Lactoferrin may inhibit the development of cancer via its immunostimulatory and immunomodulatory activities (Review)

  • Authors:
    • Sian Pan
    • Huiting Weng
    • Guohong Hu
    • Shiwen Wang
    • Tian Zhao
    • Xueping Yao
    • Libin Liao
    • Xiaopeng Zhu
    • Yanshan Ge
  • View Affiliations

  • Published online on: September 17, 2021     https://doi.org/10.3892/ijo.2021.5265
  • Article Number: 85
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lactoferrin (Lf) is secreted by ectodermal tissue and has a structure similar to that of transferrin. Although Lf seems to be multifunctional, its main function is related to the natural defense system of mammals. The present review aims to highlight the major actions of Lf, including the regulation of cell growth, the inhibition of toxic compound formation, the removal of harmful free radicals and its important role in immune response regulation. Moreover, Lf has antibacterial, antiviral, antioxidant, anticancer and anti‑inflammatory activities. In addition, the use of Lf for functionalization of drug nanocarriers, with emphasis on tumor‑targeted drug delivery, is illustrated. Such effects serve as an important theoretical basis for its future development and application. In neurodegenerative diseases and the brains of elderly people, Lf expression is markedly upregulated. Lf may exert an anti‑inflammatory effect by inhibiting the formation of hydroxyl free radicals. Through its antioxidant properties, Lf can prevent DNA damage, thereby preventing tumor formation in the central nervous system. In addition, Lf specifically activates the p53 tumor suppressor gene.

1. Introduction

Lactoferrin (Lf), which has a molecular weight of 77-80 kDa, is an iron-binding glycoprotein that has multiple functions in the body; it is involved in the apoptosis of cancer cells and can regulate various immune responses (1). Lf was first discovered in 1939 and is a 'red protein' found in milk; it can be separated and purified from human milk and cow's milk. The isolated protein structure of Lf is similar to that of serum transferrin, with a 60% sequence homology, and it reversibly binds to iron (Fe3+) ions (2). Therefore, Lf is classified as a member of the transferrin family, together with serum transferrin, melanotransferrin and ovotransferrin (3). This multifunctional protein is present in mucosal secretions, including tears, saliva, vaginal secretions, semen, nasal secretions, bronchial secretions, bile, gastrointestinal secretions, urine, cow's milk and human milk (4). Lf is also present on the mucosal surface and granules of white blood cells. Human milk and cow's milk are the most abundant sources of Lf (5). Lf is very similar between different species. In fact, the homology between the Lf of humans and cattle is 77% (6).

Lf promotes the absorption of iron by the body; it regulates cell growth, removes harmful free radicals and inhibits the formation of toxic compounds. In the regulation of immune responses, Lf exerts antibacterial, antiviral, anti-oxidation, anticancer and anti-inflammatory activities (5). Accordingly, Lf is added to a number of commercial products, including infant formula, fermented milk, cosmetics, therapeutic drinks, toothpaste and other products used in daily life (7).

In human endometrial stromal cells (8) and human embryonic kidney cells, Lf can enhance DNA synthesis in normal cells in a dose-dependent manner (9). In addition, another form of Lf, ∆Lf, can be expressed in glandular epithelium cells such as those of the prostate and salivary glands. ∆Lf is a protein subtype of Lf that lacks the leader sequence and the first 25 residues of the original protein (10). The truncated protein mRNA is detected in all normal tissues, but not in some tumor-derived cell lines. Several studies have shown that the chromosomal region encoding Lf is deleted in various tumors, which is a spontaneous process that may occur during cancer (11). ∆Lf plays a role as a transcription factor in cells, participates in the regulation of specific gene expression and plays a role in cancer (12). A number of in vivo studies have indicated the potential antitumor effect of Lf, suggesting that the oral bovine Lf (bLf) administration could decrease chemically induced carcinogenesis in rodents, along with marked cytotoxic and anti-metastatic activity against numerous cancer cell lines (13,14). The antitumor effect of Lf acts via a number of different mechanisms, including the induction of apoptosis in tumor tissues (7).

Owing to the overexpression of a number of cell surface receptors, Lf has a positive targeting effect; therefore, it is considered to be an ideal nanocarrier for certain hydrophobic therapeutic agents. In addition, as Lf can cross the blood-brain barrier (BBB), it has proven to be a good candidate for manufacturing nanocarriers to specifically deliver drugs for brain tumors. Therefore, Lf appears as a promising molecule with multiple applications in the fields of cancer treatment and nanomedicine. Lf has numerous advantages in terms of its ability to actively participate in the manufacture of nanocarriers. Furthermore, it is one of the few proteins that have a net positive charge under physiological conditions [isoelectric point (pI) 8.0-8.5]. Owing to its high pI value, Lf is positively charged over a wide range of pH values (15), is fairly stable in the gastrointestinal tract and possesses a number of intestinal receptors that facilitate the oral absorption and bioavailability of Lf-based nanocarriers within the circulation.

2. Lf exerts both immunostimulatory and immunomodu-latory activities

Lf is a natural immune modulator that plays roles in the innate and acquired immune systems, which regulate antibody formation, T- and B-cell maturation, and increase the percentage of natural killer cells in the lymphocyte population (16). The ability of Lf to regulate the activity of the immune response may be due to its ability to bind endotoxins [lipopolysaccharides (LPSs)] (17,18). In a previous study, Lf was found to alleviate the cellular inflammation induced by LPS by attenuating the nuclear factor-κB/mitogen-activated protein kinase pathways, mitigating oxidative stress and maintaining cellular barrier integrity. Such a finding implies that Lf plays an important role in immune regulation (19). When gram-negative bacteria try to invade the human host, the bacteria come into contact with various proteins of the innate immune system. Part of the bacterial outer membrane contains LPS. When this 'pathogen-related molecular pattern' is recognized by Toll-like receptor 4, it triggers a number of immune responses in various white blood cells and platelets (20,21). The combination of Lf and endotoxins released by bacteria can decrease the degree of stimulation of the immune system. This process can prevent overstimulation, which sometimes occurs in diseases such as sepsis. The hLf1-11 peptide derived from human lactoferrin (hLf) can inhibit myeloperoxidase, which is a major host defense enzyme found in a variety of white blood cells, which may further decrease the innate immune response (22). Furthermore, hLf has been shown to stimulate the maturation of dendritic cells and recruit various white blood cells (23). Therefore, Lf plays an activating role in innate and adaptive immune responses.

Lactoferrin is an allosteric enhancer of the proteolytic activity of cathepsin G, thereby affecting the function of adaptive immune cells (24). Lf has a positive charge that enables it to bind to the negatively charged surface molecules of various immune system cells, and this connection is believed to trigger signaling pathways that result in cellular responses such as activation, differentiation and proliferation. Lf can be transported to the nucleus in order to bind DNA and activate various signaling pathways (25). Lactoferrin can bind to DNA, and through its highly positively charged N-terminal region, which remains associated with the extruded DNA in the neutrophil extracellular traps, can still contribute to the bacterial killing in this process. As the granules also secrete a variety of proteolytic enzymes, Lf or other polypeptides may also be locally released from intact Lf (26).

As well as the induction of systemic immunity, skin immunity is promoted and allergic reactions are suppressed by Lf (27). The immune system is activated against skin allergens, resulting in the dose-dependent inhibition of Langerhans cell migration and dendritic cell accumulation in lymph nodes. The exposure of leukocyte Lf to cytokines, pro-inflammatory cytokines, TNF-α, IL-6 and IL-1β may be adjusted to increase and decrease. The production of these factors depends on the type of signal that is recognized by the immune system. At the cellular level, Lf can increase the number of CD4+ and CD8+ cells in natural killer (NK) cells and T cells (28), promote the recruitment of leukocytes in the blood, induce phagocytosis and regulate the process of bone marrow formation (29). Lf also increases the expression of hyaluronic acid, which is required for the formation of granulation tissue, upregulates platelet-derived growth factors and promotes the proliferation and migration of keratinocytes; this is a necessary condition for the re-epithelialization of wounds. Lf also protects cells from apoptosis (30).

