Int J Pharm Pharm Sci, Vol 7, Issue 4, 19-26Review Article


A REVIEW OF POTENTIAL ANTICANCERS FROM ANTIMICROBIAL PEPTIDES

KHAMSAH SURYATI MOHD1*, MOHAMMED AL-KASSIM HASSAN2, WAN-ATIRAH AZEMIN1, SARAVANAN DHARMARAJ2

1Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, Campus Tembila, 22200 Besut, Terengganu, Malaysia, 2Faculty of Medicine and Health Sciences, Universiti Sultan Zainal Abidin, Medical Campus, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia
Email: khamsahsuryati@unisza.edu.my

Received: 10 Nov 2014 Revised and Accepted: 04 Dec 2014


ABSTRACT

Cancer is one of the leading causes of morbidity and mortality globally. The drawbacks of conventional chemotherapy such as resistance, lack of specificity, severe toxicity warrant the need to explore alternative approach for the treatment of cancer. Antimicrobial peptides are part of the innate defense mechanism of all organisms and have been developed as potential alternatives in combatting infectious diseases. In addition, anticancer effects of many peptides have been reported with remarkable prospects in some in vitro studies especially on breast, cervical and lung cancer cell lines, and in vivo murine tumour xenografts. This review summarizes the reports on the activities of some selected anticancer peptides on various cancer cell lines.

Keywords: Antimicrobial peptides, Anticancer/antitumour peptides, Host defense peptides.


INTRODUCTION

Antimicrobial peptides (AMPs) are diverse, ancient and evolutionary conserved components of the innate immune system present in all forms of life [1] and have been isolated from various species (table 1). AMPs are also termed as host defense peptides. They are cationic and amphipathic in nature; this property played a facilitatory role in their binding and insertion into the anionic cell membrane of microorganisms. AMPs possess low propensity for developing resistance, probably due to their distinguished mode of action [2]. Consequently, they show broad spectrum antimicrobial activities against various micro-organisms, including Gram-positive and Gram-negative bacteria, fungi and viruses [3]. Over 2000 AMPs, mostly cationic, have been reported from the antimicrobial peptide database [4] and the number keep increasing with newer research discovery. In addition to their antimicrobial function, AMPs also exert antitumour effect, immunomodulatory effect, and wound-healing effect. They also find applications as drug delivery vector, contraceptive agent, mitogenic agent and signal molecules in signal transduction pathways [5].

Owing to the diversity of functions shown by the antimicrobial peptides, they can also be categorized into antibacterial peptides, antiviral peptides, antifungal peptides, antiparasitic peptides, anti-cancer/antitumour peptides, anti-HIV peptides, anti-protist peptides, insecticidal peptides, spermicidal peptides and AMPs with chemotactic activity [1]. However, one AMP may have or fall into more than one group. Although, AMPs have a certain degree of similarity among themselves regarding the biophysical properties, their sequence is rarely similar among closely related or distinct species [6].

Classification of AMPs

Host defense peptides can majorly be classified based on their secondary structure into four different groups or families which are: i) alpha (α), ii) beta (β), iii) alpha beta (αβ) and non-alpha beta (non-αβ).

The α-family consists of AMPs with helical structure, e. g. magainin-II (fig. 1E). The β-family is composed of AMPs with beta strands; e. g. looped than at in and β-sheet polyphemusin (fig. 1B and 1C, respectively). The αβ-family comprises both α-helical and β-strands in the three-dimensional structure, e. g. human β-defensin-2 and rabbit kidney β-defensin-1 (fig. 1A and 1D, respectively). The non-αβ family also referred to as extended structure contain neither α-helical nor β-strand e. g. indolicidin (Fig.1F). While some peptides belong to one of the four classes, some, however, defy this classification and have a mixed structure [11]. Further to this, AMPs can also be categorized based on their charges, disulfide bridges and amino acid-rich contents as either cationic or non-cationic. Summary of the AMPs based on their structures is shown in table 2.

