that come with the use of such protocols include bone-marrow suppression and consequently immunosuppression, inflammation of the mucosa, gastrointestinal (e.g., nausea, vomiting, and diarrhea) in addition to alopecia, exhaustion, sterility, and infertility. In addition, immunosuppression increases the risk of infections [7,8,9]. Therefore, the focus of current research is on creating customized nano-formulations that deliver drugs to cancer cells specifically while avoiding harmful side effects on healthy cells [10].

Interest in using nanostructures as a vehicle for delivering various medicinal compounds is growing. In addition to being employed as a drug delivery method, nanostructures are also utilized in molecular diagnosis, illness detection, and nanoscale immunotherapy [11, 12]. Target-based drug development regimens, in which the therapeutic agents become specific to the genotype of the cancer cell and possess strong affinity to various molecular targets, were developed by the invention of screening procedures based on nanotechnology [13, 14]. The creation of novel therapeutic techniques, improved drug distribution, and increased bioavailability are the key benefits of using nanoparticles (NPs) in medicine [15]. Numerous NPs types, including dendrimers, graphene, fullerene, metallic, magnetic, polymeric, metal oxide, quantum dots, liposomes, carbon nanotubes, and graphene, may find use in the detection and therapy of cancer [16]. Nanostructures may increase the biologically active drug's transport and deposition at the tumor site, improve therapeutic efficacy, and lessen the severity of side effects on healthy, normal tissues [17]. One can combine a medicinal medicine and a diagnostic agent into a single nanoparticle due to the internal makeup of NPs. In this manner, the drug's release, biodistribution, and accumulation may all be efficiently tracked and measured to evaluate the medication's therapeutic effectiveness [12]. Currently, organic nanomaterials such as liposomes and polymer micelles are the mainstay of Food and Drug Administration (FDA)-approved or clinically researched nanomedicine against hematological malignancies [18].

Prior to the effective clinical use of nano-based anti-leukemia drugs in humans, a number of parameters, including clearance, long-term toxicity, biocompatibility, immunogenicity, selectivity, pharmacokinetics, and biodistribution, still require a lot of investigations. Therefore, the goal of this review is to outline the background that justifies the application of NPs conjugated with various therapeutic agents in targeted therapy and cancer research, with a focus on leukemia specifically. First, we will go over leukaemia: what it is, how it is diagnosed, and what its forms are. Then, the key cellular pathways that are targeted by leukemia medications shall be discussed. The final areas include novel and emerging NPs for targeted leukemia treatment, nanocarriers for efficient anti-leukemia drug delivery, and NPs for leukemia diagnosis. Nano-based easy, rapid, selective, and early diagnosis may provide a clinically valuable information for prompt treatment. The review is devoted for the readers and experts in this important field of medicine, particularly oncology, since the same approaches may be applied for other types of cancers. Create nanomedicines in the future that are resistant to chemotherapy and targeted therapy based on the mechanism which could be the beginning of the end for leukaemia [19].

Searching strategies

Various scientific databases, including Google Scholar, PubMed, and Scopus, were used, where articles published in peer-reviewed journals and book chapters were considered. The time period was non-specified, but publications were prioritized, particularly in the last 10 years. Keywords related to the main topic of the current research, such as "Leukemia; Nanobiotechnology; Treatment; Diagnosis; Drug delivery". The search resulted in huge number of articles, which have been then filtered to include mainly molecular mechanisms-based studies and advanced formulations for drugs being promoted for in vivo, clinical trials, and approval for diagnosis and management of drug-resistant cases.

Leukemia types and diagnosis

There are four main forms of leukemia, including acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and chronic myeloid leukemia (CML), which are listed in Table 1. Since complicated and distinct group of hematological neoplasms are involved in the diagnosis of leukemia, morphologic variations, cytogenetic anomalies, immunophenotype, and clinical characteristics could distinguish each type from the others [20].

Table 1 Major four types of leukemia types and manifestations

The diagnosis of ALL involves cell morphology characterization by microscopy, immunophenotyping which is the gold standard for lineage assessment, classification, and detection of features that are important for the assessment of minimal residual disease. Chromosomal analysis with fluorescence in-situ hybridization or reverse transcription polymerase chain reaction (RT-PCR) is performed for the detection of selected genomic abnormalities, or recently in next generation sequencing have made possible whole genome sequencing [21]. In addition, AML diagnosis requires the identification of 20% or more myeloid blasts with morphological assessment of the peripheral blood or bone marrow. In addition, immunophenotyping by flow cytometry should be investigated to confirm the myeloid origin of malignant blast populations and to categorize the subtype of acute myeloid leukemia. Cytogenetic analysis and screening for the most common occurring gene mutations and rearrangements [22]. However, CLL diagnosis is determined by the presence of more than 5 × 109/l monoclonal B lymphocytes in the peripheral blood. Using flow cytometry, the clonality of these B cells should be verified. Furthermore, the leukemia cells in the blood smear have the typical tiny, mature appearance of lymphocytes, a compact nucleus devoid of distinguishable nucleoli, and partially aggregated chromatin. They also have a limited border of cytoplasm [23]. Besides, the diagnosis of CML can be made using differential excessive agranulocytosis and a distinctive blood count. The detection of the Philadelphia chromosome, i.e., 22q- or BCR-ABL1 transcripts or both, in peripheral blood or bone-marrow cells may be conclusive for CML diagnosis [24].

Cellular pathways for leukemia drug targeting

Canonical nuclear Factor Kappa B (NF-κB)-inducing kinase

Verteporfin, a benzo porphyrin derivative, evidenced anti-AML by targeting canonical NF-κB—activated in AML stem cells—which is essential for maintenance of the renewal of leukemia stem cells (LSCs). Since, the inhibition of canonical signaling by the ectopic expression of IκBα reduces AML development. Therefore, targeting the pathway is regarded as a potential therapy in AML treatment. Verteporfin impairs NF-κB-inducing kinase (NIK), eventually suppresses AML. It stabilizes NIK, and hence, activates NF-κB non-canonical signaling and inhibits NF-κB canonical signaling. Furthermore, stabilization of NIK-induced activation of NF-κB non-canonical signaling upregulates Dnmt3a to deactivate AML and downregulates Mef2c that activates AML progression. These upregulations and downregulations of the genes present exceptionally in NIK-stabilized leukemia cell supports the anti-AML of verteporfin [29]. Figure 1 summarizes pathways involved in stabilization of canonical NIK.

Fig. 1
figure 1

Stabilization of Canonical NF-κB-inducing kinase. AML: acute myeloid leukemia, caNIK: canonical nuclear factor κB inducing kinase; Mef2c: myocyte-specific enhancer factor; Dnmt3a: DNA (cytosine-5)-methyltransferase 3A, RNfkbia: nuclear factor that binds to the enhancer element of the Ig kappa light-chain of activated B cells;Tnfaip3: protein induced by TNF-mediated NF-κB activation and has a dual function in regulating NF-κB associated with inflammatory carcinogenesis in many cancer types; TRAF3: TNF receptor-associated factor

B-cell receptor (BCR)

The signal that BCR activates is one of the main targets of B-cell tumors. Several potential kinases are released downstream and drive the proliferation and survival of CLL. Consequently, a novel therapeutic strategy for CLL is the development of BCR kinase inhibitors, which specifically target the route of these kinases. A class of drugs known as BCR inhibitors has the ability to stop the target of CLL cells and release them into the peripheral blood stream, where they can be readily attacked by other substances. For instance, ibrutinib and idelalisib are Bruton’s Tyrosine Kinase (BTK) and phosphoinositide 3-kinase-δ (PI3K-δ) inhibitors, respectively, that have been approved for CLL treatment. In addition, duvelisib can inhibit both ẟ and γ isoforms of PI3K. By preventing CLL cells from homing into their specific microenvironments, kinase inhibitors drive CLL cells into the peripheral circulation, where they undergo lymphocytosis without producing hematotoxicity. BCR inhibitors can, therefore, target the CLL cells that are concealed in the lymph node and marrow indentation [30]. Nano-antitumor therapy that targets the tumor microenvironment has gained a lot of attention ascribed to the physiological differences between tumors and normal tissues [31].

Fostamatinib is a spleen tyrosine kinase (Syk) inhibitor, where SYK plays an important role in BCR-mediated survival and is mainly activated in CLL. Therefore, inhibition of SYK signaling avoids the interaction of the CLL cells with its relevant microenvironment and stimulates apoptotic signals. Fostamatinib reduces the phosphorylation of the proximal BCR signaling regulators SLP65 and PLCγ2, in addition to ERK, AKT, and NF-κB downstream pathways, eventually, decreases proliferation and survival of CLL cells [32]. Moreover, perifosine hinders the phosphorylation of Ak strain transforming (AKT) via avoiding the localization of AKT in the membrane, hence interfering with the interaction between PI3K and AKT [33]. The several BCR signaling inhibitors and mediators that are involved are displayed in Fig. 2.