3. Lf is a natural immune modulator involved in the antitumor response

Lf is considered as a key component of the first line of defense for the human body (15) and has a variety of biological effects, including regulation of the immune response, iron absorption, and anti-inflammatory and antioxidant activities. Lf exerts antitumor effects through a variety of mechanisms. Oral bLf can decrease chemically induced carcinogenesis in rodents and has significant cytotoxicity and anti-metastatic activity in numerous cancer cell lines, such as breast cancer and stomach cancer cell lines (31,32). Table I summarizes some of the anti-carcinogenic mechanisms of Lf.

Table I

Anti-carcinogenic activity of Lf against various tumor types.

Table I

Anti-carcinogenic activity of Lf against various tumor types.

Cancers associated with LfProteinOutcome(Refs.)
Breast cancerhLfArrest cancer cells in the G0/G1 phase, induction of apoptosis, and modulation of Bcl-2 and Bax expression(100)
bLfSuppression of V-H+ ATPase and decrease of the acidity of the tumor microenvironment(101)
Colorectal cancerhLf, bLfIncreased expression of TGF-β1, stimulation of IL-18 secretion in Caco-2 cells(102)
bLfEnhanced infiltration of CD4+ and CD8+ cells, increased production of IL-18(103)
GBMhLfSuppression of the proliferation of NMD and FN primary cells by a decrease in the expression of cyclin D1 and D4(104)
Lung cancerhLfAntiproliferative effects attributed to the elevated levels of hypophosphorylated Rb in H1299 cells(105)
bLfDecreased levels of TNF-α, IL-4, IL-6 and IL-10 cytokines, limiting inflammation and restricting tumor proliferation(106)
NPChLfDownregulation of PDPK1 via the MAPK/c-Jun pathway and suppression of K18-facilitated AKT stimulation(107)
OSCCbLfSelective suppression of growth through mTOR/S6K and JAK/STAT3 signaling pathways and triggering of apoptosis in OSCC(108)
Prostate cancerbLfInhibition of the plasma membrane V-ATPase, suppressing tumor progression and metastasis in PC-3 cells(14)

[i] hLf, human lactoferrin; bLf, bovine lactoferrin; NPC, nasopharyngeal arcinoma; OSCC, oral squamous cell carcinoma; GBM, glioblastoma.

Lf is a survival factor of rheumatoid synovial neutrophils, an iron-binding protein that is released from activated neutrophils in inflammatory sites, and has anti-inflammatory and antibacterial properties (33). Although the isolation of iron by Lf and the direct effect on reactive oxygen intermediates are major factors in decreasing excessive inflammatory response damage by directly controlling the development of higher-order immune functions, Lf can regulate injury and pathology caused by injury. This ultimately leads to a decrease in the pathological damage during inflammation (34). The mechanism of action of Lf involves a component that differentially regulates the cellular immune response in sepsis models in vivo. Apoptotic cells can release Lf and combine with neutrophils to inhibit the chemotaxis of neutrophils, enabling macrophages to swallow apoptotic cells, thereby exerting anti-inflammatory effects (Fig. 1) (35).

Bezault et al (36) confirmed, for the first time, the antitumor activity of Lf in fibrosarcoma and melanoma mouse models. Furthermore, an intraperitoneal injection of hLh could inhibit the growth of solid tumors and lung metastasis, independent of the iron saturation of the protein. Notably, the anticancer ability of LF is related to the presence of NK cells. To further prove the relevance of Lf in anticancer activities, Damiens et al (37) studied the role of Lf in cancer progression under inflammatory conditions. The experimental results showed that Lf regulates NK cell cytotoxicity and the sensitivity of target cells to lysis. Similar results were also obtained in experiments by Shi and Li (38). However, Iyer et al (39) pointed out that the antitumor properties of Lf may partly be due to its iron-binding properties. Free iron may act as a mutagenic promoter via the induction of oxidative damage to the nucleic acid structure (40), thereby decreasing the risk of tumors induced by oxidation (Fig. 2) (41,42).

Numerous studies (43-45) have shown that exogenous treatment with Lf and its derivatives can effectively inhibit tumor growth and decrease tumor susceptibility. Specifically, the downregulation or silencing of Lf and its derivatives can lead to an increased chance of developing a tumor (46). Conversely, the proliferation of cancer cells is prevented after the restoration of the Lf gene (47). However, these studies did not definitively conclude the mechanism underlying the anticancer effect of Lf. This review discusses the potential applications of Lf gene expression in cancer treatment and the association between Lf and cancer. To date, it has been indicated that the cytotoxicity of Lf to several cancers occurs via three methods under different conditions: i) Destruction of cell membranes; ii) induction of cell apoptosis; and iii) cell cycle arrest and cellular immune response.

Destruction of cell membranes

Lf and its derived peptides are easily endocytosed by Jurkat cells. Cutone et al (48) found that in T lymphocytes, Lf enters cells through receptor-mediated endocytosis and is almost completely degraded in lysosomes. The Lf-active peptide, Lf-B, also exhibits conformation-dependent uptake efficiency (49). The increase in membrane permeability may change the barrier function of the membrane and promote cell death. At low concentrations, Lf and its derivative peptides can increase cell lysis, and at high concentrations, Lf can regulate cell lysis, depending on the phenotype of the target cells (50). Most cancer cells contain a large amount of proteoglycans, aminoglycans and sialic acid, which all interact with Lf (51,52). This recognition may be the basis of the specificity and selectivity of Lf anticancer drugs. For example, Riedl et al (50) found that phosphatidylserine, a component of the cytoplasmic membrane mainly found in tumor cells, is a key target for the specific anticancer activity of hLf derivatives. This selective interaction through cell surface receptors is actually a cytotoxic reaction. Particularly at high concentrations, hLf and bLf and the peptides derived from them have been shown to promote cytotoxicity and cell death in prokaryotic and eukaryotic pathogens and cancer cells (53,54). This is mainly related to the cationic charge of Lf. Lf can promote electrostatic interactions with negatively charged cell surface receptors. Cationic peptides derived from LF have a low mass ratio and can enter the cell membrane and destroy its stability, thereby easily inducing cell membrane dissolution (55).

Apoptosis induction

Cell apoptosis induced by Lf has been described as the pivotal pathway whereby peptides exert their cytotoxic effects against various cancer cells. However, the apoptotic pathway that they trigger depends on the cell type (56,57).

In a previous study, stomach cancer SGC-7901 cells were treated with Lf, and phosphorylated Akt and numerous key proteins involved in the Akt signaling pathway were decreased as a result. However, the expression levels of phosphorylated caspase-9 and phosphorylated glycogen synthase kinase-3β were increased, indicating that, in stomach cancer SGC-7901 cells, Lf-induced apoptosis may be regulated via the Akt pathway (32). Lf was also found to induce a stress-related mitogen-activated protein kinase pathway in Jurkat T cells, where c-Jun N-terminal kinase (JNK) associated with Bcl-2 was hypothesized to be the pathway responsible for the apoptosis induced by Lf (58). Lf treatment induced caspase-9 and -3 activation and increased the level of Bcl-2 phosphorylation. Following the abolition of JNK activation, cell death did not occur in Lf-treated Jurkat cells. Additionally, BLf was demonstrated to induce the apoptotic extrinsic pathway by upregulating Fas signaling in the colon mucosa of azoxymethane-treated rats (59).

