Nanotechnology applications in hematological malignancies (Review)

  • Authors:
    • Ahmed Samir
    • Basma M. Elgamal
    • Hala Gabr
    • Hatem E. Sabaawy
  • View Affiliations

  • Published online on: July 2, 2015     https://doi.org/10.3892/or.2015.4100
  • Pages: 1097-1105
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A major limitation to current cancer therapies is the development of therapy-related side-effects and dose limiting complications. Moreover, a better understanding of the biology of cancer cells and the mechanisms of resistance to therapy is rapidly developing. The translation of advanced knowledge and discoveries achieved at the molecular level must be supported by advanced diagnostic, therapeutic and delivery technologies to translate these discoveries into useful tools that are essential in achieving progress in the war against cancer. Nanotechnology can play an essential role in this aspect providing a transforming technology that can translate the basic and clinical findings into novel diagnostic, therapeutic and preventive tools useful in different types of cancer. Hematological malignancies represent a specific class of cancer, which attracts special attention in the applications of nanotechnology for cancer diagnosis and treatment. The aim of the present review is to elucidate the emerging applications of nanotechnology in cancer management and describe the potentials of nanotechnology in changing the key fundamental aspects of hematological malignancy diagnosis, treatment and follow-up.

1. Introduction to nanotechnology

Nanotechnology is the management of material properties at nanometer levels. The concept of nanotechnology was first introduced by the American physicist Richard Feynman who gave a talk entitled 'There's Plenty of Room at the Bottom' at an American Physical Society meeting at Caltech in 1959. Feynman did not mention 'nanotechnology', yet considered the potential to manipulate individual atoms at the molecular level. This talk inspired the conceptual beginning for nanotechnology science through creating the concept of 'molecular manufacturing' (1). The prefix 'nano' derives from the Greek word 'nanos' which means 'a dwarf'. A nanometer (nm) measures one-billionth of a meter. Many biological structures are within the nanometer size. While a single RBC is 2,500 nm, a DNA molecule is 2.5 nm, and while a protein molecule is 1–20 nm, the biggest amino acid (tryptophan) is 1.2 nm (2).

Nanotechnology is defined as the creation and use of structures, materials, devices and systems with completely novel properties and functions obtained from their size which ranges from 1 to 100 nm. It is applied in many fields including electronics, information technology, material developments and biomedicine (3). Nanobiotechnology represents a specific category of nanotechnology, based on biological molecules and processes that involve the creation of devices and systems at the nanolevel to examine or control biological processes (4,5).

Cancer nanotechnology is a striving field (6), current methods for cancer diagnosis have limitations regarding the sensitivity and the ability to detect cancer at early stages (7). In addition, conventional treatment options have considerable side-effects due to non-target tissue toxicity and their success is limited in advanced disease stages due to the development of drug resistance. An additional diagnostic and follow-up problem is related to the detection of minimal residual diseases (MRD) after treatment (7). Therefore, there is an urgent need to develop novel therapies and cell type-specific delivery systems to deliver anticancer therapies more effectively. Hematological malignancies representing cancers that affect blood, bone marrow and lymph nodes, are associated with these same problems encountered with other types of cancer. Nanotechnology may find novel solutions to all of these problems. New nanodevices can perform different key functions such as detecting cancer cells at the initial stages and determining their location in the body. Nanotechnology, either alone or in combination with traditional diagnostic methods, provides new sensitive, specific, reproducible and cost-effective methods for diagnosis of different types of cancers including hematological malignancies. In the field of cancer therapy, nanodevices are used to carry drugs to specific target cells and tumor sites, with nanoparticle-based drug delivery approaches making remarkable success in site-specific release of different therapies due to their unique physical, chemical and biological features.

The advantages that nanotechnology offers include targeted delivery of drugs, reduced dosage, reduced frequency of dosing, better solubility, reduced immunogenicity and better half-life of these drugs when used in vivo. Multiple types of nanomaterials can be used to generate the nanoscale drug delivery systems including lipid-based nanocapsules or liposomes and natural or synthetic polymers. Liposomes are biocompatible vesicles formed of amphipathic lipid bilayer membranes. Polymers could be derived from biocompatible drug conjugates or micelles that self-assemble to form a hydrophilic outer corona and a hydrophobic inner core for drug encapsulation. The choices of different nanoscale systems are based on the components, applications, advantages and limitations of each preparation. These novel nanotherapeutics are rapidly progressing as they offer solutions to many limitations of conventional drug delivery systems such as nonspecific targeting, limited biodistribution, poor bioavailability and low therapeutic indices. Factors that determine nanomaterial design and characterization include size and shape, blood half-life, controlled drug release and active targeting of nanoparticles. Efficient cell uptakes were reported with nanoparticles with a size range of 40–50 nm (8). Studies examining the effects of nanoparticle shapes demonstrated that rod-shaped micelles continue in the circulation 10 times longer than spherical micelles (9), while these spherical micelles bind more tightly to circulating and leukemic cells (10). Moreover, nanoscale formulations can be rendered multifunctional with the concept of combination therapy in practice. In the following section, a number of applications of nanotechnology in hematological malignancies will be discussed with a particular emphasis on the utilization of nanotechnology in diagnosis, treatment and follow-up of different types of leukemias and lymphomas.

2. Nanotechnology applications in different types of leukemia

Nanotechnology applications in acute leukemias

Acute leukemias represent a group of cancers caused by malignant transformation of hematopoietic cells and usually associated with maintaining a partial capacity of differentiation of the progenitor tumor initiating cells (11). In general, acute leukemias are divided into acute myeloid (also called non-lymphocytic) leukemia (AML) (12) and acute lymphoid leukemia (ALL) (lymphoblastic leukemia/lymphoma) (13). ALL has a higher incidence in children, while AML usually predominates in adults, yet age profiles overlap. Sex distribution for AML is almost equal until the age of 60, after which a considerable male predominance occurs. The following section will discuss the different diagnostic and therapeutic applications of nanotechnology in acute leukemia.