Fig. 2
figure 2

B-cell receptor signaling inhibitors. BCR: B cell receptor; BTK: Bruton’s tyrosine kinase; SYK: spleen tyrosine kinase; BLNK: B-cell linker; PLCγ2: phospholipase Cγ2; PI3K: phosphoinositide 3-kinase, the “P” in blue circle indicates phosphorylation, LYN: a gene on chromosome 8q13 that encodes a non-receptor tyrosine protein kinase, AKT: serine/threonine kinase

Chronic lymphocytic leukemia pathogenesis and the suggested therapeutic targets

Precision medicine plays a backbone in the treatment of CLL, since the integration of molecular biomarkers with the clinical features of the disease may lead to the correct treatment strategy. These treatment strategies depend on certain molecular features of the disease. Tumor protein p53 (TP53) disruption is the strongest prognosticator of resistant cancer, accordingly the assessment of TP53 status’ evaluation is considered the first line treatment. Therefore, patients featuring TP53 disruption need drugs being able to inhibit the BCR or B-cell lymphoma 2 (BCL2) pathway. Moreover, the mutational status of immunoglobulin heavy variable (IGHV) genes directs the modification of treatment. Patients carrying mutated IGHV genes, in the absence of TP53 disruption respond positively with chemo-immunotherapy. Conversely, patients with un-mutated IGHV genes respond negatively to chemo-immunotherapy and require BCR inhibitors [34]. As revealed in Fig. 3, venetoclax can bind to BCL2 and inhibits the anti-apoptotic protein, which is damaged by several molecular mechanisms, such as del13q14, therefore, repairing the apoptosis in CLL cells. Ibrutinib, BTK inhibitor, can target BTK which is an essential kinase present downstream to the BCR pathway. Another pathway is the inhibition of the non-canonical NF-κB pathway that is fundamental to cell survival and progression as mutation of upstream signaling molecules TRAF3 leads to activation of non-canonical NF-κB. In addition, NOTCH homolog 1 (NOTCH1) mutations dislocate the PEST domain, the NOTCH intracellular domain (NICD), this leads to a vital transcription of target genes enhancing cell proliferation. The canonical activation of Notch signaling is mediated by ligand-mediated mechanisms [35]. Thus, the advance of therapeutic agents that interfere with ligand–receptor binding is regarded as an important pivot in the treatment [36].

Fig. 3
figure 3

Biological pathways for chronic lymphocytic leukemia pathogenesis and the suggested therapeutic targets. BCL2: B-cell lymphoma 2; BTK: Bruton’s tyrosine kinase; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; (NOTCH1): notch homolog 1 PEST: a peptide sequence that is rich in proline (P), glutamic acid (E), serine (S), and threonine (T) of the NICD: NOTCH intracellular domain; TP53: tumor protein 53; ATM: ataxia telangiectasia mutated proteins

NOTCH signaling

An important active NOTCH1 signaling, initiated by activating mutations in the NOTCH1 gene, is responsible for some oncogenic transformation that leads to malignant diseases, Fig. 4. T-cell acute lymphoblastic leukemia (T-ALL) is a tumor resulting from the malignant transformation of T-cell progenitors via the mutations in the NOTCH1 gene. γ-Secretase inhibitors (GSIs) can efficiently block NOTCH1 signaling in T-ALL, and could be regarded as a targeted therapy [37]. γ-Secretase complex proteolyzes the release of the intracellular portion of NOTCH1 (ICN1) and is considered a crucial step in the canonical NOTCH signaling. Therefore, GSIs that target the NOTCH receptors have been implemented in cancers, where NOTCH1 mutations are common (e.g., T-ALL and CLL). Studies showed that GSI treatment induces G0/G1 arrest along with rapid clearance of intracellular NOTCH1. However, the clinical development of GSIs has been hindered by their low cytotoxicity against human T-ALL and GIT toxicity derived from inhibition of NOTCH signaling in the gut [38]. Furthermore, anti-NOTCH1 inhibitory antibodies, small peptide inhibitors of NOTCH signaling as well as combination chemotherapy with GSIs and glucocorticoids, have lately been proposed as anti-NOTCH therapeutic protocols. Investigation of NOTCH1 mutations in chronic lymphocytic leukemia has aroused in advance the clinical significance of NOTCH signaling as a therapeutic target in human cancer.

Fig. 4
figure 4

NOTCH signaling pathway. ADAM: metalloprotease (metalloproteinase/disintegrin/cysteine-rich); ICN1: intracellular domains of NOTCH1; MAML1 co-activator (Mastermind-like 1); PEST: peptide sequence that is rich in proline (P), glutamic acid (E), serine (S), and threonine (T); HD: heterodimerization domain; LNR: lymph node ratio NOTCH1 activation shows: the interaction between the NOTCH1 receptor, Delta-like and Jagged ligands which are expressed on the surface of a near cells activating the proteolytic cleavage of the receptor; by an ADAM metalloprotease (metalloproteinase/disintegrin/cysteine-rich) (S2 cleavage) followed by the γ-secretase complex (S3 cleavage), which releases the intracellular domains of NOTCH1 (ICN1) from the membrane. ICN1 is transferred to the nucleus and interacts with DNA-binding protein and induces the MAML1 co-activator (Mastermind-like 1) to activate the expression of NOTCH1 target genes

One of the hematological malignancies is T-ALL. The novel chemotherapy approaches and/or stem cell transplantation have improved the cure rate for T-ALL; nonetheless, the relapse rate among patients necessitates alternate therapies. Remarkably, one of the main causes of this relapse is gene mutations, specifically NOTCH1 gene mutation. Targeting the NOTCH1 signaling pathway may be crucial to treating relapsed and resistant T-ALL, since NOTCH1 gene mutations are important oncogenes in T-ALL [39]. Hence, A Disintegrin and Metalloprotease (ADAM) inhibitors, gamma secretase inhibitors (GSIs), monoclonal antibody targeting NOTCH1, in addition to sarco/endoplasmic reticulum (ER) Ca2+-ATPase (SERCA) inhibitors are potential candidates for leukemia managements. In more details. These classes are illustrated in Fig. 5.

Fig. 5
figure 5

NOTCH1 signaling pathway and drug targets. ADAM: metalloproteinase/disintegrin/cysteine-rich; NRR: negative regulatory region; SERCA: Sarco/Endoplasmic Reticulum Calcium-ATPase; LNR: lymph node ratio; HD: heterodimerization domain; SAHM1: hydrocarbon-stapled synthetic peptide stapled α-helical peptide derived from mastermind-like 1 prevent the formation of MAML: co-activator (Mastermind-like-1); ICN1: intracellular domains of NOTCH1;PEST: peptide sequence is rich in proline (P), glutamic acid (E), serine (S), and threonine (T), CSL: transcription factor. ADAM inhibitors inhibit ADAM protease S2 site uptake; γ-secretase inhibitors (GSIs) prevent the cleavage of S3 site of γ-secretase; Monoclonal antibody prevent the change the spatial arrangement of the negative regulatory region (NRR) from exposing the S2 and S3 cleavage sites; SERCA inhibitors include Ca2+ ATPase inhibitors prevent transportation of NOTCH1 and the formation of heterodimerization domain (HD); SAHM1 hinder the binding complex of ICN1–CSL–MAML

Conventional treatment of Leukemia

Chemotherapy

Novel findings of chemotherapeutic agents, which are recognized as scientific and technological advance, allowed insights of cell biology of human cancer cells, and hence, targeted therapy has been applied. Granting the targeted therapy drugs with outstanding accomplishments in certain cancer cells, new therapies still cannot replace cytotoxic agents. Moreover, clinical trials have evidenced the combined effect of targeted molecules and traditional cytotoxic agents [40]. They are classified into alkylating agents (e.g., nitrogen mustards and its based hybrids) which are marketed as cyclophosphamide (Cytoxan® or Neosar®) [41].

Purine analogues as 6-mercaptopurine (6-MP) is one of the primary chemotherapeutic agents applied in acute leukemia, and still one of the most convenient drugs in maintenance therapy of ALL [42, 43]. It is marketed under the names Purinethol® and Purixan®. In addition, pyrimidine analogues are represented by cytarabine (1-β-D-arabinofuranosylcytosine; ara-C) T: Cytosar-U®, Tarabine PFS®. This drug has been approved by FDA and NCI for the management of ALL, CML and to prevent and treat meningeal leukemia. Its mechanism of action resides in the transportation of ara-C into the cell then it is phosphorylated into ara-C monophosphate (ara-CMP) by DCK and eventually to ara-C triphosphate (ara-CTP) which then contests with deoxycytidine triphosphate (dCTP) for incorporation into DNA and finally blocking DNA synthesis causing cell death [44].

Microtubule-target agents (MTA) have been also involved in the preservation of cell structure, protein transportation and mitosis. Vinca alkaloids are potential examples for this class. The most clinically important three alkaloids are vinblastine (VBL), vincristine (VCR), and vinorelbine (VRL) which have been approved in the United states by FDA [45]. Vinca alkaloids are recognized as destabilizing agents, where they depolarize microtubule, block the mitotic progression, and eventually lead to cell death by apoptosis [46]. VCR is marketed as Oncovin® and Vincasar Pfs® and commonly used in combination chemotherapy for treating pediatric leukemia.