Fig. 1: Structural classes of antimicrobial peptides. Taken from Jenssen et al. [28]
A = mixed structure of human β-defensin-2 (HBD2), B = looped thanatin, C = β-sheeted polyphemusin, D = rabbit kidney defensin-1, E = α-helical magainin-2, F = extended indolicidin

Mode of action of antimicrobial peptides

Antimicrobial peptides have been termed “natural antibiotics” due to broad spectrum activity against micro-organisms [8]. Cationic peptides interact directly with the negatively charged cellular membrane of bacteria cells resulting in enhanced membrane permeability and ultimately leading to swift cell lysis. [29]. So in effect the peptide can cause membrane disruption or membrane interaction, which may lead to the formation of transient pores and transport of the peptide into the cell thereby ensuring contact with the intracellular targets [30]. The four commonly used models to describe the mechanism of AMP action include: toroidal, carpet-like, barrel-stave and aggregate channel models.

Table 1: Diverse sources of antimicrobial peptides

Source Scientific name Host defense peptides Reference
Human Homo sapien Dermicidin (anionic)LL-37HNP-1 [7][7][7]
Insects Hyalophora cecropia(Silk moth)Apis mellifera(honey bee)Drosophilia melanogaster(fruit fly)Lucilia sericata(green bottle fly) CecropinMelittinDrosomycinLucifensin I [8][9][10][11]
Plants Leonurus cardiac(Motherwort)Viscum album L.Benincasa hispida Lipid transfer proteinViscotoxin A1Hispidalin [12][13][14]
Fishes Oreochromis mossambicus(Freshwater tilapia)Chionodraco hamatus(Marine icefish) Hepcidin TH1-5Chionodracine [15][16]
Amphibians Bufo bufo gargarizans(Asian toad) Buforin-I [17]
Xenopus laevis(S/African clawed frog) Magainin-II [18]
Reptiles Crotalus durissus terrifucus(S/American rattlesnake)Bungarus fasciatus(Branded krait)Emys orbicularis(European pond turtle) CrotamineBF-CATHTBD-1 [19][20][21]
Birds Gallus gallus(chicken)PenguinAnas platyrhynchos(mallard duck) Fowlicidin-1AvBD 103aAvBD2 [22][22][22]
Microbe Rhizopus microsporus(fungus)Pseudoplectania nigrella(fungus)Lactococus lactis(bacterium)Streptococcus pneumoniae(bacterium) Rhimisin-1NZ2114NisinPep27 anal2 [23][24][25][26]

HNP-1 human neutrophil peptide1, AvBD avian β-defensin, BF-CATH B. fungarus cathelicidin, TBD-1 turle β-defensin1, NZ2114 plectasin

Table 2: Antimicrobial peptides based on their structural features

AMP class Structural features Typical examples Structure
Cationic peptides Peptide forming helical structuresOne disulphide bridgeTwo disulphide bridgeThree disulphide bridgeGreater than three disulphide bridgesGlycine-rich peptidesHistidine-rich peptideProline-rich peptideTryptophan –rich peptide CecropinsThanatinTachyplesin IIPanaeidinsDrosomycinPyrrhocoricinDiptericinHistatinIndolicidin α-helixβ-helixβ-helixβ-helixαβ structureN/ARich in Hαβ structureExtended structure
Noncationic peptides Aspartic acid-rich peptideNeuropeptide-derived moleculesOxygen binding proteinsAromatic dipeptides DermicidinSecretolytinLactoferricinN-alanyl-5-s-glutathionyl-3,4-dihydroxylphenylalanine& P-hydroxyl cinnamaldehyde N/Aα-helixβ-turnN/A

Adapted from Pushpanathan et al. [4] N/A = not available

In toroidal model, AMPs align perpendicularly into the bilayer structure with their hydrophobic regions associated with the central part of the lipid bilayer and their hydrophilic regions facing the pore [31]. Whereas, in carpet-like mode (also known as detergent-like test), the peptide micelle touches the membrane first and coats a small area of the membrane. Thereafter, AMP penetrate the lipid layer to allow pore formation occur leaving holes behind [32]. In the barrel-stave model, staves are formed first parallel to the cell membrane. Then barrels are formed and AMPs are inserted perpendicularly to the plane of the membrane bilayer [33]. On the other hand, AMPs glue themselves to the membrane parallel to the surface in an aggregate channel model. Then reorientation of AMPs occur and they insert themselves into the membrane vertically to form sphere-like structures [32]. The schematic diagram of the models is illustrated in fig. 2.