Aptamer-conjugated nanoparticles for selective detection of leukemic cells

Diagnosis and classification of acute leukemias depend on integration of information from morphologic and cytochemical examination along with data obtained from immunophenotyping analysis using flow cytometry (FCM) and cytogenetic or molecular analyses of diagnostic genetic mutations associated with prognosis (14). As antibodies used in FCM may not detect all the molecular events associated with the development of malignancy or other factors that may be useful for clinical management, the study of cell surface proteins with nanotechnology may improve treatment strategies. Creating probes to detect surface proteins can be used to classify tumors depending on the molecular features of these cells rather than their tissues of origin (15). In this regard, using molecular aptamers is a new strategy of identifying novel biomarkers that can be used to increase effectiveness of therapy and survival of leukemic patients (16). Aptamers are synthetic nucleic acid ligands that can be produced against many targets including proteins, drugs and amino acids (17). Compared with antibodies, aptamers have several advantages. To name a few, aptamers have a higher affinity and are easily synthesized with limited toxicity. In addition, aptamers can also fold into three-dimensional conformations with unique ability to bind biomolecular ligands (18). Making use of these advantages, Herr et al (19) have chosen an aptamer for acute leukemic cells with a specific sequence. In this study, they created an assay using aptamer-conjugated nanoparticles for quick recognition of acute leukemic cells using high-affinity DNA aptamers for signal detection. An oligonucleotide of 88 base pairs, which has the capability of binding to acute leukemic cells was attached to magnetic and fluorescent nanoparticles. The fluorescent nanoparticles then provided a method of detection, while the use of magnetic nanoparticles eliminated the need for centrifugation and helped the removal of unwanted particles.

Enhanced leukemic cell detection using a novel magnetic needle and nanoparticles

Detection of MRD in leukemia is essential for follow-up of patients in order to change treatment strategies. Although the utilization of morphological examination of blood and bone marrow for detection of MRD has decreased, being replaced by more sensitive techniques, it is still being used as one of the easiest methods for detection of MRD (20). Morphological examination of peripheral blood and bone marrow has been used for years for detection of MRD. However, microscopic diagnosis is subject to the examiner's interpretations and depends on the experience of the hematologist, therefore distinguishing leukemic cells from normal lympho-hematopoietic progenitors can be challenging (21). Traditional methods for detection of MRD include FCM, polymerase chain reaction (PCR) as well as immunoglobulin (Ig) and T-cell receptor (TCR) gene rearrangements. These methods have a detection limit of 0.001% leukemic cells therefore decreasing dependence on morphological examination for detection of MRD (22). In order to improve the sensitivity of detection of leukemic cells in the bone marrow, Jaetao et al (23) developed a new device that uses antibodies coupled to superparamagnetic iron oxide nanoparticles (SPION) that were directed against the acute leukemia antigen CD34 linked with a magnetic needle biopsy. CD34-conjugated nanoparticles bound highly to CD34 expressing cell lines. In addition, the magnetic needle allowed the recognition of cells from cell lines and patient leukemic cells diluted into normal blood at low concentrations. They also observed that the magnetic needle improved the percentage of blasts visible by light microscopy by 10-fold indicating that using this needle may improve the detection levels for MRD (23).

Therapeutic nanotechnology approaches in acute leukemia

After approval of Doxil, a pegylated liposomal doxorubicin by the US Food and Drug Administration (FDA) in 1995 for treatment of metastatic ovarian cancer and AIDS-related Kaposi sarcoma, several other drugs with similar features have been developed and were either approved or in different phases of clinical trials (24). Nanotechnology has a high potential for improving current therapeutic approaches in leukemia. In 2012, vincristine sulfate liposome injection (VSLI) was approved by FDA for treatment of adult patients with Philadelphia chromosome-negative ALL when those patients are in relapse or have progressive disease after two or more anti-leukemia therapies (25). Several other approaches have also been investigated. One of the approaches tested by Krishnan et al (26) was the incorporation of dexamethasone, which is commonly used for treatment of childhood leukemia, in polymeric NPs and testing the ability of the conjugate against leukemic cells in vitro and in vivo. The cytotoxic activity of the complex was similar to that of free dexamethasone. In vivo models have shown that the complex enhanced the quality of life of mice in comparison with dexamethasone alone and that NPs were cleared from the body with time, providing evidence for the potential use of this method to deliver dexamethasone in lower doses and more efficient forms. Another approach is incorporating different compounds into lipid nanoparticles. Lipid nanoparticles have many advantages as drug carriers being biocompatible, biodegradable, with a low toxicity and able to incorporate different drugs types. This approach was tested by Rahman et al (27) who loaded zerumbone, a natural dietary compound with an antitumor effect and a low water solubility, into nano-structured lipid carriers. The activity of the formulation was tested in human T-cell ALL cell lines and has shown a sustained release and a highly effective killing effect against these ALL cells.

Conjugation of drugs with colloids was also investigated. Cosco et al (28) investigated the effect of linking cytarabine (Ara-C) with one product of squalene which is a natural product used as a precursor in the synthesis of sterols. The product was assessed against different cancer cell lines (L1210, K562 and MCF-7) and has shown an effect against resistant leukemic cells (L1210R) compared with the naked compound as the conjugate protected the drug from cellular metabolic degradation. These results were also confirmed by an in vivo leukemic model in which mice treated with the conjugate had a better survival rate, than those treated with Ara-C alone.

Nanotechnology for reversal of multidrug resistance (MDR) in leukemia

A major problem that represents an obstacle to the success of treatment of leukemia is the development of MDR, which is responsible for ~90% of cancer treatment failure (29). As MDR develops, cancer cells become resistant to the cytotoxic effects of chemotherapeutic agents (29). Using higher doses of these chemotherapies does not overcome the effects of MDR. On the contrary, it is frequently associated with several toxic effects creating a need for the development of a new approach combining conventional methods with new strategies to increase the delivery and concentration of drugs in target tissues (30). Numerous mechanisms have been implicated in the development of drug resistance in patients with AML. One of the common mechanisms is the development of efflux pumps, such as P-glycoprotein and MDR-associated protein (MRP) (31). An approach that may be used to overcome MDR is utilizing antibody drug conjugates (ADCs) by linking an antibody (or an antibody fragment) to a cytotoxic drug to improve the anticancer effect of antibodies and decrease the toxicity of the conjugated drugs (32).