Leukemia patients are treated with antibiotics (e.g., anthracyclines and anthracenediones), as one of the main classes of chemotherapeutic drugs. Two anthracyclines for AML, daunorubicin (DD) (Cerubidine®) and idarubicin (IDA) (idamycin PFS®), are thought to work by forming complexes with DNA through base pair intercalation and inhibiting topoisomerase II activity by stabilizing the DNA–topoisomerase II complex, which stops topoisomerase II from catalyzing the ligation–relegation reaction. Idarubicin may also damage DNA by free radical damage, disrupt the control of gene expression, and limit polymerase activity. Furthermore, it was stated that idarubicin outperformed daunorubicin in terms of intracellular accumulation and DNA binding ability [47, 48]. Besides, mitoxantrone (e.g., Novantrone®, Mitozantrone®) belong to anthracenediones class and has been approved for use in AML [49]. Notably, compared to anthracyclines, mitoxantrone showed reduced cardiac toxicity because of its lower generation of quinone-type free radicals.

Combination between DNT cellular therapy with conventional chemotherapy

CD3 + CD4–CD8–double negative T cells (DNTs) are immune cells that produce substantial cytotoxicity in many tumor cells in a nonspecific manner. As a result, they have emerged as functional immune cells in the field of anticancer therapy [50]. The AML resistance to chemotherapy, which ultimately results in recurrence and death, is one of the main problems with conventional chemotherapeutic drugs. To enhance the anti-leukemic effects of conventional chemotherapy and target diseases resistant to chemotherapy, a combination of DNT therapy and chemotherapy was proposed as an alternate treatment. Ara-C and daunorubicin (DNR) were used in combination with DNT in xenograft model, results revealed that the efficacy of combining an adoptive T-cell therapy and low-dose chemotherapy in reducing therapy-resistant AML. These findings suggested utilizing DNTs as an adjuvant cellular therapy following administration of chemotherapy [51].

Interferon-α (INF-α) in combination with conventional chemotherapy

Imatinib (Gleevec®) is a competitive pyridine-based inhibitor interfering with the ATP binding site, including tyrosine kinase, and hence, it is also classified among tyrosine kinase inhibitor (TKI) [52]. This mechanism has qualified imatinib as a drug of choice for the treatment for CML that is evidenced by improvement in patients with CML, though it has limitations, including the inability to eradicate CML primitive progenitors, which resulted in the elevation of the relapse rate when imatinib is withdrawn. On the other hand, interferon-α (IFN-α) (e.g., Intron®-A or Roferon®-A) directly targets CML stem cells by inducing cytogenetic remission. Consequently, patients treated with IFN-α were able to sustain durable remissions after discontinuing therapy. Nevertheless, the side effects of IFN-α hinder its administration as well as its cost was 200 times those of the conventional therapy [53]. Furthermore, clinical studies suggested that the combination of imatinib and IFN-α is superior to either therapy, due to their different mechanisms of action [54].

Radiotherapy

Radiation therapy (RD) may be used to eradicate leukemia cells or relieve pain from swollen lymph nodes, the spleen, or the liver. It is also a promising treatment for bone pain brought on by leukemia cells growing in the bone marrow. Low doses of RD may also be administered before stem cell transplantation. It includes external beam radiation therapy (EBRT), total body irradiation (TBI), and total marrow irradiation (TMI) [55]. Because lymphocyte cells are extremely sensitive to radiation, adding RD to systemic therapy has been reported to be effectively used for leukemia treatment. Therefore, use of RD is a well-established regimen prior to allogeneic stem cell transplantation [56].

Nanotechnology approaches

Nano-oncology and nanomedicine has attracted special interest based on NPs-formulated drugs holding promise for the delivery of small-molecule anticancer drugs especially leukaemia with many trials and studies focusing on the use of NPs for diagnosis and treatment [57, 58]. This interest attributes to the ability of NPs to alter drug solubility and improve their stability in vivo. In addition, they can be tailored to offer specific targeting to specific cells or tissues, thereby reducing the side effects compared to free drugs as well as to overcome resistance to the chemotherapeutics [59,60,61]. Figure 6 summarizes the various approaches of nanotechnology applied against leukemia. They include diagnosis, drug delivery, and targeted treatment. They shall be also discussed in the following subsections.

Fig. 6
figure 6

Summary of nanotechnology and nanomedicine approaches applied against leukemia

NPs with a size range of 1–100 nm are especially beneficent in cancer treatment which offer a large surface area in respect to their volume due to their small particle size which enable the sufficient loading of ligands for targeting of specific cells or organelles delivery as well as stealth such as properties yielded by hydrophilic moieties which protect the encapsulated drug from elimination and extend the drug levels in circulation in vivo thereby retaining their effect for prolonged periods and eliminating frequent dosing [62, 63]. Thus, offering many advantages, such as targeting of drugs, reduced dosage and/or dosing frequency, enhanced drug solubility, increased half-life of drugs in vivo and reduced immunogenicity [64, 65].

Nanoparticles for diagnosis of leukemia

Traditional detection techniques have certain drawbacks, like being expensive, time-consuming, and labour-intensive. They are not suitable for simple and quick medical monitoring or analysis, since they need a complex combination of equipment, have low sensitivity, and require multiple processing stages. Thus, modern diagnostic tools have been investigated to create cutting-edge tactics in response to the need for the development of improved diagnostic and therapeutic approaches with the highest level of specificity, efficiency, and minimal toxicity [61]. Therefore, rapid, sensitive, easy-to-use, and reasonably priced instruments are becoming more and more necessary for the diagnosis of leukaemia. These tools should not only enable correct diagnosis but also help doctors choose the best course of treatment. Karyotyping, immunophenotyping by flow cytometry or microarray, which combines bone marrow and peripheral blood cytochemical tests, and PCR-based techniques—which are costly and involve numerous steps—are among the diagnostic methods for leukaemia [65]. In this context, NPs have demonstrated their ability to offer substitute approaches to these challenges by offering quick, secure, and precise leukaemia diagnosis techniques.

Specific aptamers which bind to specific target molecules have been widely exploited loading on NPs for detection of leukaemia cells has been applied by many researchers due to their specificity of targeting the desired cells based on loading the NPs with specific targeting molecules with negligible cytotoxicity [63, 66]. One example is Sgc8 aptamer which has been employed in studies because of its selectivity to leukaemia cell. Tan, et al. utilized a simple method to detect leukaemia based on amine-labelled Sgc8 aptamer-modified fluorescent silica NPs (FSNPs) prepared via amide coupling between amino and carboxyl groups which detected leukaemia cells with high accuracy and sensitivity assessed by flow cytometry and fluorescence microscopy. Sgc8-specific targeting combined with FSNPs' showed stronger fluorescence intensity and photostability, and thus, it may provide a clinically valuable tool for early diagnosis and prompt treatment [67]. In addition, Khoshfetrat et al. used thiolated Sgc8c aptamer immobilized on gold NPs-coated magnetic Fe3O4 NPs (Apt-GMNPs) with a hairpin structure to form Aptasensors and intercalating Ethidium bromide (EB) for the quantification of the leukaemia cells. When leukaemia cancer cells are introduced onto the Aptasensors, the hairpin structure of the aptamer is disrupted and the intercalated molecules are released, resulting in a decrease of the electrochemical signal which is detected. This new diagnostic tool showed a linear response and was successful in detection of leukaemia cancer cells in complex media, such as plasma, without significant interference [68].

Grechkin et al. also utilized Sgc8-aptamer conjugated with amino- and carboxyl-modified silica-coated terbium (III) thiacalix arenesulfonate to form luminescent NPs that were able to detect leukaemia cells using fluorescence microscopy without induction of cell apoptosis or necrosis, which can be common in bioimaging applications. At this experiment, CCRF–CEM (human T lymphoblasts cell line) and Jurkat cells (an immortalized line of human T lymphocyte) were investigated as models. The findings indicated that CCRF–CEM had a larger shift in fluorescence intensity than Jurkat cells, which may be related to the latter's lower concentration of a tyrosine protein kinase 7 (PTK7) on the cellular membrane [69].

The labelling of NPs or receptor ligands by fluorescent markers is a well-established method for cancer diagnosis as well as the identification of cell phenotype. Maclaughlin et al. harnessed this strategy by developing an antibody-targeted, PEG-coated surface-enhanced Raman scattering (SERS) gold NPs (AuNPS) which can be used to label three cell surface markers on cancerous B cells from the LY10 lymphoma cell line. Using confocal Raman mapping and SERS from cell suspensions, the specificity of the particles' cell labelling was shown on primary CLL and LY10 cells, respectively. The results were also confirmed through the flow cytometer which was used to validate the binding of SERS probes to LY10 over vast cell populations [70]. Moreover, Haghighi et al. combined fluorescent gold nanoclusters (GNCs) with a super paramagnetic (Fe3O4/GNCs) nanoprobe for imaging and detection of human leukaemia cancer cells (HL-60) using (γ-Mercaptopropyl) trimethoxysilane (MPS) as a stabilizer decorated with the thiol-modified KH1C12 aptamer. The aptamer-functionalized Fe3O4/GNCs nanoprobe proved its ability to uptake and -image HL-60 cancer cells as well as detection of cancer cells from a range of concentrations 10–200 cells μL−1 which can help in early diagnosis of lekemia. The nanoprobe's magnetic and fluorescent properties are particularly significant for MRI-based fluorescence imaging and the collection of HL-60 cancer cells, which may aid in the early detection of extremely aggressive human leukaemia [71]. Asides, AuNPS were fabricated as biosensors by Ensafi et al. by modification of a gold electrode with AuNPS coated with ZAP70 oligonucleotide probe to detect specific sequence of ZAP70 gene, which is a predictor of the IgVH mutation status and serve as prognostic indicator for distinguishing Ig-mutated from Ig-unmutated CLL. The biosensor can detect with a detection limit ZAP70 DNA sequence of 4.0 × 10−15molL−1 and a decent calibration range between 2.0 × 10–14 and 1.0 × 10−9molL−1. This biosensor is thought to be the most informative stage-independent prognostic indicator for CLL patients [72]. Another application of AuNPS was done by Mazloum-Ardakani et al. to synthesize a stable composite with electrical properties, on the surface of a biosensor by synthesizing poly (catechol) with enhanced conducting properties by modification with graphene sheets and loading on AuNPS, i.e., made up of conductive polymers and nanoparticles. With a detection limit of 1.0 pM for the DNA strand, the peak currents of the electroactive probe catechol were linearly correlated with the logarithm of the target DNA concentrations in the range of 100.0 μM to 10.0 pM. This study looked into a novel, stable composite that was used to identify AML [73].