Fig. 2: Illustration of the models of lipid membrane permeabilization by antimicrobial peptides. Adapted from Silva et al. [34]

AMPs as anticancer therapeutics

Cancer is the most malignant disease threatening the health of man. It has been predicted that the number of death from this disease will continue to increase with an estimated 11.5 million deaths by the year 2030 [35]. Conventional chemotherapeutics has many drawbacks such as resistance; lack of selectivity, severe toxicity hence the need to explore alternative approach for cancer remission. AMPs have emerged as potentially better alternative as they have broad anticancer spectrum with low propensity for resistance, selectivity for cancer cells, rapid cell kill, ability to destroy and prevent metastasis of primary tumours and are harmless to vital organs [36]. This review highlights some of the antimicrobial peptides that have been reported to possess anticancer properties.

Anticancer mechanism of AMPs

Three anticancer mechanisms have been proposed for cytotoxic AMPs namely: a) cell membrane lysis; b) inhibition of angiogenesis; and c) activation of extrinsic apoptotic pathways [8].

Cell membrane lysis

This mechanism is similar to the mode of action of antimicrobial peptides that disrupt the bacterial cell wall and the effect may involve selective lysis of the cancer cell membrane. Anticancer peptides (ACPs) membranolytic and selective mode of action on tumor cells can be due to the increased anionicity of the cytoplasmic membrane of these cells [37]. This follows the same pattern as the “carpet” and “barrel-stave” models. According to the carpet mechanism [38], the peptides align themselves parallel to the negatively charged membrane surfaces in a carpet-like fashion until a critical threshold concentration is attained, after which the peptide permeates the membrane. Conversely, for “barrel-stave” model, the lytic cationic amphiphilic peptides self-aggregate via hydrophobic interactions in the membrane and form transmembrane channels or pores [39]. Other membranolytic activities involve the permeation and swelling of mitochondria with release of cytochrome c and apoptosis events [40]. Examples of some oncolytic peptides are listed in table 3.

Inhibition of tumour angiogenesis

Angiogenesis defined as the process of new blood vessel formation is essential during cancer progression [41]. During this process, migration and proliferation and entry into tumour surrounding tissue, of endothelial cells occur forming a capillary network [42]. It plays an essential role in the growth, invasion and metastasis of solid tumours, which could provide necessary nutrients and oxygen and discard metabolic wastes [43]. Present in the tumour cells are angiogenic growth factors that stimulate the process of angiogenesis. These include vascular endothelial growth factor-A(VGEF-A), fibroblast growth factor, epidermal growth factor, platelet-derived growth factor, placental growth factor, angio poietin and other activators [41]. Many peptides could unleash beneficial antiangiogenesis and antitumour effects mainly by interrupting the interactions between growth factors and their receptors [44]. Attempts at controlling tumourigenesis involve targeting the angiogenesis processes [45].

Activation of extrinsic apoptotic pathway

Multicellular organisms have developed apoptosis to control cell metastasis in response to DNA damage, during cell development and cellular stress [46]. In the course of apoptosis, the nucleus and cytoplasm shrink, condense and then fragment, releasing small membrane-bound apoptotic bodies, which are phagocytosed by macrophages or adjacent cells [47]. The extrinsic apoptotic pathway is receptor–mediated and regulated by tumour necrosis factor (TNF) super family namely Fas (CD95) and TNF-related apoptosis-inducing ligand (TRAIL) receptors. Binding of these receptors with their ligands such as Fas ligand (FasL) and TRAIL respectively activate this pathway [48]. Increasing evidence suggest that apoptosis could play a significant part in getting rid of cancer cells without eliciting damage to the healthy normal cells or surrounding tissues [49]. Targeting apoptosis pathways in premalignant and malignant cells will be effective strategies for cancer prevention and treatment [50]. Conventional chemotherapeutic drugs may induce apoptosis by enhancing Fas expression [48]. Peptides that could induce apoptosis in tumor cells are slowly becoming important candidates for the development of new anticancer drugs [44].