Cell cycle arrest and cellular immune responses

Mammalian cell cycles are usually strictly controlled by hormones and growth factors, and abnormal regulation may lead to tumors. Cyclins, cyclin-dependent kinases (CdKs) and their antagonists, CdK inhibitors, are key factors that regulate cell cycle progression. In breast cancer MDA-MB-231 cells, hLF was found to inhibit cell growth during the transition phase from G1 to S of the cell cycle. At the molecular level, hLF induces a significant decrease in the protein level and activity of Cdk2 and Cdk4, activates cyclins D and E, and plays a key role in the transition from the G1 to the S phase (60). Similar hLf effects have also been reported in four head and neck cancer cell models, with blockage of the G1 to S phase after hLf treatment (61). Lf was reported to induce cell growth arrest by reducing phospho-Akt resulting in increased expression and activity of p21Cip1 and p27Kip1 (62).

Lf has been confirmed to enhance the adaptive immune response and is an effective anti-inflammatory drug (63). Although its molecular mechanism still needs to be revealed, researchers have found that both hLf and bLf can enter the host cell nucleus and bind to DNA to regulate gene expression, thereby exerting their anti-inflammatory activity (64). hLf has significantly increased NK cell-mediated cytotoxicity in breast cancer and colon cancer cell lines (65).

4. Lf has anticancer effects on tumors of the central nervous system

Tammam et al (66) revealed that the cytotoxicity of Lf to gliomas can be attributed to its cytoplasmic distribution. The nuclear transmission of Lf induces cell proliferation rather than cytotoxicity, suggesting that the mode of action of Lf in glioma is related to cell location.

It is well know that tumor cells overexpress Lf receptors in order to fulfill the increased nutritional demands of these highly proliferative cells (67). Lf is an ideal nanocarrier for certain hydrophobic therapeutics due to its active targeting potential as a result of its receptor being overexpressed on the surface of a number of cells. Moreover, Lf is good potential candidate for fabricating nanocarriers that specifically deliver drugs to brain tumors, as Lf can cross the BBB. Consequently, Lf appears as a promising molecule with multiple applications in the fields of cancer therapy and nanomedicine (68). Song et al (69) demonstrated the potential utility of Lf-conjugated GO@Fe3O4 nanocomposites for therapeutic applications in the treatment of gliomas (69). Lf-conjugated iron oxide nanoparticles can be used as tracers for targeted brain glioma imaging using magnetic particle imaging (70). Lf/phenylboronic acid-functionalized hyaluronic acid (HA) nanogel crosslinked with a disulfide bond crosslinker was generated as a reduction-sensitive dual-targeting glioma therapeutic platform for doxorubicin hydrochloride (DOX) delivery (71). Lf-HA-DOX significantly increased drug delivery to the glioma and may thus serve as a promising anti-glioma therapy (72).

5. Lf affects tumor progression by exerting anti-bacterial and anti-viral activities

Lf has a broad inhibitory effect on anti-bacterial infections

Inflammatory bowel disease is a chronic inflammatory and relapsing condition of the gastrointestinal tract (73). Normally, the gut microbiota is composed of 90% Bacteroidetes and Firmicutes, with rare phyla, such as Proteobacteria and Actinobacteria, as well as fungi, viruses, and protists, composing the remaining 10%. Anti-microbial activity has been described as the first Lf function linked to the ancestral host defense-linked mechanisms to target pathogen infections. This activity, evaluated in several in vitro (74,75) and in vivo (76) models, can be both independent and dependent of the iron-binding ability of Lf. The anticancer activity of Lf via host immunomodulation has been widely reported, particularly in colorectal cancer (43).

Lf is a multifunctional natural defense protein with significant antibacterial activity (77); its function is mainly reflected in the absorption of Fe3+, which limits the use of Fe3+ by bacteria in the infected site, and inhibits the growth and reproduction of these microorganisms and the expression of their virulence factors. The bactericidal effect of Lf is mainly mediated by its interaction with the bacterial surface. In vivo (78) and in vitro (79) studies have shown that Lf prevents certain bacteria from adhering to host cells (15) (Fig. 3).

Lf destroys the outer membrane of gram-negative bacteria by interacting with LPS. The positively charged n-terminus of Lf prevents LPS and bacterial cations (Ca2+ and Mg2+) from interacting, resulting in the release of LPS from the cell wall, increasing membrane permeability and subsequently causing damage to the bacteria. The interaction between Lf and LPS also enhances the effect of natural antibacterial agents, such as lysozyme, which are secreted from the mucosa at high concentrations together with Lf via the BBB (15). Dialysis chamber research indicates that bacterial killing requires direct contact with Lf, and work with purified LPS suggests that this relates to direct LPS-binding by the protein. As Lf and lysozyme are both present in mucosal secretions and neutrophil granules, their interaction may help the host defense (80).

The ability of Helicobacter pylori to use hLf as a source of iron depends on the contact between cells and proteins. As Lf is abundant in gastrectomy specimens of patients with superficial or atrophic gastritis, the uptake of iron by H. pylori through specific hLf receptors may play a major role in the virulence of H. pylori infection (Fig. 4) (81). The combination of bovine Lf and Streptococcus pneumoniae surface protein is poor, and human transferrin does not bind to S. pneumoniae surface protein (82). Breast milk Lf inactivates two putative colonization factors expressed by Haemophilus influenzae. Breast milk Lf may decrease the pathogenic potential of H. influenzae by the selective inactivation of iga1 protease and hap, thereby interfering with the colonization of the bacteria (83).

Lf has a broad inhibitory effect on DNA and RNA viruses

Lf is an iron-binding glycoprotein found in some mucosal secretions and with antiviral activity against DNA and RNA viruses, such as HIV and rotavirus. The antiviral effect of LF occurs in the early stages of infection. Lf prevents the virus from entering the host cell by blocking cell receptors (84). In particular, Lf has antiviral and immune responses, such as demonstrated by Lf against SARS-CoV, which is closely related to SARS-CoV-2 that causes COVID-19 (85).

Lf can prevent the internalization of certain viruses into host cells, such as poliovirus type 1, which causes human polio, herpes simplex virus types I and II, and giant cell viruses. For other viruses, such as hepatitis C virus (HCV) and rotavirus, instead of preventing their entry, Lf inhibits virus replication in the host cell. At present, the specific inhibitory mechanism remains to be studied; however, a widely accepted hypothesis is the binding and blockage of glycosaminoglycan virus receptors, particularly heparan sulfate (HS), by Lf. LF and HS in combination stops the first contact between the host and virus cells, thereby preventing infection. In vitro studies have shown that in human plasma and milk proteins, LF exerts a strong activity on HIV, and this effect is due to the inhibition of virus replication in the host cells (15). The HCV envelope protein binds to Lf. hLf and bLf, a multifunctional immunomodulator, combines two HCV envelope proteins. Based on western blotting with milk separated by sodium lauryl sulfate-polyacrylamide gel electrophoresis or immunopurification, bacteria expressing e1 and e2 can bind to Lf (86-88).

6. Lf has enzyme activity functions

The effect of Lf on gram-positive bacteria is mainly based on the combination of cations on the surface of Lf and anions on the surface of the bacteria, thereby neutralizing the negative charges on the surface of the gram-positive bacteria. For example, lipoteichoic acid decreases the negative charge on the cell wall, thereby facilitating the contact between lysozyme and the peptidoglycan under the cell wall, ultimately exerting an enzymatic effect (15).

Lf functions as an enzyme in certain reactions. Lf is the milk protein with the highest activity of DNase, RNase, ATPase and amylase. However, these activities are not the only enzymatic activities of Lf. bLf binds two HCV envelope proteins (88). Lf has DNA-binding properties (89) and can participate in the transcriptional activation of specific DNA sequences (90) or as a signal transduction mediator (91). Celiac disease has the highest amylase and ATP activities (92). The discovery of the properties of the Lf enzyme helps to clarify a number of its physiological functions.

7. Lf plays an extensive role in nanotechnology

Lf is used as a nanocarrier of DOX, as its receptor is highly expressed on the surface of highly proliferating cells (such as cancer cells). DOX is an effective cytotoxic anticancer drug, but has been reported to exhibit extensive toxicity to the heart and spleen, in addition to its limited oral absorption (93,94). Drug-loaded preparations have shown good physical stability, indicating that damage to the red blood cell membrane is negligible. Drug delivery through nano-formulations not only minimizes the cardiotoxicity of DOX, but also improves the efficacy and bioavailability of the drug in a targeted-specific manner (95).