Chen et al (33) prepared Fe3O4-MNPs (magnetic nanoparticles) loaded with adriamycin and tetrandrine to interfere with MDR of K562/A02 cells. Measurement of several biological parameters has shown that adriamycin- and tetrandrine-loaded Fe3O4-MNPs can help in counteracting the effects of MDR in K562/A02 cells, due to the buildup of drugs caused by its polymerization with Fe3O4-MNPs. In another study, Chen et al (34) synthesized tetraheptylammonium-capped MNPs-Fe3O4 causing an increase in the concentration of daunorubicin in MDR leukemia K562/A02 cells and augmenting the therapeutic effect of daunorubicin in MDR leukemic K562/A02 cells. This study also showed that utilizing daunorubicin only is less sensitive than daunorubicin associated with MNPs-Fe3O4. In vitro cytotoxicity assays provided evidence that MNPs-Fe3O4s are biocompatible materials. Combination of Fe3O4-MNP with other therapeutic forms can also represent an alternative strategy to overcome MDR in leukemia. Magnetic drug targeting (MDT) is a novel approach developed to increase the concentration of anticancer drugs in the tumor and decrease the amount of leakage into the surrounding healthy tissues (35). Ren et al (3) investigated this approach and combined Fe3O4-MNP with hyperthermia and chemotherapy in nude mice harboring tumor xenografts. Pathological examination of the tumors and markers of apoptosis were assessed. According to their results, tumors became smaller in size and apoptosis was observed in cells from the treated group, providing evidence for the benefits of using this approach to reverse MDR in leukemia.

Another promising approach to overcome MDR is the use of nanodiamonds (NDs). Several advantages of NDs favor their use in the field of medicine such as being inert, transparent, having high surface area and being biocompatible (36). NDs can be used as carriers for drugs or therapeutic nucleic acids. Man et al (37) adjusted different parameters such as pH and concentration for loading daunorubicin into NDs. The resultant complex was tested in K562 human leukemic cells, with MDR induced by gradual exposure to daunorubicin. The results showed that NDs could help the release of daunorubicin and overcome mechanisms of drug efflux, which induce resistance, therefore adding further evidence for the potential use of NDs to overcome MDR. In another study, Ghoneum et al (38) examined the effects of using a mixture of NDs and nanoplatinum (NP) known as DPV576 to overcome MDR in MDR human myeloid leukemic cells (HL60/AR) and MDR-sensitive cells (HL60). According to their results, the combination of daunorubicin with DPV576 was associated with higher incidence of the development of cell membrane holes in the MDR cells in comparison with the control cells that allowed better delivery of the drugs inside such cells.

Modifications of all-trans retinoic acid for treatment of acute promyelocytic leukemia

Acute promyelocytic leukemia (APL) is a specific subtype of AML. The hallmark of APL is the presence of specific t(15;17) chromosomal translocation. The translocation occurs between retinoic acid receptor-α (RARα) gene and the PML gene on chromosomes 17 and 15, respectively (39). APL cells are induced by all-trans retinoic acid (ATRA) to differentiate into mature myeloid cells and ATRA may induce complete remission in APL patients (40). However, several complications appear to be associated with the use of ATRA including hypertriglyceridermia, mucocutaneous dryness and headache. In addition, ATRA resistance may develop due to lower plasma concentration of the drug leading to recurrence of the disease. Kim et al (41) developed new nanoparticles based on creating a complex between low molecular weight water-soluble chitosan (LMWSC) and ATRA. LMWSC has many advantages, in addition to being water soluble, including ease of modification and potential use as a gene or drug carrier. In their study, Kim et al (41) observed that ATRA was released from the nanoparticles for 10 days and had superior effects on CT-26 colon carcinoma cells on day 1, but the same cytotoxicity on day 2. These results provide proof-of-concept of using LMWSC nanoparticles in the drug delivery field. However, overcoming treatment resistance in APL is still a challenge. Understanding the molecular mechanisms associated with resistance and development of targeted therapy could be the ultimate solution to this problem (42).

Nanotechnology applications in chronic myeloid leukemia (CML)

Due to their unique properties, gold nanoparticles (AuNPs) are among the interesting materials for use in the field of nanotechnology. AuNPs have many optical properties that make them suitable for bioimaging applications (43). AuNPs have superior absorption and scattering properties than traditional organic dyes. The size and shape of AuNPs affect their optical properties where they can absorb light from visible to the near-infrared (NIR) region raising interest for their medicinal use (44). Light absorption, scattering and emission are enhanced in AuNPs due to the interaction of light with the free electrons in the particles. Accordingly, the field of light causes oscillation of the conduction band electrons at the surface of the particles. This oscillation of free electrons in resonance with the electro-magnetic field is called the surface plasmon resonance (SPR), which is responsible for the unique properties of AuNPs (43).

AuNPs have also proven to be biocompatible, have limited toxicity and low susceptibility to photobleaching making them suitable for use in studies with human cells (45). These exceptional optical properties together with easy synthesis and functionality of AuNPs with different biomolecules allow the use of AuNPs for molecular assays and permit the detection of very low concentrations of nucleic acids, therefore providing five times higher sensitivity than fluorescence based techniques (46). Using gold nanoparticles in molecular assays can improve current technique performances or help in the development of completely new assays, which can be faster, more sensitive and less expensive. This is of great importance particularly for use in developing countries (45,47).

CML is one of the examples in which AuNPs have been used for molecular diagnosis. CML is one of the most common myeloproliferative disorders. It is a clonal disorder of hematopoietic stem cells that passes through three stages. The first phase is the chronic phase characterized by a marked increase in the total leukocytic count with predominance of progenitors and mature cells of myeloid nature. With time, the disease progresses into an accelerated and finally a blastic phase characterized by maturation arrest concomitant with the appearance of an increased number of blasts (48). Most CML patients, ~90%, have an acquired translocation between the long arms of chromosomes 9 and 22 t(9;22 q34;q11) resulting in what is known as the Philadelphia chromosome (Ph). The translocation results in the formation of fusion gene between BCR and ABL genes with production of fusion protein that has a greater tyrosine kinase activity than normal. The new gene encodes p210 BCR-ABL1 protein, instead of the p145 Abelson protein causing growth factor independence and unlimited growth of leukemic cells (49). Common procedures for the diagnosis of Philadelphia chromosome include karyotype analysis, fluorescence in-situ hybridization (FISH) as well as reverse transcriptase polymerase chain reaction analyses (RT-PCR). However, those methods are costly and time-consuming creating the need for developing methods that are sensitive and cost effective at the same time (50).