Furthermore, Bordbar et al. utilized a different and simple technique by employing volatile organic compounds (VOCs) in blood samples as biomarkers to diagnose leukemia based on a paper-based sensor array for detecting leukemia which contains 16 NPs deposited on a sheet of hydrophobic paper in a 4 × 4 array format. The diagnosis was made by comparing the image of sensors recorded by a scanner before and after exposing to the blood vapour. The accuracy of the rock curve in differentiating patients from the control group was 96%. According to the logistic regression model, this approach properly categorised 93.6% of healthy individuals and 93.2% of patients. In addition, for every 20 units rise in overall response, the likelihood of being classified as a patient fell by 10%. With improved sensitivity and specificity, it is a low-cost, non-invasive method of identifying new leukaemia cases [74]. Recently, Wang et al. have reviewed the use of iron oxides NPs which have gained increasing attention in diagnosis and treatment of leukemia. The ability to detect magnetic resonance imaging (MRI) as effective contrast agents, magnetic biosensors, and targeted administration of anti-leukemia medications by coating various targeting moieties are among the benefits of iron oxide nanoparticles [75].

Nanocarriers for efficient delivery of anti-leukemia drugs

The application of NPs and nanocarriers has improved the delivery of chemotherapeutic agents, primarily to lessen their toxicity to normal tissue. Other benefits include improving the bioavailability, reducing side effects, solubility, blood circulation time. In addition, anti-tumor drug candidates were found to be improved by nano-delivery systems, which also use targeting or sensing techniques to release medications selectively at specific locations [61, 64, 76]. The same is true for anti-leukemia drugs, which is why many research have looked into different ways to distribute them using nanocarriers.

Polymeric NPs are one example of nanocarriers widely used by researchers for loading of drugs due to their versatility and potential low toxicity [77]. Simon et al. loaded all-trans retinoic acid (ATRA) in poly-D, L-lactadie-co-glycolide (PLGA) polymeric NPs for treatment of acute promyelocytic leukemia. This strategy allowed ATRA to be administered intravenously instead of orally which is valuable for patients who have difficulties in swallowing, which is common in case for leukemia patients [78]. PLGA was also utilized by Leung et al. for encapsulation of curcumin (Cur-PLGA NP) using a microfluidic-assisted nano-precipitation surfactant free technique that showed excellent colloidal and drug stability as well as a dose-dependent cytotoxicity in leukemia Jurkat cells without altering the viability of fibroblast NIH3T3 cells [79]. Hasandoost et al. prepared imatinib mesylate-loaded polybutylcyanoacrylate (PBCA) NPs which enhanced the cytotoxic effects of imatinib mesylate compared to free drug alone. The miniemulsion polymerization process was used to create the formulation demonstrating that the drugs were physically entrapped by nanoscale particles with a high encapsulation efficiency of 86% [80].

Combined therapy is often used in leukemia treatment to avoid resistance to treatment. Cortese et al. research team focused on this strategy and benefited from NPs ability to load several drug molecules by formulating a poly (ε-caprolactone) (PCL) system with biodegradable pH-sensitive core for nilotinib release and an enzymatic sensitive outer shell for imatinib release which allowed for combined therapy of both drugs. This system showed better cytotoxicity to leukemia cells compared to free drug [81]. The same team used the PCL loaded NPs with imatinib in combination with doxorubicin loaded sensitive polyelectrolyte complexes (PECs) which allowed a down-regulation of BCR–ABL in a sustained manner as well as significant CML stem cell death as both drugs worked in a synergistic manner with maximized cytotoxicity with minimized probability of cell resistance to any of these drugs [82]. The same research team also utilized PCL NPs to encapsulate RNA–protamine complex encapsulated for the intracellular delivery of mRNA molecules. These NPs had a core–shell structure with an mRNA-containing inner core surrounded by PCL layers which increased the in vivo stability of the mRNA and allowing their efficient delivery [83].

The loading of SiRNA was attempted by Valencia-Serna et al. using lipopolymers, namely, α-linolenic acid (ALA) substitution on low molecular weight polyethyleneimine (PEI) to deliver specific siRNAs to CML cells for therapeutic gene silencing of BCR–ABL gene. The results showed that siRNA/PEI–ALA NPs successfully allowed the silencing of the BCR–ABL gene and BCR–ABL protein, thereby decreasing the growth on CML K562 cells in vitro and arresting the growth of localized tumors in a CML mouse model [84].

Inorganic NPs have been widely exploited as nanocarriers for leading of ant-leukemic drugs. Mesoporous silica NPs were coated with folic acid PEG–lipid bilayer membrane loaded with paclitaxel and tanshinone by Li et al. It allowed a sustained release of both drugs with enhanced uptake of the NPs by NB4 cells due to the presence of folic acid leading to targeting of folic acid receptors on NB4 cells [85]. Drug resistance was diminished by Deng et al. through combined delivery of AS1411, doxorubicin and anti-221 via multifunctional AuNPS which significantly decreased the proliferation and clonogenic potential as well as facilitating the apoptosis of drug resistant leukemia cells. These systems also showed marked down regulation of miR-221 and DNMT1, leading to restoring the expression of p27kip1 and p15ink4b tumor suppressors as well as miR-221-mediated decrease of P-glycoprotein (P-gp) expression thereby providing a more efficient treatment [86]. AuNPS were also utilized by Petrushev et al. to conjugate TKIs that inhibit FLT3 kinase thereby targeting the FLT#3 mutation causing relapse in AML patients. The new prepared bioconjugate had better therapeutic effect that TKI alone as well as effective trans-membrane delivery [87].

Carbon-based NPs have been utilized due to their biocompatibility and high loading efficiencies. Shahiri.et al. synthesized multi-walled carbon nanotubes (MWCNTs) coated with silver NPs (Ag-NPs) which showed efficacy as antioxidant and cytotoxic agents against AML and acute T-cell leukemia cell lines in a dose depended manner [88]. Moreover, Felix et al. formulated imatinib loaded quantum dots, which graphene NPs are known for their safety and internalization efficiency in the cells. The results of this research confirmed the ability of the quantum dots loaded with Imatinib to sufficiently internalize and induce apoptosis and killing of cancer cells [89]. Quantum dots were also used by Esmaeili et al. as zinc oxide quantum dot NPs (ZnO QD NPs) as nanofluid (NF) to treat acute promyelocytic leukemia (APL)-derived NB4 cells. The resultant NPs successfully lowered the proliferation of NB4 cells through p21-mediated G1 cell cycle arrest and enabled their apoptosis via reactive oxygen species (ROS)-dependent up regulation of FoxO3a and SIRT1 as well as peroxisome proliferator-activated receptors gamma (PPARγ) up regulation in NB4 cells leading to elevated expression of pro-apoptotic molecules in NB4 cells [90].

Furthermore, Anu et al. synthesized selenium nanoparticles (SeNPs) as green biogenesis NPs from Cassia auriculata and shown their effectiveness against leukemia cancer cells in vitro, as well as greater solubility and decreased toxicity on normal cells [91]. Niosomal formulations to encapsulate ifosfamide, alkylating anticancer drug, was developed to control its release and alleviate its toxicity [92]. Nanocarriers for efficient delivery of anti-leukemia drugs are summarized in Table 2.

Table 2 Nanocarriers for efficient delivery of anti-leukemia drugs

Novel and emerging nanoparticles for targeted treatment

Several efforts have focused on the development of approaches that allowed targeted delivery of chemotherapeutic drugs as most of these drugs lack selectivity which cause major side-effects as well as lower effectiveness in many patients. Thus, the need arises to smart drug formulations that deliver the drugs selectively to cancer cells, thereby enhancing drug efficacy and reducing nonspecific cytotoxic effects to normal cells [59, 94]. The novel and emerging NPs for targeted treatment of leukemia are summarized in Table 3.