A study has also shown that Lf nanoparticles are used to encapsulate antiviral drugs. Zidovudine nano-encapsulation into Lf nanoparticles has been achieved through sol-oil chemistry. Zidovudine is an effective antiviral drug with good bioavailability (50-75%); however, it can cause bone marrow suppression, neutropenia and organ toxicity. In the study by Kumar et al (96), the size of the prepared nanoparticles was 50-60 nm, the drug encapsulation efficiency was 67% and good physical stability was observed at room temperature and at 4°C. Furthermore, there was no significant change in particle size or drug content. Oral administration of efavirenz-loaded Lf nanoparticles resulted in anti-HIV-1 effects comparable to those of the free drug. In addition, compared with free efavirenz, drug-loaded nanoparticles showed improved pharmacokinetic characteristics and lower organ toxicity, indicating that this nanoformulation is a safe nanoplatform that can enhance drug delivery (96).

The cationic nature of Lf can be used to form complexes with negatively charged DNA through electrostatic complexation. In this context, in a previous study, plasmid pGFPC1, which encodes the green fluorescent protein, was used as a cargo gene (97). Lf nanoparticles loaded with plasmids were prepared using the sol-oil method. The diameter of the prepared Lf nanoparticles was 60 nm and the PDI was low, indicating the uniformity of the preparation. The prepared Lf nanoparticles also showed enhanced physical stability at a temperature of 4°C for ≤10 weeks without the particle size exhibiting significant changes. Incubation in DMEM containing 10% serum at 37°C for 8 h also did not result in changes to the particle size, which would result in longer plasma levels. This improved stability may be related to the strong electrostatic interactions between the positively charged Lf and negatively charged DNA. According to reports, Lf has a DNA-binding domain, which may help to further promote DNA binding and the formation of a tighter DNA-Lf nanocomplex (98).

The methods used to prepare Lf nanoparticles include nanoparticle albumin-bound (NAB) and thermal denaturation methods. The NAB technology is primarily dependent on the presence of an oily phase, which is slowly added to the aqueous phase containing Lf. As a nanoparticle, Lf forms a gel when exposed to heat treatment, ionic strength or changes in pH. Generally, the thermal gelation of a protein starts with a heating step to denature the protein, followed by the addition of salt to induce protein aggregation (87).

Lf can participate in the production of polyelectrolyte complex nanocarriers. The synthesis of such nanocarriers is based on the use of two oppositely charged molecules, such as a positively charged protein and a negatively charged natural polysaccharide. Nanocarriers based on polyelectrolyte complexes are likely stabilized by strong electrostatic interactions between cationic proteins and anionic polysaccharides. This stabilization can also enhance the stability of the encapsulated active ingredients (99). Table II summarizes representative examples of Lf-based nanocarriers for drug delivery applications.

Table II

Lf-based nanocarriers for drug delivery applications.

Table II

Lf-based nanocarriers for drug delivery applications.

DrugIndicationOutcome(Refs.)
Rapamycin and wogonin, or dasatinib and Fe3O4 NPsBreast cancerEnhanced synergistic cytotoxicity and suppression of MCF-7 cells and EAT tumor growth(109,110)
DOX and Ellagic acidNSCLCHigher cytotoxic effect and uptake into A549 cancer cells triggered by Tf and CD44 receptors(111)
TMZGliomaSignificant decrease of tumor volume and improved median survival time(112)
DOXHCCMinimized cardiotoxicity of DOX and enhanced its efficacy and bioavailability(95)
5-FUMelanomaHigher intracellular uptake and 2.7-fold improved cytotoxicity against B16F10 melanoma cells(113)
Shikonin JQ1Colorectal cancerNPs elicited immunogenic cell death and repolarized TAMs to M1 macrophages(114)
EFVHIVTwo-fold increased anti-HIV-1 activity in comparison with free drug(115)
Gambogic acidHCCEnhanced oral bioavailability and anticancer effect of gambogic acid, thus decreasing its toxicity(116)
DOXProstate cancerOral Fe-bLf-DOX inhibited tumor growth, prolonged survival and decreased DOX toxicity(117)

[i] DOX, doxorubicin; 5-FU, 5-fluorouracil; EFV, efavirenz; TMZ, temozolomide; EAT, Ehrlich ascites tumor; NP, nanoparticle; HIV-1, human immunodeficiency virus; Fe-bLf-DOX, Dox conjugated to iron-saturated bovine Lf.

8. Conclusion and perspectives

This review provides an in-depth summary of the biological characteristics of Lf, including the structure of Lf biomolecules, its binding affinity to iron, and the interaction between Lf and the host. Lf plays an important role in the regulation of the immune response, and has immune stimulation, immune activation, anti-inflammatory activity, antibacterial, anti-viral effects and, in particular, anticancer activity. Based on the results of several studies, it is known that Lf-based nanocarriers can be easily prepared by simple methods and have excellent active targeting potential for tumor tissues, particularly brain tumors. The manufacture of Lf-based nanocarriers can broadly enhance the therapeutic potential of encapsulating active molecules. The present review discussed the latest preparation methods for Lf-based nanocarriers prepared by the sol-oil method, NAB technology and thermal denaturation method.

Availability of data and materials

Not applicable.

Authors' contributions

HW, YG, XZ, GH, LL and SP collected the related papers and completed the manuscript and figures. YG and XZ provided constructive guidance and performed critical revisions. XY, SW, TZ, HW and SP participated in the design of this review. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Abbreviations:

Lf

lactoferrin

LPS

lipopolysaccharide

HCV

hepatitis C virus

HS

heparan sulfate

CdKs

cyclin-dependent kinases

NK

natural killer

References

1 

Rascon-Cruz Q, Espinoza-Sanchez EA, Siqueiros-Cendon TS, Nakamura-Bencomo SI, Arévalo-Gallegos S and Iglesias-Figueroa BF: Lactoferrin: A glycoprotein involved in immunomodulation, anticancer, and antimicrobial processes. Molecules. 26:2052021. View Article : Google Scholar

2 

Baker EN, Baker HM and Kidd RD: Lactoferrin and transferrin: Functional variations on a common structural framework. Biochem Cell Biol. 80:27–34. 2002. View Article : Google Scholar

3 

Lambert LA, Perri H and Meehan TJ: Evolution of duplications in the transferrin family of proteins. Comp Biochem Physiol B Biochem Mol Biol. 140:11–25. 2005. View Article : Google Scholar

4 

Pierce A, Legrand D and Mazurier J: Lactoferrin: A multifunctional protein. Med Sci (Paris). 25:361–369. 2009.In French. View Article : Google Scholar

5 

Hao L, Shan Q, Wei J, Ma F and Sun P: Lactoferrin: Major physiological functions and applications. Curr Protein Pept Sci. 20:139–144. 2019. View Article : Google Scholar

6 

Garcia-Montoya IA, Cendon TS, Arevalo-Gallegos S and Rascon-Cruz Q: Lactoferrin a multiple bioactive protein: An overview. Biochim Biophys Acta. 1820:226–236. 2012. View Article : Google Scholar

7 

Legrand D, Pierce A, Elass E, Carpentier M, Mariller C and Mazurier J: Lactoferrin structure and functions. Adv Exp Med Biol. 606:163–194. 2008. View Article : Google Scholar

8 

Yanaihara A, Toma Y, Saito H and Yanaihara T: Cell proliferation effect of lactoferrin in human endometrial stroma cells. Mol Hum Reprod. 6:469–473. 2000. View Article : Google Scholar

9 

Huang N, Bethell D, Card C, Cornish J, Marchbank T, Wyatt D, Mabery K and Playford R: Bioactive recombinant human lactoferrin, derived from rice, stimulates mammalian cell growth. In Vitro Cell Dev Biol Anim. 44:464–471. 2008. View Article : Google Scholar