Conde et al (51) presented an Au-nanoparticle-based method for the diagnosis and quantification of the BCR-ABL fusion transcript associated with CML. The technique depends on the optical properties of AuNPs. According to the size, Au-nanoprobes absorb light in different regions of the spectrum. Au-nanoprobes of 13 nm are red and have a narrow SPR band ~520 nm. On the other hand, when aggregated Au-nanoprobes are present, the solution is blue. The colors of the solutions are compared before and after aggregation induced by salt either visually and/or using spectroscopy. As shown in Fig. 1, the existence of a complementary target does not allow aggregation and the solution appears red (SPR peak at ±520 nm), while the absence of this target allows aggregation of Au-nanoprobes, associated with alteration of color from red into blue (red-shift of the SPR peak to 600–650 nm).

The same team worked on another idea within the same scope. Baptista et al (52) have developed nanoparticles in the form of an alloy with different metal compositions and have linked them with different thiol-modified single stranded DNA (nanoprobes). This was used for simultaneous detection of multiple targets depending on the different colors detected by the different alloy composition. In this study, the targets detected were various gene products in CML; ABL, BCR and BCR-ABL fusion product allowing identification of multiple targets at the same time. The presence of BCR-ABL fusion gene transcript also provided the base for targeted therapy for CML. Imatinib mesylate (IM), a tyrosine kinase inhibitor which was introduced in 1998, acts selectively against the oncoprotein BCR-ABL with a high success rate (53). Despite major hematological and cytogenetic responses achieved by IM, resistance to IM develops in a significant number of patients, resulting in disease relapse (54). Palamà et al (55) developed new polyelectrolyte complexes to act as carriers for IM in KU812 CML cell line and CD34-positive cells collected from patients. The results of these studies have shown that the used complexes allowed longer BCR-ABL kinase inactivation even at lower doses of IM compared with the previously formed microscale formulation polyelectrolyte microcapsules, giving hope that such complexes can be used with success to overcome drug resistance and prevent relapse in CML patients (55). A major limitation to this approach is that failure of therapy in most CML cases is due to persistence of leukemic stem cells (LSCS) in which tyrosine kinase therapy (TKI) is not effective. A better understanding of the pathways regulating LSC survival and the mechanisms involved in disease progression is a major need to overcome resistance of therapy and relapse (56).

Nanotechnology applications in B-chronic lymphocytic leukemia (B-CLL)

Chronic lymphocytic leukemia (CLL) is a clonal disorder characterized by accumulation of mature-appearing lymphocytes in the peripheral blood, bone marrow, lymph nodes as well as the spleen. CLL cells are frequently monoclonal B-lymphocytes that express CD19, CD5 and CD23, with weak or no expression of surface immunoglobulin (Ig), CD20, CD79b and FMC7 (57). CLL cells are also characterized by resistance to apoptosis. Although most CLL cells are non-replicating cells in G0 phase of the cell cycle, a small percentage of cells are replicating and these account for disease progression. The arrest of cells at this stage might explain the resistance of these CLL cells to cytotoxic drugs, which work through induction of apoptosis. Bcl-2 overexpression is one of the specific genes that is overexpressed in CLL and may induce such resistance (58).

The other finding that can affect growth and development of CLL is the presence of abnormal vascularization. CLL cells manufacture and release vascular endothelial growth factor (VEGF) and express VEGF membrane receptors (VEGF-R1 and VEGF-R2). VEGF also plays a significant role in CLL cells resistance to apoptosis (59). These findings can create the base for using targeted therapy with anti-VEGF antibodies (avastin; bevacizumab) to induce apoptosis in CLL cells. It has been shown that avastin selectively induces apoptosis in CLL cell through the mitochondrial pathway of caspase activation while sparing normal lymphocytes (60). However, a limitation to such an approach is the high concentration of the antibody necessary to achieve reasonable effects (58). Mukherjee et al (58) studied the functionality of gold nanoparticles with anti-VEGF antibodies on the release of the drug. In their study, all samples examined have shown a response to gold-AbVF that was considerably superior to CLL cells exposed to only AbVF or GNP with downregulation of anti apoptotic proteins. GNP alone has shown some response with induction of apoptosis in some of these CLL cells. Phase II clinical studies conducted on 46 CLL patients concluded that while anti-VEGF therapy, including avastin, remains a viable therapy for CLL, using a single agent anti-VEGF monotherapy had limited activity in CLL patients, and combination therapy is a more feasible approach particularly for patients with relapsed/refractory CLL (61). The combination of monoclonal antibodies, such as anti-CD20 or rituximab, with other types of chemotherapy is another promising approach for treatment in CLL (62).

Another strategy, developed later by Yu et al (63), recognized that anti-CD37 monoclonal antibody immunoliposomes could be used as carriers for specific targeting of B-CLL cells. Anti-CD19 or anti-CD20 was combined with anti-CD37 to form dual immunoliposomes that were used to induce apoptosis in B-CLL cells. Their results have proven that using the dual liposomes have higher effects regarding induction of apoptosis than using either anti-CD19 immunoliposome or anti-CD20 immunoliposome suggesting that this strategy can be favorable for personalized treatment of B-CLL and B-cell malignancies in general.

3. Nanotechnology applications in lymphomas

Nanotechnology applications in Hodgkin lymphoma (HL)

CD30 is an antigen that is expressed on T-cells, activated B-cells as well as natural killer cells and has a very low expression on normal cells. Therefore, CD30 is an ideal target in classical HL. Brentuximab vedotin (BV) is a CD30-directed antibody-drug conjugate that received approval from FDA for the treatment of patients with HL after relapse (64). CD30 has also been investigated as a target for photothermal therapy using gold nanoparticles. Gold nanoparticles have several advantages that enhance their potential as important agents in nanotechnology. In addition to their absorption and scattering properties, gold nanoparticles can absorb light and switch it into heat. This property can be used to induce killing of cancer cells through protein denaturation and induction of apoptosis. Photothermal therapy also allows for monitoring of the process that eventually will lead to the death of the cancer cells (65). Accordingly, Zharov et al made use of this property for photothermal therapy of HL. They developed two gold nanoparticle-antibody conjugates, one of them was combined with an anti-CD30 receptor which binds to CD30 on the surface of L-428 Hodgkin cells and the other with an anti-CD25-receptor as a control. High killing power was achieved using appropriate doses of laser irradiation and gold concentration for gold-targeted L-428 cells with little to no effect on neighboring non-targeted cancer cells. These data further support previous findings for the potential use of gold nanoparticles as a safe modality for treatment of cancer.