The mode of action for anticancer peptides may not be limited to those highlighted above. It is not uncommon, however, to find a peptide with multiple mechanisms in exerting their effect [37].

A12L/A20L & Pep27anal2

Huang et al; [66] demonstrated the role of helicity on the anticancer mechanism of action of cationic helical peptides. The 26-residue amphipathic α-helical peptide A12L/A20L was modified by replacing the original L-amino acid with the D-form on either polar or non-polar face of the helix. This results in improved therapeutic index of the peptide against human cervix adenocarcinoma cells (HeLa) by 9-fold and 22-fold respectively. The anticancer activity of these peptides can also be correlated to their hydrophobicity. When challenged with eight different cell lines including HeLa, human breast adenocarcinoma cell line (MCF7), human lung carcinoma cell line (A549), mouse melanoma cell line (B16), the peptides showed greater anticancer activity against cervical carcinoma cells with IC50 of 2 and 1.2 µmol/l with a necrotic-like membrane disruption mechanism. Thus increasing hydrophobicity can lead to increased anticancer effect [67]. Similar phenomenon stem from introducing tryptophan in the peptide pep27 analogues with increased anticancer activity shown by Pepanal2 against acute myelogenous leukaemia cell line (AML-2), acute promyelocytic leukaemia cell line (HL-60), gastric cancer cell line (SNU-601), Jurkat T cell leukaemia cell line and MCF7 [26].

Cecropin A & B

Cecropin A and B are members of cecropin-family of AMP first derived from the giant silk moth Hyalophora cecropia. These peptides exerted selective inhibitory and antiproliferative efficacy against bladder tumour cell lines namely RT4, 647V, J82, 486P in a dose-dependent fashion. However, both peptides remained benign to fibroblasts of murine and human origin [68]. In a related research, Wu et al; [69] reported that another derivative of cecropin, synthetic peptide CB1a, showed selective anticancer activity against leukemia, stomach carcinoma and lung cancer cells but low toxicity against non-cancer cells. The IC50 of this potential anticancer agent in leukemia and stomach carcinoma were 2-8 folds lower than the parent peptide cecropin. CB1a also displayed high selective toxicity against two lung cancer cells – NCI-H460 and NCI-H520. The peptide had an interaction time with the target cell (IT50) of 7 mins, which suggest that the drug was fast acting. More extensive study was done on this peptide where it, at 50mg/kg, inhibited the growth of lung tumours in an in vivo mouse model xenografted with human tumorigenic NCI-H460 lung cancer cells [64]. Comparatively, the peptide was toxic to cancerous cells and less toxic to normal cells than conventional antitumour drug, docetaxel. The peptide can survive long enough in the bloodstream to exert its effect before protease digests it; this longevity is due to its design of three repeated amphipathic sequence [64].