10 

Siebert PD and Huang BC: Identification of an alternative form of human lactoferrin mRNA that is expressed differentially in normal tissues and tumor-derived cell lines. Proc Natl Acad Sci USA. 94:2198–2203. 1997. View Article : Google Scholar

11 

Klein G, Imreh S and Zabarovsky ER: Why do we not all die of cancer at an early age? Adv Cancer Res. 98:1–16. 2007. View Article : Google Scholar

12 

Mariller C, Hardiville S, Hoedt E, Huvent I, Pina-Canseco S and Pierce A: Delta-lactoferrin, an intracellular lactoferrin isoform that acts as a transcription factor. Biochem Cell Biol. 90:307–319. 2012. View Article : Google Scholar

13 

Tsuda H, Ohshima Y, Nomoto H, Fujita K, Matsuda E, Iigo M, Takasuka N and Moore MA: Cancer prevention by natural compounds. Drug Metab Pharmacokinet. 19:245–263. 2004. View Article : Google Scholar

14 

Guedes JP, Pereira CS, Rodrigues LR and Corte-Real M: Bovine milk lactoferrin selectively kills highly metastatic prostate cancer PC-3 and osteosarcoma MG-63 cells in vitro. Front Oncol. 8:2002018. View Article : Google Scholar

15 

Gonzalez-Chavez SA, Arevalo-Gallegos S and Rascon-Cruz Q: Lactoferrin: Structure, function and applications. Int J Antimicrob Agents. 33:301.e1–e8. 2009. View Article : Google Scholar

16 

Actor JK, Hwang SA and Kruzel ML: Lactoferrin as a natural immune modulator. Curr Pharm Des. 15:1956–1973. 2009. View Article : Google Scholar

17 

Appelmelk BJ, An YQ, Geerts M, Thijs BG, de Boer HA, MacLaren DM, de Graaff J and Nuijens JH: Lactoferrin is a lipid A-binding protein. Infect Immun. 62:2628–2632. 1994. View Article : Google Scholar

18 

Elass-Rochard E, Roseanu A, Legrand D, Trif M, Salmon V, Motas C, Montreuil J and Spik G: Lactoferrin-lipopolysaccharide interaction: Involvement of the 28-34 loop region of human lactoferrin in the high-affinity binding to Escherichia coli 055B5 lipopolysaccharide. Biochem J. 312:839–845. 1995. View Article : Google Scholar

19 

Hu P, Zhao F, Wang J and Zhu W: Lactoferrin attenuates lipopolysaccharide-stimulated inflammatory responses and barrier impairment through the modulation of NF-kappaB/MAPK/Nrf2 pathways in IPEC-J2 cells. Food Funct. 11:8516–8526. 2020. View Article : Google Scholar

20 

Baker HM and Baker EN: A structural perspective on lactoferrin function. Biochem Cell Biol. 90:320–328. 2012. View Article : Google Scholar

21 

Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S, Giannias B, Bourdeau F, Kubes P and Ferri L: Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 123:3446–3458. 2013. View Article : Google Scholar

22 

van der Does AM, Hensbergen PJ, Bogaards SJ, Cansoy M, Deelder AM, van Leeuwen HC, Drijfhout JW, van Dissel JT and Nibbering PH: The human lactoferrin-derived peptide hLF1-11 exerts immunomodulatory effects by specific inhibition of myeloperoxidase activity. J Immunol. 188:5012–5019. 2012. View Article : Google Scholar

23 

Park HW, Park SH, Jo HJ, Kim TG, Lee JH, Kang SG, Jang YS and Kim PH: Lactoferrin induces tolerogenic bone marrow-derived dendritic cells. Immune Netw. 20:e382020. View Article : Google Scholar

24 

Eipper S, Steiner R, Lesner A, Sienczyk M, Palesch D, Halatsch ME, Zaczynska E, Heim C, Hartmann MD, Zimecki M, et al: Lactoferrin is an allosteric enhancer of the proteolytic activity of cathepsin G. PLoS One. 11:e1515092016. View Article : Google Scholar

25 

Penco S, Scarfi S, Giovine M, Damonte G, Millo E, Villaggio B, Passalacqua M, Pozzolini M, Garrè C and Benatti U: Identification of an import signal for, and the nuclear localization of, human lactoferrin. Biotechnol Appl Biochem. 34:151–159. 2001. View Article : Google Scholar

26 

Vogel HJ: Lactoferrin, a bird's eye view. Biochem Cell Biol. 90:233–244. 2012. View Article : Google Scholar

27 

Takayama Y and Aoki R: Roles of lactoferrin on skin wound healing. Biochem Cell Biol. 90:497–503. 2012. View Article : Google Scholar

28 

Kabanov AV and Batrakova EV: New technologies for drug delivery across the blood brain barrier. Curr Pharm Des. 10:1355–1363. 2004. View Article : Google Scholar

29 

Elzoghby AO, Abdelmoneem MA, Hassanin IA, Abd Elwakil MM, Elnaggar MA, Mokhtar S, Fang JY and Elkhodairy KA: Lactoferrin, a multi-functional glycoprotein: Active therapeutic, drug nanocarrier & targeting ligand. Biomaterials. 263:1203552020. View Article : Google Scholar

30 

Thangavel P, Ramachandran B, Chakraborty S, Kannan R, Lonchin S and Muthuvijayan V: Accelerated healing of diabetic wounds treated with L-Glutamic acid loaded hydrogels through enhanced collagen deposition and angiogenesis: An in vivo study. Sci Rep. 7:107012017. View Article : Google Scholar

31 

Iigo M, Alexander DB, Long N, Xu J, Fukamachi K, Futakuchi M, Takase M and Tsuda H: Anticarcinogenesis pathways activated by bovine lactoferrin in the murine small intestine. Biochimie. 91:86–101. 2009. View Article : Google Scholar

32 

Xu XX, Jiang HR, Li HB, Zhang TN, Zhou Q and Liu N: Apoptosis of stomach cancer cell SGC-7901 and regulation of Akt signaling way induced by bovine lactoferrin. J Dairy Sci. 93:2344–2350. 2010. View Article : Google Scholar

33 

Wong SH, Francis N, Chahal H, Raza K, Salmon M, Scheel-Toellner D and Lord JM: Lactoferrin is a survival factor for neutrophils in rheumatoid synovial fluid. Rheumatology (Oxford). 48:39–44. 2009. View Article : Google Scholar

34 

Kruzel ML, Zimecki M and Actor JK: Lactoferrin in a context of inflammation-induced pathology. Front Immunol. 8:14382017. View Article : Google Scholar

35 

Bournazou I, Pound JD, Duffin R, Bournazos S, Melville LA, Brown SB, Rossi AG and Gregory CD: Apoptotic human cells inhibit migration of granulocytes via release of lactoferrin. J Clin Invest. 119:20–32. 2009.