Nanotechnology applications in anaplastic large cell lymphoma (ALCL)

ALCL is a type of T-cell lymphoma with aggressive features and poor prognosis. One of the main features that characterize ALCL cells is abnormal expression of the anaplastic lymphoma kinase (ALK) together with the surface expression of CD30. Based on the presence or the absence of ALK translocation, the tumor is classified into either ALK-positive or -negative-ALCL. The presence of these unique and specific features can provide the base of targeted therapy for treatment of ALCL (66). For example, BV has been approved, in addition to its role in treatment of HL, for treatment of relapsed systemic ALCL (67). Crizotinib, an ALK inhibitor, is another FDA-approved drug for treatment of ALK-positive anaplastic large-cell lymphoma. However, resistance to crizotinib may develop limiting its use for long-term therapy (68). Using an approach that combines both characteristics, Zhao et al (69) have developed an RNA aptamer, which binds only to CD30 protein in solution. The complex produced was formed by inserting an RNA-based CD30 aptamer probe and ALK siRNA into polyethyleneimine-citrate carriers in nanosize (Fig. 2). According to their results, this complex allowed silencing of the expression of ALK gene in ALCL cells, stopping the growth of these cells with induction of apoptosis.

Nanotechnology applications in mantle cell lymphoma

Mantle cell lymphoma (MCL) is a high-risk subtype of NHL characterized by an aggressive clinical course and by having the worst prognosis among B-cell NHL (70). These characters create a need for development of new agents and targets against this type of cancer. One of the agents that has attracted attention is lenalidomide, an immunomodulatory agent, that has been tested with some success for treatment of refractory MCL (71). Using high-throughput techniques can provide a better understanding of the mechanisms underlying excessive proliferation and apoptotic resistance in MCL, thus allowing the identification of new targets for therapy (72). One of these potential targets is SYK, which is one of the factors that control apoptosis. Overexpression of SYK is associated with several B-cell malignancies including MCL. Cely et al (73) designed a nanotechnology platform targeting specifically a SYK inhibitor in the MCL cells. According to their results, the developed liposomal nanoparticles (NP) loaded with a product called compound 61 (C-61) were able to induce apoptosis of the MCL cells in one day offering the base for using this therapeutic innovation against a large spectrum of lymphoid malignancies, including MCL.

4. Conclusions and future prospects

Nanotechnology represents a promising technology that can be of great value in the war against cancer. However, attention should be put towards safety issues and ethical concerns. Since particles at the nanosize have unique physical and chemical properties, care must be taken when dealing with particles or products for in vivo applications. Possible changes in the reactions of the particles with cells of the body may occur and changes in the absorption and secretion properties of such particles should be well investigated. FDA has recommended that meticulous research should be conducted in areas related to these novel properties that may cause toxicity. FDA also recommended that further research for development of devices and methods that can be used for measurement of nanoscale materials should be developed. Another important issue that is related to nanotechnology, being an emerging technology, is the ethical concern. In many cases, as science leaps forward, ethics lag behind. Part of the funding should be directed towards ethical, legal and social issues related to nanotechnology. One of the most famous examples for this readiness was during the Human Genome Project when James Watson suggested that 3–5% of the budget be devoted for study of ethical, legal and social issues.

The future prospects of nanotechnology and nanomedicine are very promising. Nanotechnology development for targeted therapy of cancer is one of the most interesting areas in research currently underway where drugs are delivered specifically to the diseased tissues or cells of interest without affecting nearby normal cells. Development of biomarkers for early detection and imaging of cancer cells is another important topic for research using nanotechnology. Scientists hope that progress in proteomics and bioinformatics can be used by nanotechnology to identify and kill malignant cells with the highest efficacy and the least possible side-effects, therefore bringing us closer to the goal of more effective cancer care.

Acknowledgments

We would like to thank Dr Joseph Bertino and Dr Robert DiPaola (Rutgers Cancer Institute of New Jersey) and Mervat El Ansary (Cairo University School of Medicine) for their helpful discussions. We also appreciate the excellent work of all the personnel at the Central Hematology Laboratory, Department of Clinical Pathology, Faculty of Medicine and National Cancer Institute, Cairo University. The authors of this review are generously supported by Rutgers Cancer Institute of New Jersey, New Jersey Health Foundation, Cairo University, and the Egyptian Academy for Scientific Research and Technology. H.E.S. is a founder and stockholder, and H.G. is a collaborator of Celvive, Inc., a company with a profile focused on stem cell therapy technologies. No products or off label use of any products of Celvive have been discussed in this review.

References

1 

Feynman R: There's plenty of room at the bottom: An invitation to enter a new field of physics. Caltech Engineering and Science. 23(5): 22–36. 1959.

2 

Jain KK: Introduction to nanomedicine. The handbook of nanomedicine. Jain KK: Hamana press; pp. 1–5. 2008, View Article : Google Scholar

3 

Ren Y, Zhang H, Chen B, Cheng J, Cai X, Liu R, Xia G, Wu W, Wang S, Ding J, et al: Multifunctional magnetic Fe3O4 nanoparticles combined with chemotherapy and hyperthermia to overcome multidrug resistance. Int J Nanomed. 7:2261–2269. 2012.