Table 3 Novel and emerging nanoparticles for targeted treatment of leukemia

One strategy of targeting is to prepare or coat the NPs with a specific target for an over-expressed receptor on a cancer cell. Sun et al. developed curcumin loaded hyaluronic acid-modified liposomes (HA-Cur-LPs) which targeted the CD44 receptor and consequently curcumin is targeted to these cells with high stability as CD44 is overexpressed on the surface of leukemia cells. HA-Cur-LPs also provided mice with longer survival time compared to free curcumin treatment [95]. Hyaluronic acid was also utilized by Zhang et al. through formulating hyaluronic acid-coated AgNPs (HA-AgNPs) via electrostatic interaction between the negatively charged HA molecules and positively charged AgNPs which exert their anti-leukemic action through ROS overproduction thereby inhibited the leukemia cells viability by specific binding of HA with CD44 receptors causing apoptosis while having decreased systemic toxicity [96].

Mesoporous silica NPs were used by Durfee et al. used to form a template surrounded by a liposome layer on them to form Protocells. These Protocells were then coated with antibiotic protein combined with biotinylated EGFR antibody. The prepared NPs-targeted leukemia cells efficiently as well as offering a targeted release of gemcitabine which prevented proliferation of myeloid leukemia stem cells and induced apoptosis [97].

Davis et al. loaded leukemia inhibitory factor (LIF) on poly (ethylene glycol)–poly (lactic acid) (PEG–PLA) NPs (NanoLIF) then modified their surface with CD11b antibody to target activated peripheral macrophage. The resultant NPs significantly decreased M1 cell proliferation over 72 h compared to free LIF [98].

Another strategy is oligopeptides and oligonucleotides which can be synthesized to target specific biomarkers or even synthesize personalized drugs for individual patients. Zhao et al. formulated an oligonucleotide single-strand DNA aptamer for targeting CD117 biomarker, which is over expressed on AML cells. Furthermore, they also designed an aptamer − methotrexate complex which could target primary AML cells without affecting the background marrow cells [99]. Fan et al. synthesized a double oligopeptide coupled NPs for targeting of osteoblastic and vascular niches in bone marrow. The prepared NPs also enhanced the stability and loading efficiency of Arsenic trioxide and targeted it to the cancer lesions eliminating primitive CML cells as well as improving arsenic trioxide protection to the original CML cells, and reducing the size of the K562–HUVEC organoid [100].

RNA-based therapies were exploited for treatment of leukemia as they can silence gene expression, express proteins, edit genes, thereby serving as a tailored treatment for specific types of leukemia; however, they suffer from poor in vivo stability and thus need to be encapsulated in NPs which have the stealth ability thereby preserving them from degradation [59]. Lee et al. loaded siRNA polyplexes on folate–PEG oligoaminoamide that protected siRNA against degradation as well as targeted its delivery to leukemia cells as it has folic acid receptor over expression. [101]. Lui et al. targeted a CRISPR/Cas9 system for treatment of CML by encapsulating a CRISPR/Cas9 plasmid (pCas9) which expresses gRNA to target the BCR–ABL gene (pCas9/gBCR–ABL) with poly(ethylene glycol)-b-poly(lactic acid-co-glycolic acid) (PEG–PLGA)-based cationic lipid-assisted polymeric NPs (CLANs). The prepared NPs were able to disrupt the CML-related BCR–ABL gene without affecting the same gene in normal cells thereby improving the survival rate in a CML mouse model [102]. AuNPS decorated with an antisense oligonucleotide were used for silencing the BCR–ABL1 chimeric gene and enhancing leukemia cell apoptosis significantly with loss of viability of imatinib-resistant K562 cells [103]. Exosomes are natural vesicles secreted by cells were utilized by Min et al. through extracting exosomes from imatinib-resistant CML cells and using them as vectors to deliver miR-365. Successful uptake of exosomes CML cells was achieved as well as successful targeted receptors on target cells, thereby accumulating miRNA in target tissues [104].

Cyclodextrin NPs were synthesized by Guo et al. and decorated with FAB antibody to target AML which specifically targets human leukemia stem cells (LSC) surface antigen IL-3 receptor α-chain. Furthermore, it had a synergistic therapeutic effect when combined the drug ara-C [105]. Dai et al. developed transferrin paclitaxel loaded lipid NPs (TPLN) which showed enhanced targeting to the HL-60 cancer cells compared to that of the paclitaxel-loaded NPs (PLN) due to the ability of transferrin to target leukemia cells leading to remarkable apoptosis of leukemia cells [106].

Abdellatif et al. utilized quantum dots, a form of carbon-based NPs to formulate amino-PEGylated quantum dots (QDs) which were succinylated using succinic anhydride and coated with octreotide (QD–OCD) to target somatostatin receptor type-II (SSTR) which is expressed in leukemia cells. QD–OCD accumulated in blood cells indicated by their fluorescence intensity due to SSTR targeting thereby showing potential for leukemia treatment [107]. In addition, Nui et al. synthesized a fluorescent lanthanide oxyfluoride nanoparticle-based multifunctional peptide drug delivery system for AML treatment via targeting mouse double minute 2 (MDM2) and/or MDMX by a dodecameric peptide antagonist (PMI) and an overexpressed cell surface receptor, CD33 through humanized monoclonal antibody. The resultant nanoparticle had no toxicity on normal cells and enabled apoptosis induction of AML cell lines and primary leukemic cells. An additional advantage of the prepared NPs is their fluorescence which facilitated real-time visualization of apoptotic events in AML cells, which is useful in tracking the response and treatment efficiency [108].

Finally, inflammation and bacterial oral infection may accompany and worsen the case of leukemia patients as a result of chemotherapy and immunosuppression. A bimetallic clusterzyme was created by Pt atom replacement in the structure of gold nanocluster (Au/Pt NCs) coupled with glucose oxidase (GOX) Au/Pt NCs@GOX). The in vivo investigations demonstrated that it is possible to successfully cure periodontitis in rats, and enhance periodontal tissue regeneration [109]. In addition, a modified photocross-linkable gelatin-based polymer (GelMA) containing cationic quaternary ammonium salt groups may be helpful in preventing accompanied infections [110]. The associated coagulation and hemostasis problems should be investigated by synthesis of biocompatible anticoagulant and hemostatic materials through surface-functionalized designs [111].

Clinical trials

Using carefully designed materials to create innovative treatments and devices that may lessen toxicity while improving treatment delivery and efficacy is known as nanotechnology in oncology. Notably, the most well-known nano-based medications, Abraxane® and Doxil®, were approved by the FDA a few years ago and have since been effectively employed in clinical settings. Currently, various nano delivery systems have undergone experimental stage and animal models. However, many factors need to be established before its successful clinical applications on humans, such as clearance, long-term toxicity, biocompatibility, selectivity, immunogenicity, pharmacokinetics and bio distribution which still lake sufficient data [112]. Nevertheless, many NPs drug products have gone successfully through clinical trials or even gained approval for marketing.

One example is Vyxeos® by Jazz pharmaceuticals which obtained FDA approval in 2017 which is a liposome injection loaded with daunorubicin and ara-C. This drug product utilized the advantage of loading several drugs in the same carrier or vesicle, offering the advantage of combined therapy which is beneficial for treatment of leukemia compared to a single drug therapy and consequently offered a significantly increased survival rate [113]. Marqibo® is another FDA approved drug product comprising sphingomyelin and cholesterol liposome NPs loaded with VCR. The formulations aim to eliminate the limitations of dosing and pharmacokinetics of conventional VCR drug products in addition to increase the circulation time, optimized delivery to target tissues and enhanced dosing concentrations without increasing toxicity. In addition, patients treated weekly with this new formula achieved complete remission of 20% with an overall response rate of 35%. This new formulation also counteracted the dose-dependent toxicity of VCR administration, such as neuropathy [114].

According to clinicaltrials.gov, current clinical trials involve a study done in University Hospital Zurich, Switzerland for the use of magnetic NPs, coated with epithelial cell adhesion molecule (EpCAM)–antibodies to target epithelial tumor cells. The study aims to see if circulating tumor cells (EpCAM or CD52 as marker) can be removed out of the blood with the help of these particles for the aim of leukemia diagnosis. Another study is conducted by AstraZeneca in phase I/II to determine the maximum tolerated dose as well as the safety, pharmacokinetics, and pharmacodynamics of AZD2811 NPs either as single therapy or in combination with other drugs for treatment of relapsed/refractory AML patients or for treatment of naïve AML patients who are not eligible for intensive induction therapy.

Currently in phase I, Memorial Sloan Kettering Cancer Centre in collaboration with National Cancer Institute (NCI) and Actinium Pharmaceuticals are conducting a clinical study on Targeted Atomic Nano-Generators (Actinium-225-Labeled Humanized Anti-CD33 Monoclonal Antibody HuM195) to determine the safety and dosing of actinium-225 when it is labelled to HuM195. The medication is acknowledged as a clinical-stage radioimmunotherapy that targets CD33 and has demonstrated single-agent activity in AML treatment. The result of this research revealed that its combination with venetoclax is a potential candidate for the treatment of patients with venetoclax-resistant AML [115].

A phase I study by Mayo clinic and national cancer institute currently investigates the safe therapeutic doses as well as the adverse effects of paclitaxel albumin–nanoparticle formulation (nab-paclitaxel) and rituximab-coated nanoparticle for treatment of unresponsive or relapsing B-cell non-Hodgkin lymphoma (NHL) in patients.