36 

Bezault J, Bhimani R, Wiprovnick J and Furmanski P: Human lactoferrin inhibits growth of solid tumors and development of experimental metastases in mice. Cancer Res. 54:2310–2312. 1994.PubMed/NCBI

37 

Damiens E, Mazurier J, el Yazidi I, Masson M, Duthille I, Spik G and Boilly-Marer Y: Effects of human lactoferrin on NK cell cytotoxicity against haematopoietic and epithelial tumour cells. Biochim Biophys Acta. 1402:277–287. 1998. View Article : Google Scholar

38 

Shi H and Li W: Inhibitory effects of human lactoferrin on U14 cervical carcinoma through upregulation of the immune response. Oncol Lett. 7:820–826. 2014. View Article : Google Scholar

39 

Iyer S, Yip TT, Hutchens TW and Lonnerdal B: Lactoferrin-receptor interaction. Effect of surface exposed histidine residues. Adv Exp Med Biol. 357:245–252. 1994. View Article : Google Scholar

40 

Rastogi N, Singh A, Pandey SN, Sinha M, Bhushan A, Kaur P, Sharma S and Singh TP: Structure of the iron-free true C-terminal half of bovine lactoferrin produced by tryptic digestion and its functional significance in the gut. FEBS J. 281:2871–2882. 2014. View Article : Google Scholar

41 

Teng CT: Factors regulating lactoferrin gene expression. Biochem Cell Biol. 84:263–267. 2006. View Article : Google Scholar

42 

Ieni A, Barresi V, Licata L, Cardia R, Fazzari C, Nuciforo G, Caruso F, Caruso M, Adamo V and Tuccari G: Immunoexpression of lactoferrin in triple-negative breast cancer patients: A proposal to select a less aggressive subgroup. Oncol Lett. 13:3205–3209. 2017. View Article : Google Scholar

43 

Tsuda H, Kozu T, Iinuma G, Ohashi Y, Saito Y, Saito D, Akasu T, Alexander DB, Futakuchi M, Fukamachi K, et al: Cancer prevention by bovine lactoferrin: From animal studies to human trial. Biometals. 23:399–409. 2010. View Article : Google Scholar

44 

Gibbons JA, Kanwar RK and Kanwar JR: Lactoferrin and cancer in different cancer models. Front Biosci (Schol Ed). 3:1080–1088. 2011. View Article : Google Scholar

45 

Digumarti R, Wang Y, Raman G, Doval DC, Advani SH, Julka PK, Parikh PM, Patil S, Nag S, Madhavan J, et al: A randomized, double-blind, placebo-controlled, phase II study of oral talactoferrin in combination with carboplatin and paclitaxel in previously untreated locally advanced or metastatic non-small cell lung cancer. J Thorac Oncol. 6:1098–1103. 2011. View Article : Google Scholar

46 

Hoedt E, Hardiville S, Mariller C, Elass E, Perraudin JP and Pierce A: Discrimination and evaluation of lactoferrin and delta-lactoferrin gene expression levels in cancer cells and under inflammatory stimuli using TaqMan real-time PCR. Biometals. 23:441–452. 2010. View Article : Google Scholar

47 

Zadvornyi TV, Lukianova NY, Borikun TV and Chekhun VF: Effects of exogenous lactoferrin on phenotypic profile and invasiveness of human prostate cancer cells (DU145 and LNCaP) in vitro. Exp Oncol. 40:184–189. 2018. View Article : Google Scholar

48 

Cutone A, Ianiro G, Lepanto MS, Rosa L, Valenti P, Bonaccorsi di Patti MC and Musci G: Lactoferrin in the prevention and treatment of intestinal inflammatory pathologies associated with colorectal cancer development. Cancers (Basel). 12:38062020. View Article : Google Scholar

49 

Duchardt F, Ruttekolk IR, Verdurmen WP, Lortat-Jacob H, Bürck J, Hufnagel H, Fischer R, van den Heuvel M, Löwik DW, Vuister GW, et al: A cell-penetrating peptide derived from human lactoferrin with conformation-dependent uptake efficiency. J biol Chem. 284:36099–36108. 2009. View Article : Google Scholar

50 

Riedl S, Leber R, Rinner B, Schaider H, Lohner K and Zweytick D: Human lactoferricin derived di-peptides deploying loop structures induce apoptosis specifically in cancer cells through targeting membranous phosphatidylserine. Biochim Biophys Acta. 1848:2918–2931. 2015. View Article : Google Scholar

51 

Kühnle A, Veelken R, Galuska CE, Saftenberger M, Verleih M, Schuppe HC, Rudloff S, Kunz C and Galuska SP: Polysialic acid interacts with lactoferrin and supports its activity to inhibit the release of neutrophil extracellular traps. Carbohydr Polym. 208:32–41. 2019. View Article : Google Scholar

52 

El YI, Legrand D, Nuijens J, Slomianny MC, van Berkel P and Spik G: The binding of lactoferrin to glycosaminoglycans on enterocyte-like HT29-18-C1 cells is mediated through basic residues located in the N-terminus. Biochim Biophys Acta. 1568:197–204. 2001. View Article : Google Scholar

53 

Jiang R and Lönnerdal B: Bovine lactoferrin and lactoferricin exert antitumor activities on human colorectal cancer cells (HT-29) by activating various signaling pathways. Biochem Cell Biol. 95:99–109. 2017. View Article : Google Scholar

54 

Sharma A, Shandilya UK, Sodhi M, Mohanty AK, Jain P and Mukesh M: Evaluation of milk colostrum derived lactoferrin of sahiwal (Bos indicus) and karan fries (Cross-Bred) cows for its anti-cancerous potential. Int J Mol Sci. 20:63182019. View Article : Google Scholar

55 

Arias M, Hilchie AL, Haney EF, Bolscher JG, Hyndman ME, Hancock RE and Vogel HJ: Anticancer activities of bovine and human lactoferricin-derived peptides. Biochem Cell Biol. 95:91–98. 2017. View Article : Google Scholar

56 

Zhou Y, Zeng Z, Zhang W, Xiong W, Wu M, Tan Y, Yi W, Xiao L, Li X, Huang C, et al: Lactotransferrin: A candidate tumor suppressor-Deficient expression in human nasopharyngeal carcinoma and inhibition of NPC cell proliferation by modulating the mitogen-activated protein kinase pathway. INT J Cancer. 123:2065–2072. 2008. View Article : Google Scholar

57 

Onishi J, Roy MK, Juneja LR, Watanabe Y and Tamai Y: A lactoferrin-derived peptide with cationic residues concentrated in a region of its helical structure induces necrotic cell death in a leukemic cell line (HL-60). J Pept Sci. 14:1032–1038. 2008. View Article : Google Scholar

58 

Lee SH, Park SW, Pyo CW, Yoo NK, Kim J and Choi SY: Requirement of the JNK-associated Bcl-2 pathway for human lactoferrin-induced apoptosis in the Jurkat leukemia T cell line. Biochimie. 91:102–108. 2009. View Article : Google Scholar

59 

Fujita K, Matsuda E, Sekine K, Iigo M and Tsuda H: Lactoferrin enhances Fas expression and apoptosis in the colon mucosa of Azoxymethane-treated rats. Carcinogenesis. 25:1961–1966. 2004. View Article : Google Scholar

60 

Gopal SH and Das SK: Role of lactoferrin in the carcinogenesis of triple-negative breast cancer. J Cancer Clin Trials. 1:e1052016.

61 

Velliyagounder K, Bahdila D, Pawar S and Fine DH: Role of lactoferrin and lactoferrin-derived peptides in oral and maxillofacial diseases. Oral Dis. 25:652–669. 2019. View Article : Google Scholar

62 

Damiens E, El YI, Mazurier J, Duthille I, Spik G and Boilly-Marer Y: Lactoferrin inhibits G1 cyclin-dependent kinases during growth arrest of human breast carcinoma cells. J Cell Biochem. 74:486–498. 1999. View Article : Google Scholar

63 

Ibrahim HM, Mohamed AH, Salem ML, Osman GY and Morsi DS: Anti-neoplastic and immunomodulatory potency of co-treatment based on bovine lactoferrin and/or muramyl dipeptide in tumor-bearing mice. Toxicol Res (Camb). 9:137–147. 2020. View Article : Google Scholar

64 

Sangermano R, Pernarella S, Straker M, Lepanto MS, Rosa L, Cutone A, Valenti P and Ottolenghi L: The treatment of black stain associated with of iron metabolism disorders with lactoferrin: A litterature search and two case studies. Clin Ter. 170:e373–e381. 2019.PubMed/NCBI

65 

Li HY, Li M, Luo CC, Wang JQ and Zheng N: Lactoferrin exerts antitumor effects by inhibiting angiogenesis in a HT29 human colon tumor model. J Agric Food Chem. 65:10464–10472. 2017. View Article : Google Scholar