4 

Fakruddin M, Hossain Z and Afroz H: Prospects and applications of nanobiotechnology: A medical perspective. J Nanobiotechnol. 10:312012. View Article : Google Scholar

5 

Kim ES, Ahn EH, Chung E and Kim DH: Recent advances in nanobiotechnology and high-throughput molecular techniques for systems biomedicine. Mol Cells. 36:477–484. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Cai W, Gao T, Hong H and Sun J: Applications of gold nanoparticles in cancer nanotechnology. Nanotechnol Sci Appl. 1:17–32. 2008.PubMed/NCBI

7 

Bharali DJ and Mousa SA: Emerging nanomedicines for early cancer detection and improved treatment: Current perspective and future promise. Pharmacol Ther. 128:324–335. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Jiang W, Kim BY, Rutka JT and Chan WC: Nanoparticle-mediated cellular response is size-dependent. Nat Nanotechnol. 3:145–150. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Geng Y, Dalhaimer P, Cai S, Tsai R, Tewari M, Minko T and Discher DE: Shape effects of filaments versus spherical particles in flow and drug delivery. Nat Nanotechnol. 2:249–255. 2007. View Article : Google Scholar

10 

Verma A and Stellacci F: Effect of surface properties on nanoparticle-cell interactions. Small. 6:12–21. 2010. View Article : Google Scholar

11 

Olsen RJ, Chang CC, Herrick JL, Zu Y and Ehsan A: Acute leukemia immunohistochemistry: A systematic diagnostic approach. Arch Pathol Lab Med. 132:462–475. 2008.PubMed/NCBI

12 

Ferrara F and Schiffer CA: Acute myeloid leukaemia in adults. Lancet. 381:484–495. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Inaba H, Greaves M and Mullighan CG: Acute lymphoblastic leukaemia. Lancet. 381:1943–1955. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Salem DA and Abd El-Aziz SM: Flowcytometric immunopheno-typic profile of acute leukemia: Mansoura experience. Indian J Hematol Blood Transfus. 28:89–96. 2012. View Article : Google Scholar :

15 

Sefah K, Tang ZW, Shangguan DH, Chen H, Lopez-Colon D, Li Y, Parekh P, Martin J, Meng L, Phillips JA, et al: Molecular recognition of acute myeloid leukemia using aptamers. Leukemia. 23:235–244. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Yang M, Jiang G, Li W, Qiu K, Zhang M, Carter CM, Al-Quran SZ and Li Y: Developing aptamer probes for acute myelogenous leukemia detection and surface protein biomarker discovery. J Hematol Oncol. 7:52014. View Article : Google Scholar : PubMed/NCBI

17 

Mallikaratchy PR, Ruggiero A, Gardner JR, Kuryavyi V, Maguire WF, Heaney ML, McDevitt MR, Patel DJ and Scheinberg DA: A multivalent DNA aptamer specific for the B-cell receptor on human lymphoma and leukemia. Nucleic Acids Res. 39:2458–2469. 2011. View Article : Google Scholar :

18 

Lakhin AV, Tarantul VZ and Gening LV: Aptamers: Problems, solutions and prospects. Acta Naturae. 5:34–43. 2013.

19 

Herr JK, Smith JE, Medley CD, Shangguan D and Tan W: Aptamer-conjugated nanoparticles for selective collection and detection of cancer cells. Anal Chem. 78:2918–2924. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Shook D, Coustan-Smith E, Ribeiro RC, Rubnitz JE and Campana D: Minimal residual disease quantitation in acute myeloid leukemia. Clin Lymphoma Myeloma. 9(Suppl 3): S281–S285. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Campana D and Coustan-Smith E: Measurements of treatment response in childhood acute leukemia. Korean J Hematol. 47:245–254. 2012. View Article : Google Scholar

22 

Campana D: Minimal residual disease in acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program. 2010:7–12. 2010. View Article : Google Scholar

23 

Jaetao JE, Butler KS, Adolphi NL, Lovato DM, Bryant HC, Rabinowitz I, Winter SS, Tessier TE, Hathaway HJ, Bergemann C, et al: Enhanced leukemia cell detection using a novel magnetic needle and nanoparticles. Cancer Res. 69:8310–8316. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Pillai JJ, Thulasidasan AK, Anto RJ, Chithralekha DN, Narayanan A and Kumar GS: Folic acid conjugated cross-linked acrylic polymer (FA-CLAP) hydrogel for site specific delivery of hydrophobic drugs to cancer cells. J Nanobiotechnol. 12:252014. View Article : Google Scholar

25 

Davis T and Farag SS: Treating relapsed or refractory Philadelphia chromosome-negative acute lymphoblastic leukemia: Liposome-encapsulated vincristine. Int J Nanomed. 8:3479–3488. 2013.

26 

Krishnan V, Xu X, Barwe SP, Yang X, Czymmek K, Waldman SA, Mason RW, Jia X and Rajasekaran AK: Dexamethasone-loaded block copolymer nanoparticles induce leukemia cell death and enhance therapeutic efficacy: A novel application in pediatric nanomedicine. Mol Pharm. 10:2199–2210. 2013. View Article : Google Scholar

27 

Rahman HS, Rasedee A, How CW, Abdul AB, Zeenathul NA, Othman HH, Saeed MI and Yeap SK: Zerumbone-loaded nanostructured lipid carriers: Preparation, characterization, and antileukemic effect. Int J Nanomed. 8:2769–2781. 2013. View Article : Google Scholar

28 

Cosco D, Rocco F, Ceruti M, Vono M, Fresta M and Paolino D: Self-assembled squalenoyl-cytarabine nanostructures as a potent nanomedicine for treatment of leukemic diseases. Int J Nanomed. 7:2535–2546. 2012.

29 

Lai BB, Chen BA, Cheng J, Gao F, Xu WL, Ding JH, Gao C, Sun XC, Li GH, Chen WJ, et al: Daunorubicin-loaded magnetic nanoparticles of Fe3O4 greatly enhance the responses of multi-drug-resistant K562 leukemic cells in a nude mouse xenograft model to chemotherapy. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 17:345–351. 2009.PubMed/NCBI

30 

He Q, Gao Y, Zhang L, Zhang Z, Gao F, Ji X, Li Y and Shi J: A pH-responsive mesoporous silica nanoparticles-based multi-drug delivery system for overcoming multi-drug resistance. Biomaterials. 32:7711–7720. 2011. View Article : Google Scholar : PubMed/NCBI

31 

De Boer AB, De Lange EL, Van der Sandt IC and Breimer DD: Transporters and the blood-brain barrier (BBB). Int J Clin Pharmacol Ther. 36:14–15. 1998.PubMed/NCBI

32 

Cianfriglia M: Targeting MDR1-P-glycoprotein (MDR1-Pgp) in immunochemotherapy of acute myeloid leukemia (AML). Ann Ist Super Sanita. 49:190–208. 2013.PubMed/NCBI

33 

Chen B, Sun Q, Wang X, Gao F, Dai Y, Yin Y, Ding J, Gao C, Cheng J, Li J, et al: Reversal in multidrug resistance by magnetic nanoparticle of Fe3O4 loaded with adriamycin and tetrandrine in K562/A02 leukemic cells. Int J Nanomed. 3:277–286. 2008.