Despite of the significant advancements in the field of nanomedicine, the potential harmful effects of nanomaterials on humans remain a source for concern. Significant studies have been performed in recent years to try to understand these harmful effects, and as a result of cellular exposure to nanomaterials, a number of pathways, mechanisms, and important influencing parameters have been linked. The immunological changes brought on by these nanoparticles have also been clarified by these assessments of nanotoxicology. Understanding the process by which nanomaterials trigger immunological responses is necessary to regulate the immunomodulatory effect of these materials in applications [116].

Conclusion, limitations, and future perspectives

Frequent dosing, serious side effects, and a lack of specificity in anticancer medications are all part of the traditional chemotherapy for leukemia. Consequently, devising a plan to administer the anti-leukemic medication straight to the cancerous location. NPs have recently shown promise in the diagnosis and treatment of many forms of leukemia due to their easy manufacturing process and ability to encapsulate medications with a variety of chemical compositions. In addition to their capacity for sustained drug release and intracellular delivery, NPs for the treatment of leukemia are distinguished by their capacity to reduce the toxicity of the drug on healthy, normal tissues. Hence, to demonstrate the safety and selectivity of such nanotechnology, additional clinical trials will be necessary before it can be used to treat leukemia.

Through the use of smart NPs, the field of nanomedicine has steadily advanced over the past few decades in the accurate diagnosis and focused treatment of many cancers. However, before these advancements can completely replace traditional cancer treatment methods, such as chemotherapy and/or radiation therapy, they must first show in clinical trials that they are both safe and effective. Despite that most of these nano-formulations have shifted to target and deal with solid tumors, aiming to influence their ability to metastasize, spread, implantation, and trigger an inflammatory response. Later, novel methods for the diagnosis and therapy of some liquid tumor types, such as leukemia, which develops slowly, have also been developed. Leukemia has the ability to invade the blood and lymphatic systems, allowing it to spread throughout the body and remain localized in certain regions, such as the bone marrow. Although the potential advantages of nanotechnology are presented in the current article, expected outcomes in clinical uses have not yet been satisfactory. Furthermore, understanding the process by which nanomaterials trigger immunological responses is necessary to regulate the immunomodulatory effect of these materials in applications. Therefore, more sophisticated, and up-to-date NPs-based tactics are required to eradicate various cancer types with more efficacy, selectivity, and less toxicity supported with clinical trials.

Data availability

No data were used for the research described in the article.

References

  1. Shroff GS, et al. Leukemic involvement in the thorax. Radiographics. 2019;39(1):44–61.

    Article  PubMed  Google Scholar 

  2. Lockwood W. Leukemia: AML, CML, ALL and CLL. 2019.

  3. Sajana T, et al. Classification of leukemia patients with different clinical presentation of blood cells. Mater Today Proc. 2020. https://doi.org/10.1016/j.matpr.2020.10.619.

    Article  Google Scholar 

  4. Rosenzweig SA. Acquired resistance to drugs targeting receptor tyrosine kinases. Biochem Pharmacol. 2012;83(8):1041–8.

    Article  PubMed  CAS  Google Scholar 

  5. Gultekin M, et al. Gynecologic oncology training systems in Europe: a report from the European network of young gynaecological oncologists. Int J Gynecol Cancer. 2011. https://doi.org/10.1097/IGC.0b013e3182202d17.

    Article  PubMed  Google Scholar 

  6. Braicu C, et al. Novel insight into triple-negative breast cancers, the emerging role of angiogenesis, and antiangiogenic therapy. Exp Rev Mol Med. 2016;18:e18. https://doi.org/10.1017/erm.2016.17.

    Article  CAS  Google Scholar 

  7. Shylasree TS, Bryant A, Athavale R. Chemotherapy and/or radiotherapy in combination with surgery for ovarian carcinosarcoma. Cochrane Database Syst Rev. 2013;2013(2):cd00246.

    Google Scholar 

  8. Akbarali HI, et al. Chemotherapy induced gastrointestinal toxicities. Adv Cancer Res. 2022;155:131–66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Kubeček O, Paterová P, Novosadová M. Risk factors for infections, antibiotic therapy, and its impact on cancer therapy outcomes for patients with solid tumors. Life. 2021;11(12):1387.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Banu H, et al. Doxorubicin loaded polymeric gold nanoparticles targeted to human folate receptor upon laser photothermal therapy potentiates chemotherapy in breast cancer cell lines. J Photochem Photobiol B. 2015;149:116–28.

    Article  PubMed  CAS  Google Scholar 

  11. Jurj A, et al. The new era of nanotechnology, an alternative to change cancer treatment. Drug Des Dev Ther. 2017;11:2871.

    Article  CAS  Google Scholar 

  12. Irimie AI, et al. Novel technologies for oral squamous carcinoma biomarkers in diagnostics and prognostics. Acta Odontol Scand. 2015;73(3):161–8.

    Article  PubMed  CAS  Google Scholar 

  13. Herbrink M, et al. Variability in bioavailability of small molecular tyrosine kinase inhibitors. Cancer Treat Rev. 2015;41(5):412–22.

    Article  PubMed  CAS  Google Scholar 

  14. Vlad C, et al. Evaluation of clinical, morphopathological and therapeutic prognostic factors in rectal cancer. Experience of a tertiary oncology center. J Buon. 2015;20(1):92–9.

    PubMed  Google Scholar 

  15. Wang Y, Chen L. Quantum dots, lighting up the research and development of nanomedicine. Nanomed Nanotechnol Biol Med. 2011;7(4):385–402.

    Article  CAS  Google Scholar 

  16. Alrushaid N, et al. Nanotechnology in cancer diagnosis and treatment. Pharmaceutics. 2023;15(3):1025.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Baetke SC, Lammers T, Kiessling F. Applications of nanoparticles for diagnosis and therapy of cancer. Br J Radiol. 2015;88(1054):20150207.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Li J, et al. Development and application of nanomaterials, nanotechnology and nanomedicine for treating hematological malignancies. J Hematol Oncol. 2023;16(1):65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Wang R, et al. Advanced nanoparticles that can target therapy and reverse drug resistance may be the dawn of leukemia treatment: a bibliometrics study. Front Bioeng Biotechnol. 2022;10:1027868.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Blackburn LM, Bender S, Brown S. Acute leukemia: diagnosis and treatment. Semin Oncol Nurs. 2019. https://doi.org/10.1016/j.soncn.2019.150950.

    Article  PubMed  Google Scholar 

  21. Malard F, Mohty MJTL. Acute lymphoblastic leukaemia. Lancet. 2020;395(10230):1146–62.

    Article  PubMed  CAS  Google Scholar 

  22. Short NJ, Rytting ME, Cortes JE. Acute myeloid leukaemia. Lancet. 2018;392(10147):593–606.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Eichhorst B, et al. Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021;32(1):23–33.

    Article  PubMed  CAS  Google Scholar 

  24. Hochhaus A, et al. Chronic myeloid leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28:iv41–51.

    Article  PubMed  CAS  Google Scholar 

  25. Terwilliger T, Abdul-Hay MJ. Acute lymphoblastic leukemia: a comprehensive review and 2017 update. Blood Cancer J. 2017;7(6):e577.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Bosch F, Dalla-Favera R. Chronic lymphocytic leukaemia: from genetics to treatment. Nat Rev Clin Oncol. 2019;16(11):684–701.

    Article  PubMed  CAS  Google Scholar 

  27. Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2020 update on diagnosis, therapy and monitoring. Am J Hematol. 2020;95(6):691–709.

    Article  PubMed  CAS  Google Scholar 

  28. Eden RE, Coviello JM. Chronic myelogenous leukemia. In: StatPearls. 2020: StatPearls Publishing.

  29. Xiu Y, et al. Stabilization of NF-κB-inducing kinase suppresses MLL-AF9-induced acute myeloid leukemia. Cell Rep. 2018;22(2):350–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Balakrishnan K, et al. Hide and seek: the game between chronic lymphocytic leukaemia cells and B cell receptor signalling inhibitors. EMJ. 2017;2(1):24–30.

    Article  Google Scholar 

  31. Li XT, et al. Nanoparticle-delivered therapeutic agents targeting the tumor microenvironment for antitumor therapy. Discov Med. 2021;32(166):93–107.

    PubMed  Google Scholar 

  32. Hoellenriegel J, et al. Selective, novel spleen tyrosine kinase (Syk) inhibitors suppress chronic lymphocytic leukemia B-cell activation and migration. Leukemia. 2012;26(7):1576–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Friedman DR, et al. Perifosine treatment in chronic lymphocytic leukemia: results of a phase II clinical trial and in vitro studies. Leuk Lymphoma. 2014;55(5):1067–75.

    Article  PubMed  CAS  Google Scholar 

  34. Moia R, et al. Precision medicine management of chronic lymphocytic leukemia. Cancers. 2020;12(3):642.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Chiang MY, Radojcic V, Maillard I. Oncogenic Notch signaling in T and B cell lymphoproliferative disorders. Curr Opin Hematol. 2016;23(4):362.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Cuesta-Mateos C, et al. Monoclonal antibody therapies for hematological malignancies: not just lineage-specific targets. Front Immunol. 2018;8:1936.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Tzoneva G, Ferrando AA. Recent advances on NOTCH signaling in T-ALL. In: Radtke F, editor. Notch regulation of the immune system. Berlin: Springer; 2012. p. 163–82.