66 

Tammam SN, Azzazy H and Lamprecht A: Nuclear and cytoplasmic delivery of lactoferrin in glioma using chitosan nanoparticles: Cellular location dependent-action of lactoferrin. Eur J Pharm Biopharm. 129:74–79. 2018. View Article : Google Scholar

67 

Golla K, Bhaskar C, Ahmed F and Kondapi AK: A target-specific oral formulation of Doxorubicin-protein nanoparticles: Efficacy and safety in hepatocellular cancer. J Cancer. 4:644–652. 2013. View Article : Google Scholar

68 

Sabra S and Agwa MM: Lactoferrin, a unique molecule with diverse therapeutical and nanotechnological applications. Int J Biol Macromol. 164:1046–1060. 2020. View Article : Google Scholar

69 

Song MM, Xu HL, Liang JX, Xiang HH, Liu R and Shen YX: Lactoferrin modified graphene oxide iron oxide nanocomposite for glioma-targeted drug delivery. Mater Sci Eng C Mater Biol Appl. 77:904–911. 2017. View Article : Google Scholar

70 

Tomitaka A, Arami H, Gandhi S and Krishnan KM: Lactoferrin conjugated iron oxide nanoparticles for targeting brain glioma cells in magnetic particle imaging. Nanoscale. 7:16890–16898. 2015. View Article : Google Scholar

71 

Zhang M, Asghar S, Tian C, Hu Z, Ping Q, Chen Z, Shao F and Xiao Y: Lactoferrin/phenylboronic acid-functionalized hyaluronic acid nanogels loading doxorubicin hydrochloride for targeting glioma. Carbohydr Polym. 253:1171942021. View Article : Google Scholar

72 

Yin Y, Fu C, Li M, Li X, Wang M, He L, Zhang LM and Peng Y: A pH-sensitive hyaluronic acid prodrug modified with lactoferrin for glioma dual-targeted treatment. Mater Sci Eng C Mater Biol Appl. 67:159–169. 2016. View Article : Google Scholar

73 

Guan Q: A comprehensive review and update on the pathogenesis of inflammatory bowel disease. J Immunol Res. 2019:72472382019. View Article : Google Scholar

74 

Frioni A, Conte MP, Cutone A, Longhi C, Musci G, di Patti MC, Natalizi T, Marazzato M, Lepanto MS, Puddu P, et al: Lactoferrin differently modulates the inflammatory response in epithelial models mimicking human inflammatory and infectious diseases. Biometals. 27:843–856. 2014. View Article : Google Scholar

75 

Sessa R, Di Pietro M, Filardo S, Bressan A, Rosa L, Cutone A, Frioni A, Berlutti F, Paesano R and Valenti P: Effect of bovine lactoferrin on chlamydia trachomatis infection and inflammation. Biochem Cell Biol. 95:34–40. 2017. View Article : Google Scholar

76 

Valenti P, Frioni A, Rossi A, Ranucci S, De Fino I, Cutone A, Rosa L, Bragonzi A and Berlutti F: Aerosolized bovine lactoferrin reduces neutrophils and pro-inflammatory cytokines in mouse models of Pseudomonas aeruginosa lung infections. Biochem Cell Biol. 95:41–47. 2017. View Article : Google Scholar

77 

Svendsen J, Grant TM, Rennison D, Brimble MA and Svenson J: Very short and stable lactoferricin-derived antimicrobial peptides: Design principles and potential uses. Acc Chem Res. 52:749–759. 2019. View Article : Google Scholar

78 

Dial EJ, Romero JJ, Headon DR and Lichtenberger LM: Recombinant human lactoferrin is effective in the treatment of Helicobacter Felis-infected mice. J Pharm Pharmacol. 52:1541–1546. 2000. View Article : Google Scholar

79 

Rodríguez-Franco DA, Vázquez-Moreno L and Ramos-Clamont MG: Antimicrobial mechanisms and potential clinical application of lactoferrin. Rev Latinoam Microbiol. 47:102–111. 2005.In Spanish.

80 

Ellison RR and Giehl TJ: Killing of gram-negative bacteria by lactoferrin and lysozyme. J Clin Invest. 88:1080–1091. 1991. View Article : Google Scholar

81 

Dhaenens L, Szczebara F and Husson MO: Identification, characterization, and immunogenicity of the lactoferrin-binding protein from Helicobacter pylori. Infect Immun. 65:514–518. 1997. View Article : Google Scholar

82 

Kuipers ME, de Vries HG, Eikelboom MC, Meijer DK and Swart PJ: Synergistic fungistatic effects of lactoferrin in combination with antifungal drugs against clinical Candida isolates. Antimicrob Agents Chemother. 43:2635–2641. 1999. View Article : Google Scholar

83 

Hakansson A, Roche H, Mirza S, McDaniel LS, Brooks-Walter A and Briles DE: Characterization of binding of human lactoferrin to pneumococcal surface protein A. Infect Immun. 69:3372–3381. 2001. View Article : Google Scholar

84 

van der Strate BW, Beljaars L, Molema G, Harmsen MC and Meijer DK: Antiviral activities of lactoferrin. Antiviral Res. 52:225–239. 2001. View Article : Google Scholar

85 

Campione E, Cosio T, Rosa L, Lanna C, Di Girolamo S, Gaziano R, Valenti P and Bianchi L: Lactoferrin as protective natural barrier of respiratory and intestinal mucosa against coronavirus infection and inflammation. Int J Mol Sci. 21:49032020. View Article : Google Scholar

86 

Kell DB, Heyden EL and Pretorius E: The biology of lactoferrin, an iron-binding protein that can help defend against viruses and bacteria. Front Immunol. 11:12212020. View Article : Google Scholar

87 

Fu Q, Sun J, Zhang W, Sui X, Yan Z and He Z: Nanoparticle albumin-bound (NAB) technology is a promising method for anti-cancer drug delivery. Recent Pat Anticancer Drug Discov. 4:262–272. 2009. View Article : Google Scholar

88 

Yi M, Kaneko S, Yu DY and Murakami S: Hepatitis C virus envelope proteins bind lactoferrin. J Virol. 71:5997–6002. 1997. View Article : Google Scholar

89 

Bennett RM, Merritt MM and Gabor G: Lactoferrin binds to neutrophilic membrane DNA. Br J Haematol. 63:105–117. 1986. View Article : Google Scholar

90 

He J and Furmanski P: Sequence specificity and transcriptional activation in the binding of lactoferrin to DNA. Nature. 373:721–724. 1995. View Article : Google Scholar

91 

Brandl N, Zemann A, Kaupe I, Marlovits S, Huettinger P, Goldenberg H and Huettinger M: Signal transduction and metabolism in chondrocytes is modulated by lactoferrin. Osteoarthritis Cartilage. 18:117–125. 2010. View Article : Google Scholar

92 

Kanyshkova TG, Babina SE, Semenov DV, Isaeva N, Vlassov AV, Neustroev KN, Kul'minskaya AA, Buneva VN and Nevinsky GA: Multiple enzymic activities of human milk lactoferrin. Eur J Biochem. 270:3353–3361. 2003. View Article : Google Scholar

93 

Fang JH, Lai YH, Chiu TL, Chen YY, Hu SH and Chen SY: Magnetic core-shell nanocapsules with dual-targeting capabilities and co-delivery of multiple drugs to treat brain gliomas. Adv Healthc Mater. 3:1250–1260. 2014. View Article : Google Scholar

94 

Wei M, Guo X, Tu L, Zou Q, Li Q, Tang C, Chen B, Xu Y and Wu C: Lactoferrin-modified PEGylated liposomes loaded with doxorubicin for targeting delivery to hepatocellular carcinoma. Int J Nanomedicine. 10:5123–5137. 2015.PubMed/NCBI

95 

Golla K, Cherukuvada B, Ahmed F and Kondapi AK: Efficacy, safety and anticancer activity of protein nanoparticle-based delivery of doxorubicin through intravenous administration in rats. PLoS One. 7:e519602012. View Article : Google Scholar