34 

Chen BA, Lai BB, Cheng J, Xia GH, Gao F, Xu WL, Ding JH, Gao C, Sun XC, Xu CR, et al: Daunorubicin-loaded magnetic nanoparticles of Fe3O4 overcome multidrug resistance and induce apoptosis of K562-n/VCR cells in vivo. Int J Nanomed. 4:201–208. 2009. View Article : Google Scholar

35 

Janko C, Dürr S, Munoz LE, Lyer S, Chaurio R, Tietze R, Löhneysen S, Schorn C, Herrmann M and Alexiou C: Magnetic drug targeting reduces the chemotherapeutic burden on circulating leukocytes. Int J Mol Sci. 14:7341–7355. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Zhu Y, Li J, Li W, Zhang Y, Yang X, Chen N, Sun Y, Zhao Y, Fan C and Huang Q: The biocompatibility of nanodiamonds and their application in drug delivery systems. Theranostics. 2:302–312. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Man HB, Kim H, Kim HJ, Robinson E, Liu WK, Chow EK and Ho D: Synthesis of nanodiamond-daunorubicin conjugates to overcome multidrug chemoresistance in leukemia. Nanomedicine. 10:359–369. 2014. View Article : Google Scholar :

38 

Ghoneum A, Sharma S and Gimzewski J: Nano-hole induction by nanodiamond and nanoplatinum liquid, DPV576, reverses multidrug resistance in human myeloid leukemia (HL60/AR). Int J Nanomed. 8:2567–2573. 2013. View Article : Google Scholar

39 

Grignani F, Testa U, Fagioli M, Barberi T, Masciulli R, Mariani G, Peschle C and Pelicci PG: Promyelocytic leukemia-specific PML-retinoic acid alpha receptor fusion protein interferes with erythroid differentiation of human erythroleukemia K562 cells. Cancer Res. 55:440–443. 1995.PubMed/NCBI

40 

Imaizumi M, Suzuki H, Yoshinari M, Sato A, Saito T, Sugawara A, Tsuchiya S, Hatae Y, Fujimoto T, Kakizuka A, et al: Mutations in the E-domain of RARα portion of the PML/RARα chimeric gene may confer clinical resistance to all-trans retinoic acid in acute promyelocytic leukemia. Blood. 92:374–382. 1998.PubMed/NCBI

41 

Kim DG, Jeong YI, Choi C, Roh SH, Kang SK, Jang MK and Nah JW: Retinol-encapsulated low molecular water-soluble chitosan nanoparticles. Int J Pharm. 319:130–138. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Fung TK and So CW: Overcoming treatment resistance in acute promyelocytic leukemia and beyond. Oncotarget. 4:1128–1129. 2013.PubMed/NCBI

43 

Huang X, Jain PK, El-Sayed IH and El-Sayed MA: Gold nanoparticles: Interesting optical properties and recent applications in cancer diagnostics and therapy. Nanomedicine. 2:681–693. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Choi YE, Kwak JW and Park JW: Nanotechnology for early cancer detection. Sens Basel. 10:428–455. 2010. View Article : Google Scholar

45 

Radwan SH and Azzazy HM: Gold nanoparticles for molecular diagnostics. Expert Rev Mol Diagn. 9:511–524. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Hwang SH, Im SG, Hah SS, Cong VT, Lee EJ, Lee YS, Lee GK, Lee DH and Son SJ: Effects of upconversion nanoparticles on polymerase chain reaction. PLoS One. 8:e734082013. View Article : Google Scholar : PubMed/NCBI

47 

Gormally E, Vineis P, Matullo G, Veglia F, Caboux E, Le Roux E, Peluso M, Garte S, Guarrera S, Munnia A, et al: TP53 and KRAS2 mutations in plasma DNA of healthy subjects and subsequent cancer occurrence: A prospective study. Cancer Res. 66:6871–6876. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Silver RT, Woolf SH, Hehlmann R, Appelbaum FR, Anderson J, Bennett C, Goldman JM, Guilhot F, Kantarjian HM, Lichtin AE, et al: An evidence-based analysis of the effect of busulfan, hydroxyurea, interferon, and allogeneic bone marrow transplantation in treating the chronic phase of chronic myeloid leukemia: Developed for the American Society of Hematology. Blood. 94:1517–1536. 1999.PubMed/NCBI

49 

Shet AS, Jahagirdar BN and Verfaillie CM: Chronic myelogenous leukemia: Mechanisms underlying disease progression. Leukemia. 16:1402–1411. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Gabert J, Beillard E, van der Velden VH, Bi W, Grimwade D, Pallisgaard N, Barbany G, Cazzaniga G, Cayuela JM, Cavé H, et al: Standardization and quality control studies of 'real-time' quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia - a Europe Against Cancer program. Leukemia. 17:2318–2357. 2003. View Article : Google Scholar : PubMed/NCBI

51 

Conde J, Doria G and Baptista P: Noble metal nanoparticles applications in cancer. J Drug Deliv. 2012:7510752012. View Article : Google Scholar

52 

Baptista PV, Doria G, Quaresma P, Cavadas M, Neves CS, Gomes I, Eaton P, Pereira E and Franco R: Nanoparticles in molecular diagnostics. Prog Mol Biol Transl Sci. 104:427–488. 2011. View Article : Google Scholar : PubMed/NCBI

53 

El-Metnawy WH, Mattar MM, El-Nahass YH, Samra MA, Abdelhamid HM, Abdlfattah RM and Hamed AR: Predictive value of pretreatment BCR-ABL (IS) transcript level on response to imatinib therapy in Egyptian patients with chronic phase chronic myeloid leukemia (CPCML). Int J Biomed Sci. 9:48–53. 2013.PubMed/NCBI

54 

Kang Y, Hodges A, Ong E, Roberts W, Piermarocchi C and Paternostro G: Identification of drug combinations containing imatinib for treatment of BCR-ABL+ leukemias. PLoS One. 9:e1022212014. View Article : Google Scholar : PubMed/NCBI

55 

Palamà IE, Coluccia AM and Gigli G: Uptake of imatinib-loaded polyelectrolyte complexes by BCR-ABL+ cells: A long-acting drug-delivery strategy for targeting oncoprotein activity. Nanomedicine. 9:2087–2098. 2014. View Article : Google Scholar