    Chapter  Google Scholar 

  38. Palomero T, Ferrando A. Therapeutic targeting of NOTCH1 signaling in T-cell acute lymphoblastic leukemia. Clin Lymphoma Myeloma. 2009;9:S205–10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Zheng R, et al. Advances of target therapy on NOTCH1 signaling pathway in T-cell acute lymphoblastic leukemia. Exp Hematol Oncol. 2020;9(1):1–9.

    Article  CAS  Google Scholar 

  40. dos Santos Guimarães I, et al. Conventional cancer treatment. In: Rangel L, editor., et al., Cancer treatment-conventional and innovative approaches. London: Intech; 2013.

    Google Scholar 

  41. Hughes E, et al. T-cell modulation by cyclophosphamide for tumour therapy. Immunology. 2018;154(1):62–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Stanczyk M, et al. Role of base-excision repair in the treatment of childhood acute lymphoblastic leukaemia with 6-mercaptopurine and high doses of methotrexate. Mutat Res. 2012;741(1–2):13–21.

    Article  PubMed  CAS  Google Scholar 

  43. Karran P, Attard N. Thiopurines in current medical practice: molecular mechanisms and contributions to therapy-related cancer. Nat Rev Cancer. 2008;8(1):24–36.

    Article  PubMed  CAS  Google Scholar 

  44. Im Kim K, et al. Combined interaction of multi-locus genetic polymorphisms in cytarabine arabinoside metabolic pathway on clinical outcomes in adult acute myeloid leukaemia (AML) patients. Eur J Cancer. 2013;49(2):403–10.

    Article  Google Scholar 

  45. Moudi M, et al. Vinca alkaloids. Int J Prev Med. 2013;4(11):1231.

    PubMed  PubMed Central  Google Scholar 

  46. Dumontet C, Jordan MA. Microtubule-binding agents: a dynamic field of cancer therapeutics. Nat Rev Drug Discov. 2010;9(10):790–803.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Fukushima T, et al. Action mechanism of idarubicin (4-demethoxydaunorubicin) as compared with daunorubicin in leukemic cells. Int J Hematol. 1993;57(2):121–30.

    PubMed  CAS  Google Scholar 

  48. Owattanapanich W, et al. Efficacy and toxicity of idarubicin versus high-dose daunorubicin for induction chemotherapy in adult acute myeloid leukemia: a systematic review and meta-analysis. Clin Lymphoma Myeloma Leuk. 2018;18(12):814-821 e3.

    Article  PubMed  Google Scholar 

  49. Trifilio SM, et al. Mitoxantrone and etoposide with or without intermediate dose cytarabine for the treatment of primary induction failure or relapsed acute myeloid leukemia. Leuk Res. 2012;36(4):394–6.

    Article  PubMed  CAS  Google Scholar 

  50. Li Y, et al. DNT cell-based Immunotherapy: progress and applications. J Cancer. 2020;11(13):3717–24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Chen B, et al. Targeting chemotherapy-resistant leukemia by combining DNT cellular therapy with conventional chemotherapy. J Exp Clin Cancer Res. 2018;37(1):1–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Elagamy A, Elghoneimy LK, Arafa RK. Pyridine ring as an important scaffold in anticancer drugs. In: Singh P, editor. Recent developments in the synthesis and applications of pyridines. Amsterdam: Elsevier; 2023. p. 375–410.

    Chapter  Google Scholar 

  53. Tura S, et al. Interferon alfa-2a as compared with conventional chemotherapy for the treatment of chronic myeloid leukemia. N Engl J Med. 1994;330(12):820–5.

    Article  PubMed  Google Scholar 

  54. Talpaz M, et al. Re-emergence of interferon-α in the treatment of chronic myeloid leukemia. Leukemia. 2013;27(4):803–12.

    Article  PubMed  CAS  Google Scholar 

  55. Köksal M, et al. Total marrow irradiation versus total body irradiation using intensity-modulated helical tomotherapy. J Cancer Res Clin Oncol. 2023;149(9):5965–73. https://doi.org/10.1007/s00432-022-04565-2.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Oertel M, Eich HT. Radiotherapy for the treatment of leukemia. Gynakologe. 2022;28(6):511–8.

    PubMed  PubMed Central  Google Scholar 

  57. Sabit H, et al. Nanocarriers: a reliable tool for the delivery of anticancer drugs. Pharmaceutics. 2022;14(8):1566.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Tian H, et al. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol. 2022;15(1):132.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Shen J, et al. Advances of nanoparticles for leukemia treatment. ACS Biomater Sci Eng. 2020;6(12):6478–89.

    Article  PubMed  CAS  Google Scholar 

  60. Rizvi SAA, Saleh AM. Applications of nanoparticle systems in drug delivery technology. Saudi Pharm J. 2018;26(1):64–70.

    Article  PubMed  Google Scholar 

  61. Khademi R, et al. Nanotechnology-based diagnostics and therapeutics in acute lymphoblastic leukemia: a systematic review of preclinical studies. Nanoscale Adv. 2023;5(3):571–95.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Cheng Z, et al. Nanomaterials for cancer therapy: current progress and perspectives. J Hematol Oncol. 2021;14(1):85.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Zhu R, et al. Current progress in cancer treatment using nanomaterials. Front Oncol. 2022;12: 930125.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Krishnan V, Rajasekaran AK. Clinical nanomedicine: a solution to the chemotherapy conundrum in pediatric leukemia therapy. Clin Pharmacol Ther. 2014;95(2):168–78.

    Article  PubMed  CAS  Google Scholar 

  65. Soni G, Yadav KS. Applications of nanoparticles in treatment and diagnosis of leukemia. Mater Sci Eng C Mater Biol Appl. 2015;47:156–64.

    Article  PubMed  CAS  Google Scholar 

  66. Tan Y, Li Y, Tang F. Nucleic acid aptamer: a novel potential diagnostic and therapeutic tool for leukemia. Onco Targets Ther. 2019;12:10597–613.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Tan J, et al. Aptamer-functionalized fluorescent silica nanoparticles for highly sensitive detection of leukemia cells. Nanoscale Res Lett. 2016;11(1):298.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Khoshfetrat SM, Mehrgardi MA. Amplified detection of leukemia cancer cells using an aptamer-conjugated gold-coated magnetic nanoparticles on a nitrogen-doped graphene modified electrode. Bioelectrochemistry. 2017;114:24–32.

    Article  PubMed  CAS  Google Scholar 

  69. Grechkin YA, et al. Aptamer-conjugated Tb(III)-doped silica nanoparticles for luminescent detection of leukemia cells. Biomedicines. 2020;8(1):14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. MacLaughlin CM, et al. Surface-enhanced raman scattering dye-labeled Au nanoparticles for triplexed detection of leukemia and lymphoma cells and SERS flow cytometry. Langmuir. 2013;29(6):1908–19.

    Article  PubMed  CAS  Google Scholar 

  71. Haghighi FH, et al. Aptamer/magnetic nanoparticles decorated with fluorescent gold nanoclusters for selective detection and collection of human promyelocytic leukemia (HL-60) cells from a mixture. Nanotechnology. 2019;31(2): 025605.

    Article  PubMed  Google Scholar 

  72. Ensafi AA, et al. A novel diagnostic biosensor for distinguishing immunoglobulin mutated and unmutated types of chronic lymphocytic leukemia. Biosens Bioelectron. 2016;77:409–15.

    Article  PubMed  CAS  Google Scholar 

  73. Mazloum-Ardakani M, et al. A new composite consisting of electrosynthesized conducting polymers, graphene sheets and biosynthesized gold nanoparticles for biosensing acute lymphoblastic leukemia. Bioelectrochemistry. 2018;121:38–45.

    Article  PubMed  CAS  Google Scholar 

  74. Bordbar MM, et al. A non-invasive tool for early detection of acute leukemia in children using a paper-based optoelectronic nose based on an array of metallic nanoparticles. Anal Chim Acta. 2021;1141:28–35.

    Article  PubMed  CAS  Google Scholar 

  75. Wang Y, et al. Application of iron oxide nanoparticles in the diagnosis and treatment of leukemia. Front Pharmacol. 2023;14:1177068.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Mitchell MJ, et al. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2020;20(2):101–24. https://doi.org/10.1038/s41573-020-0090-8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Abdelkader DH, et al. Insight into fucoidan-based PEGylated PLGA nanoparticles encapsulating methyl anthranilic acid: in vitro evaluation and in vivo anti-inflammatory study. Mar Drugs. 2022;20:694. https://doi.org/10.3390/md20110694.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Simon AM, et al. Poly (D, L-lactic-co-glycolide) nanoparticles for the improved therapeutic efficacy of all-trans-retinoic acid: a study of acute myeloid leukemia (AML) cell differentiation in vitro. Med Chem. 2012;8(5):805–10.

    Article  PubMed  CAS  Google Scholar 

  79. Leung MHM, Shen AQ. Microfluidic assisted nanoprecipitation of PLGA nanoparticles for curcumin delivery to leukemia jurkat cells. Langmuir. 2018;34(13):3961–70.

    Article  PubMed  CAS  Google Scholar 

  80. Hasandoost L, et al. In vitro effect of imatinib mesylate loaded on polybutylcyanoacrylate nanoparticles on leukemia cell line K562. Artif Cells Nanomed Biotechnol. 2017;45(3):665–9.