96 

Kumar P, Lakshmi YS, C B, Golla K and Kondapi AK: Improved safety, bioavailability and pharmacokinetics of zidovudine through lactoferrin nanoparticles during oral administration in rats. PLoS One. 10:e1403992015. View Article : Google Scholar

97 

Kumari S and Kondapi AK: Receptor-mediated targeted delivery of DNA using Lactoferrin nanoparticles. Int J Biol Macromol. 108:401–407. 2018. View Article : Google Scholar

98 

Elfinger M, Maucksch C and Rudolph C: Characterization of lactoferrin as a targeting ligand for nonviral gene delivery to airway epithelial cells. Biomaterials. 28:3448–3455. 2007. View Article : Google Scholar

99 

Yan JK, Qiu WY, Wang YY and Wu JY: Biocompatible polyelectrolyte complex nanoparticles from lactoferrin and pectin as potential vehicles for antioxidative curcumin. J Agric Food Chem. 65:5720–5730. 2017. View Article : Google Scholar

100 

Wang J, Li Q, Li K, Ou Y, Han Z, Gao D and Li J: Effects of adenovirus vectors mediated human lactoferrin cDNA on mice bearing EMT6 breast carcinoma. Pharmazie. 66:704–709. 2011.

101 

Pereira CS, Guedes JP, Goncalves M, Loureiro L, Castro L, Gerós H, Rodrigues LR and Côrte-Real M: Lactoferrin selectively triggers apoptosis in highly metastatic breast cancer cells through inhibition of plasmalemmal V-H+-ATPase. Oncotarget. 7:62144–62158. 2016. View Article : Google Scholar

102 

Lonnerdal B, Jiang R and Du X: Bovine lactoferrin can be taken up by the human intestinal lactoferrin receptor and exert bioactivities. J Pediatr Gastroenterol Nutr. 53:606–614. 2011. View Article : Google Scholar

103 

Machado S, Alves R, Lima M, Leal I and Massa A: Cutaneous necrobiotic xanthogranuloma (NXG)-successfully treated with low dose chlorambucil. Eur J Dermatol. 11:458–462. 2001.PubMed/NCBI

104 

Arcella A, Oliva MA, Staffieri S, Aalberti S, Grillea G, Madonna M, Bartolo M, Pavone L, Giangaspero F, Cantore G and Frati A: In vitro and in vivo effect of human lactoferrin on glioblastoma growth. J Neurosurg. 123:1026–1035. 2015. View Article : Google Scholar

105 

Son HJ, Lee SH and Choi SY: Human lactoferrin controls the level of retinoblastoma protein and its activity. Biochem Cell Biol. 84:345–350. 2006. View Article : Google Scholar

106 

Tung YT, Chen HL, Yen CC, Lee PY, Tsai HC, Lin MF and Chen CM: Bovine lactoferrin inhibits lung cancer growth through suppression of both inflammation and expression of vascular endothelial growth factor. J Dairy Sci. 96:2095–2106. 2013. View Article : Google Scholar

107 

Deng M, Zhang W, Tang H, Ye Q, Liao Q, Zhou Y, Wu M, Xiong W, Zheng Y, Guo X, et al: Lactotransferrin acts as a tumor suppressor in nasopharyngeal carcinoma by repressing AKT through multiple mechanisms. Oncogene. 32:4273–4283. 2013. View Article : Google Scholar

108 

Chea C, Miyauchi M, Inubushi T, Febriyanti Ayuningtyas N, Subarnbhesaj A, Nguyen PT, Shrestha M, Haing S, Ohta K and Takata T: Molecular mechanism of inhibitory effects of bovine lactoferrin on the growth of oral squamous cell carcinoma. PLoS One. 13:e1916832018. View Article : Google Scholar

109 

Sabra SA, Elzoghby AO, Sheweita SA, Haroun M, Helmy MW, Eldemellawy MA, Xia Y, Goodale D, Allan AL and Rohani S: Self-assembled amphiphilic zein-lactoferrin micelles for tumor targeted co-delivery of rapamycin and wogonin to breast cancer. Eur J Pharm Biopharm. 128:156–169. 2018. View Article : Google Scholar

110 

Sabra SA, Sheweita SA, Haroun M, Ragab D, Eldemellawy MA, Xia Y, Goodale D, Allan AL, Elzoghby AO and Rohani S: Magnetically guided self-assembled protein micelles for enhanced delivery of dasatinib to human triple-negative breast cancer cells. J Pharm Sci. 108:1713–1725. 2019. View Article : Google Scholar

111 

Abd Elwakil MM, Mabrouk MT, Helmy MW, Abdelfattah EA, Khiste SK, Elkhodairy KA and Elzoghby AO: Inhalable lactoferrin-chondroitin nanocomposites for combined delivery of doxorubicin and ellagic acid to lung carcinoma. Nanomedicine (Lond). 13:2015–2035. 2018. View Article : Google Scholar

112 

Kumari S, Ahsan SM, Kumar JM, Kondapi AK and Rao NM: Overcoming blood brain barrier with a dual purpose Temozolomide loaded lactoferrin nanoparticles for combating glioma (SERP-17-12433). Sci Rep. 7:66022017. View Article : Google Scholar

113 

Kumari S and Kondapi AK: Lactoferrin nanoparticle mediated targeted delivery of 5-fluorouracil for enhanced therapeutic efficacy. Int J Biol Macromol. 95:232–237. 2017. View Article : Google Scholar

114 

Wang H, Tang Y, Fang Y, Zhang M, Wang H, He Z, Wang B, Xu Q and Huang Y: Reprogramming Tumor Immune Microenvironment (TIME) and metabolism via biomimetic targeting Codelivery of Shikonin/JQ1. Nano Lett. 19:2935–2944. 2019. View Article : Google Scholar

115 

Kumar P, Lakshmi YS and Kondapi AK: An oral formulation of efavirenz-loaded lactoferrin nanoparticles with improved biodistribution and pharmacokinetic profile. HIV Med. 18:452–462. 2017. View Article : Google Scholar

116 

Zhang ZH, Wang XP, Ayman WY, Munyendo WL, Lv HX and Zhou JP: Studies on lactoferrin nanoparticles of gambogic acid for oral delivery. Drug Deliv. 20:86–93. 2013. View Article : Google Scholar

117 

Shankaranarayanan JS, Kanwar JR, Al-Juhaishi AJ and Kanwar RK: Doxorubicin conjugated to immunomodulatory anticancer lactoferrin displays improved cytotoxicity overcoming prostate cancer chemo resistance and inhibits tumour development in TRAMP mice. Sci Rep. 6:320622016. View Article : Google Scholar

Related Articles

Journal Cover

November-2021
Volume 59 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pan S, Weng H, Hu G, Wang S, Zhao T, Yao X, Liao L, Zhu X and Ge Y: Lactoferrin may inhibit the development of cancer via its immunostimulatory and immunomodulatory activities (Review). Int J Oncol 59: 85, 2021
APA
Pan, S., Weng, H., Hu, G., Wang, S., Zhao, T., Yao, X. ... Ge, Y. (2021). Lactoferrin may inhibit the development of cancer via its immunostimulatory and immunomodulatory activities (Review). International Journal of Oncology, 59, 85. https://doi.org/10.3892/ijo.2021.5265
MLA
Pan, S., Weng, H., Hu, G., Wang, S., Zhao, T., Yao, X., Liao, L., Zhu, X., Ge, Y."Lactoferrin may inhibit the development of cancer via its immunostimulatory and immunomodulatory activities (Review)". International Journal of Oncology 59.5 (2021): 85.
Chicago
Pan, S., Weng, H., Hu, G., Wang, S., Zhao, T., Yao, X., Liao, L., Zhu, X., Ge, Y."Lactoferrin may inhibit the development of cancer via its immunostimulatory and immunomodulatory activities (Review)". International Journal of Oncology 59, no. 5 (2021): 85. https://doi.org/10.3892/ijo.2021.5265