56 

Jamieson CH: Chronic myeloid leukemia stem cells. Hematology Am Soc Hematol Educ Program. 2008:436–442. 2008. View Article : Google Scholar

57 

Vilpo J, Hulkkonen J, Hurme M and Vilpo L: Surface membrane antigen expression changes induced in vitro by exogenous growth factors in chronic lymphocytic leukemia cells. Leukemia. 16:1691–1698. 2002. View Article : Google Scholar : PubMed/NCBI

58 

Mukherjee P, Bhattacharya R, Bone N, Lee YK, Patra CR, Wang S, Lu L, Secreto C, Banerjee PC, Yaszemski MJ, et al: Potential therapeutic application of gold nanoparticles in B-chronic lymphocytic leukemia (BCLL): Enhancing apoptosis. J Nanobiotechnol. 5:42007. View Article : Google Scholar

59 

Lee YK, Bone ND, Strege AK, Shanafelt TD, Jelinek DF and Kay NE: VEGF receptor phosphorylation status and apoptosis is modulated by a green tea component, epigallocatechin-3-gallate (EGCG), in B-cell chronic lymphocytic leukemia. Blood. 104:788–794. 2004. View Article : Google Scholar : PubMed/NCBI

60 

Bogusz J, Majchrzak A, Mędra A, Cebula-Obrzut B, Robak T and Smolewski P: Mechanisms of action of the anti-VEGF monoclonal antibody bevacizumab on chronic lymphocytic leukemia cells. Postepy Hig Med Dosw Online. 67:107–118. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Shanafelt TD, Gunderson H and Call TG: Commentary: Chronic lymphocytic leukemia - the price of progress. Oncologist. 15:601–602. 2010. View Article : Google Scholar

62 

Żołnierczyk JD, Borowiak A, Błoński JZ, Cebula-Obrzut B, Rogalińska M, Kotkowska A, Wawrzyniak E, Smolewski P, Robak T and Kiliańska ZM: In vivo and ex vivo responses of CLL cells to purine analogs combined with alkylating agent. Pharmacol Rep. 65:460–475. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Yu B, Mao Y, Yuan Y, Yue C, Wang X, Mo X, Jarjoura D, Paulaitis ME, Lee RJ and Byrd JC: Targeted drug delivery and cross-linking induced apoptosis with anti-CD37 based dual-ligand immunoliposomes in B chronic lymphocytic leukemia cells. Biomaterials. 34:6185–6193. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Siddiqi T, Thomas SH and Chen R: Role of brentuximab vedotin in the treatment of relapsed or refractory Hodgkin lymphoma. Pharmgenomics Pers Med. 7:79–85. 2014.PubMed/NCBI

65 

Zharov VP, Mercer KE, Galitovskaya EN and Smeltzer MS: Photothermal nanotherapeutics and nanodiagnostics for selective killing of bacteria targeted with gold nanoparticles. Biophys J. 90:619–627. 2006. View Article : Google Scholar

66 

Kutok JL and Aster JC: Molecular biology of anaplastic lymphoma kinase-positive anaplastic large-cell lymphoma. J Clin Oncol. 20:3691–3702. 2002. View Article : Google Scholar : PubMed/NCBI

67 

Heidegger S, Beer AJ, Geissinger E, Rosenwald A, Peschel C, Ringshausen I and Keller U: Combination therapy with brentuximab vedotin and cisplatin/cytarabine in a patient with primarily refractory anaplastic lymphoma kinase positive anaplastic large cell lymphoma. Onco Targets Ther. 7:1123–1127. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Zdzalik D, Dymek B, Grygielewicz P, Gunerka P, Bujak A, Lamparska-Przybysz M, Wieczorek M and Dzwonek K: Activating mutations in ALK kinase domain confer resistance to structurally unrelated ALK inhibitors in NPM-ALK-positive anaplastic large-cell lymphoma. J Cancer Res Clin Oncol. 140:589–598. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Zhao N, Bagaria HG, Wong MS and Zu Y: A nanocomplex that is both tumor cell-selective and cancer gene-specific for anaplastic large cell lymphoma. J Nanobiotechnol. 9:22011. View Article : Google Scholar

70 

Zinzani PL, Vose JM, Czuczman MS, Reeder CB, Haioun C, Polikoff J, Tilly H, Zhang L, Prandi K, Li J, et al: Long-term follow-up of lenalidomide in relapsed/refractory mantle cell lymphoma: Subset analysis of the NHL-003 study. Ann Oncol. 24:2892–2897. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Desai M, Newberry K, Ou Z, Wang M and Zhang L: Lenalidomide in relapsed or refractory mantle cell lymphoma: Overview and perspective. Ther Adv Hematol. 5:91–101. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Pighi C, Gu TL, Dalai I, Barbi S, Parolini C, Bertolaso A, Pedron S, Parisi A, Ren J, Cecconi D, et al: Phospho-proteomic analysis of mantle cell lymphoma cells suggests a pro-survival role of B-cell receptor signaling. Cell Oncol. 34:141–153. 2011. View Article : Google Scholar

73 

Cely I, Yiv S, Yin Q, Shahidzadeh A, Tang L, Cheng J and Uckun FM: Targeting mantle cell lymphoma with anti-SYK nanoparticles. J Anal Oncol. 1:1–9. 2012.

Related Articles

Journal Cover

September-2015
Volume 34 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Samir A, Elgamal BM, Gabr H and Sabaawy HE: Nanotechnology applications in hematological malignancies (Review). Oncol Rep 34: 1097-1105, 2015
APA
Samir, A., Elgamal, B.M., Gabr, H., & Sabaawy, H.E. (2015). Nanotechnology applications in hematological malignancies (Review). Oncology Reports, 34, 1097-1105. https://doi.org/10.3892/or.2015.4100
MLA
Samir, A., Elgamal, B. M., Gabr, H., Sabaawy, H. E."Nanotechnology applications in hematological malignancies (Review)". Oncology Reports 34.3 (2015): 1097-1105.
Chicago
Samir, A., Elgamal, B. M., Gabr, H., Sabaawy, H. E."Nanotechnology applications in hematological malignancies (Review)". Oncology Reports 34, no. 3 (2015): 1097-1105. https://doi.org/10.3892/or.2015.4100