    Article  PubMed  CAS  Google Scholar 

  81. Cortese B, D’Amone S, Palama IE. Wool-like hollow polymeric nanoparticles for CML chemo-combinatorial therapy. Pharmaceutics. 2018;10(2):52.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Palamà IE, et al. Coupled delivery of imatinib mesylate and doxorubicin with nanoscaled polymeric vectors for a sustained downregulation of BCR-ABL in chronic myeloid leukemia. Biomater Sci. 2015;3(2):361–72.

    Article  PubMed  Google Scholar 

  83. Palamà IE, et al. mRNA delivery using non-viral PCL nanoparticles. Biomater Sci. 2015;3(1):144–51.

    Article  PubMed  Google Scholar 

  84. Valencia-Serna J, et al. siRNA/lipopolymer nanoparticles to arrest growth of chronic myeloid leukemia cells in vitro and in vivo. Eur J Pharm Biopharm. 2018;130:66–70.

    Article  PubMed  CAS  Google Scholar 

  85. Li Z, et al. Folic acid modified lipid-bilayer coated mesoporous silica nanoparticles co-loading paclitaxel and tanshinone IIA for the treatment of acute promyelocytic leukemia. Int J Pharm. 2020;586: 119576.

    Article  PubMed  CAS  Google Scholar 

  86. Deng R, et al. Multifunctional gold nanoparticles overcome microRNA regulatory network mediated-multidrug resistant leukemia. Sci Rep. 2019;9(1):5348.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Petrushev B, et al. Gold nanoparticles enhance the effect of tyrosine kinase inhibitors in acute myeloid leukemia therapy. Int J Nanomedicine. 2016;11:641–60.

    PubMed  PubMed Central  CAS  Google Scholar 

  88. Shahriari M, et al. Decoration of silver nanoparticles on multi-walled carbon nanotubes: Investigation of its anti-acute leukemia property against acute myeloid leukemia and acute T cell leukemia. Appl Organ Chem. 2020;34(4):e5476.

    Article  CAS  Google Scholar 

  89. Felix DM, et al. Graphene quantum dots decorated with imatinib for leukemia treatment. J Drug Deliv Sci Technol. 2020;61: 102117.

    Article  Google Scholar 

  90. Esmaeili S, et al. ZnO Q-dots-induced apoptosis was coupled with the induction of PPARγ in acute promyelocytic leukemia cells; proposing a novel application of nanoparticles in combination with pioglitazone. J Cluster Sci. 2021. https://doi.org/10.1007/s10876-021-01992-y.

    Article  Google Scholar 

  91. Anu K, et al. Biogenesis of selenium nanoparticles and their anti-leukemia activity. J King Saud Univ Sci. 2020;32(4):2520–6.

    Article  Google Scholar 

  92. Hajinezhad MR, et al. Development of a new vesicular formulation for delivery of Ifosfamide: evidence from in vitro, in vivo, and in silico experiments. Arab J Chem. 2023;16(9): 105086.

    Article  CAS  Google Scholar 

  93. Miao Y, Mahdavi B, Zangeneh MM. Anti-leukemia properties of cadmium nanoparticles in vitro and in vivo conditions: a chemobiological study. Arch Med Sci. 2021. https://doi.org/10.5114/aoms/142465.

    Article  Google Scholar 

  94. Haggag YA, et al. Fucoidan in pharmaceutical formulations: a comprehensive review for smart drug delivery systems. Mar Drugs. 2023;21:112. https://doi.org/10.3390/md21020112.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Sun D, et al. Novel curcumin liposome modified with hyaluronan targeting CD44 plays an anti-leukemic role in acute myeloid leukemia in vitro and in vivo. ACS Appl Mater Interfaces. 2017;9(20):16857–68.

    Article  PubMed  CAS  Google Scholar 

  96. Zhang J, et al. Anti-leukemia activity of hyaluronic acid coated silver nanoparticles for selective targeting to leukemic cells. J Biomater Tissue Eng. 2018;8(6):906–10.

    Article  CAS  Google Scholar 

  97. Durfee PN, et al. Mesoporous silica nanoparticle-supported lipid bilayers (protocells) for active targeting and delivery to individual leukemia cells. ACS Nano. 2016;10(9):8325–45.

    Article  PubMed  CAS  Google Scholar 

  98. Davis SM, et al. Leukemia inhibitory factor-loaded nanoparticles with enhanced cytokine metabolic stability and anti-inflammatory activity. Pharm Res. 2018;35(1):6.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Zhao N, et al. Oligonucleotide aptamer-drug conjugates for targeted therapy of acute myeloid leukemia. Biomaterials. 2015;67:42–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Fan L, et al. Dual oligopeptides modification mediates arsenic trioxide containing nanoparticles to eliminate primitive chronic myeloid leukemia cells inside bone marrow niches. Int J Pharm. 2020;579: 119179.

    Article  CAS  Google Scholar 

  101. Lee D-J, et al. Systemic delivery of folate-PEG siRNA lipopolyplexes with enhanced intracellular stability for in vivo gene silencing in leukemia. Bioconjug Chem. 2017;28(9):2393–409.

    Article  PubMed  CAS  Google Scholar 

  102. Liu Y, et al. Systemic delivery of CRISPR/Cas9 with PEG-PLGA nanoparticles for chronic myeloid leukemia targeted therapy. Biomater Sci. 2018;6(6):1592–603.

    Article  PubMed  CAS  Google Scholar 

  103. Vinhas R, Fernandes AR, Baptista PV. Gold nanoparticles for BCR-ABL1 gene silencing: improving tyrosine kinase inhibitor efficacy in chronic myeloid leukemia. Mol Ther Nucleic Acids. 2017;7:408–16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Min QH, et al. Exosomes derived from imatinib-resistant chronic myeloid leukemia cells mediate a horizontal transfer of drug-resistant trait by delivering miR-365. Exp Cell Res. 2018;362(2):386–93.

    Article  PubMed  CAS  Google Scholar 

  105. Guo J, et al. Antibody-targeted cyclodextrin-based nanoparticles for siRNA delivery in the treatment of acute myeloid leukemia: physicochemical characteristics, in vitro mechanistic studies, and ex vivo patient derived therapeutic efficacy. Mol Pharm. 2017;14(3):940–52.

    Article  PubMed  CAS  Google Scholar 

  106. Dai Y, et al. Antiproliferative and apoptosis triggering potential of paclitaxel-based targeted-lipid nanoparticles with enhanced cellular internalization by transferrin receptors-a study in leukemia cells. Nanoscale Res Lett. 2018;13(1):271.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Abdellatif AAH, et al. Fluorescent nanoparticles coated with a somatostatin analogue target blood monocyte for efficient leukaemia treatment. Pharm Res. 2020;37(11):217.

    Article  PubMed  CAS  Google Scholar 

  108. Niu F, et al. Lanthanide-doped nanoparticles conjugated with an anti-CD33 antibody and a p53-activating peptide for acute myeloid leukemia therapy. Biomaterials. 2018;167:132–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Wang Y, et al. Ultra-small Au/Pt NCs@GOX clusterzyme for enhancing cascade catalytic antibiofilm effect against F. nucleatum-induced periodontitis. Chem Eng J. 2023;466:143292.

    Article  CAS  Google Scholar 

  110. Liu Y, et al. Biodegradable and cytocompatible hydrogel coating with antibacterial activity for the prevention of implant-associated infection. ACS Appl Mater Interfaces. 2023;15(9):11507–19.

    Article  PubMed  CAS  Google Scholar 

  111. Wang Y, et al. Surface-functionalized design of blood-contacting biomaterials for preventing coagulation and promoting hemostasis. Friction. 2023;11(8):1371–94.

    Article  CAS  Google Scholar 

  112. Chen H, et al. Rethinking cancer nanotheranostics. Nat Rev Mater. 2017;2:17024.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Bulbake U, et al. Liposomal formulations in clinical use: an updated review. Pharmaceutics. 2017;9(2):12.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Silverman JA, Deitcher SR. Marqibo(R) (vincristine sulfate liposome injection) improves the pharmacokinetics and pharmacodynamics of vincristine. Cancer Chemother Pharmacol. 2013;71(3):555–64.

    Article  PubMed  CAS  Google Scholar 

  115. Garg R, et al. 225Ac-labeled CD33-targeting antibody reverses resistance to Bcl-2 inhibitor venetoclax in acute myeloid leukemia models. Cancer Med. 2021;10(3):1128–40.

    Article  PubMed  CAS  Google Scholar 

  116. Lenders V, et al. Biomedical nanomaterials for immunological applications: ongoing research and clinical trials. Nanoscale Adv. 2020;2(11):5046–89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).

Author information

Authors and Affiliations

Authors

Contributions

Writing—original draft preparation, MMS, NMA, NME, and SME; writing—review and editing AZ and SME; Data collection and analysis: AZ and SME. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Shahira M. Ezzat.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Salama, M.M., Aborehab, N.M., El Mahdy, N.M. et al. Nanotechnology in leukemia: diagnosis, efficient-targeted drug delivery, and clinical trials. Eur J Med Res 28, 566 (2023). https://doi.org/10.1186/s40001-023-01539-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-023-01539-z

Keywords