Table 3: Sequence of some selected oncolytic host defense peptides

Peptide name Sequence Reference
Aurein 1.2 GLFDIIKKIAESF [51]
Hepcidin TH1-5 GIKCRFCCGCCTPGICGVCCRF [52]
Melittin GIGAVLKVLTTGLPALISWIKRKRQQ [53]
Epinecidin-1 GFIFHIIKGLFHAGKMIHGLV [54]
Tachyplesin KWCFRVCYRGICYRRCR [55]
LL-37 LLGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES [43]
Cecropin (CB1a) KWKVFKKIEK-KWKVFKKIEK-AGPKWKVFKKIEK [56]
Lactoferricin FKCRRWQWRMKKLGAPSITCVRRAF [57]
TsAP-1/TsAP-2 FLSLIPSLVGGSISAFK/FLGMIPGLIGGLISAFK [58]
Buforin-II TRSSRAGLQFPVGRVHRLLRK [53]
Magainin-II GIGKFLHSAKKFGKAFVGEIMNS [59]
Hepcidin TH2-3 QSHLSLCRWCCNCCRSNKGC [15]
Gomesin ZCRRLCYKQRCVTYCRGR [60]
Temporin-1CEa FVDLKKIANIINSIFGK [61]
Pardaxin GFFALIPKIISSPLFKTLLSAVGSALSSSGGQE [62]
Piscidin-1 FFHHIFRGIVHVGKTIHRLVTG [63]
HNP-1 ACYCRIPACIAGERRYGTCIYQGRLWAFCC [53]
GW-H1 GYNYAKKLANLAKKFANALW [64]
Pep27anal2 MWKWFHNVLSWWWLLADKRPARDYNRK [26]
Cecropin A KWKLFKKIEKVGQNIRDGIIKAGPAVAVVGQATQIAK [65]
Cecropin B KWKVFKKIEKMGRNIRNGIVKAGPAIAVLGEAKAL [73]

Magainin-II

Magainin-II belongs to the magainins AMP family, and the parent peptide was initially isolated from the skin of African clawed frog, Xenopus laevis [3]. Magainins and its derivatives have been shown to exert anticancer effect on a number of cancer cells. Significant cytotoxicity of magainin-II have been reported against cancer cells including bladder tumours [70], HeLa cells [71], human melanoma [72], lung cancer cells [73]. In vivo, magainins have been shown to improve the recovery of animals with ascites-producing tumours [74]. In addition, nude mice xenografted with melanoma tumour were able to recuperate completely after local treatment with magainin-II. [72]. Selectivity for neoplastic cells over normal cells is one of the main positives of magainin peptides [70], they are also reported to resist degradation by proteases [75].

Melittin

Melittin is a 26 amino acid sequence linear peptide that is the principal component of bee venom. It is a potent activator of phospholipase A2, caspase and matrix metalloproteinase that kill tumour cells [76]. It has been reported as a lytic peptide with broad spectrum of anticancer effects, but its toxicity towards normal cells has limited its potentials as a therapeutic agent [9]. However, this drawback can be subjugated by introducing proper delivery vehicle in the form of melittin nanoparticles that can safely convey the peptide in significant amount intravenously to target and destroy the cells [9]. Other AMP and their analogues derived from bees venom such as halictin HAL-1 (GMWSKILGHLIR), macropin MAC-1 (GFGMALKLLKKVL), and lasioglossins LL-III (VNWKKILGKIIKVVK) have also been reported to be cytotoxic especially to HeLaS3, a clonal derivative of parent HeLa cell line [77].

LL-37/hCAP-18

Human cationic AMP of 18 kDa is the human cathelicidin-derived peptide initially synthesized as a preprotein (hCAP-18) before being subsequently converted into its active form LL-37 of 4.5 kDa [78]. A COOH moiety of hCAP-18 consisting amino acid residue 109-135 (hCAP109-135) and which correspond to LL-376-32 residue induce apoptosis in human oral squamous carcinoma cell line by mitochondrial depolarization without any detectable activation of caspase-3 [79]. This finding support the proposition that the oncolytic activity of LL-37/hCAP-18 is due to LL-37 (17-29), which is a 13-amino acid fragment attached to COOH-terminal region corresponding to amino acid residue 17-29 [80].

Although LL-37 (17–29) is equally cytotoxic to drug-sensitive and drug-resistant variants of the KB human squamous cancer cell line, it also kills untransformed human endothelial cells. Native LL-37 is similarly cytotoxic to human peripheral blood lymphocytes [81]. This toxicity against normal human cells has limited their therapeutic potentials.

Pardaxin

Pardaxin is a 33-amino acid sequence amphipathic polypeptide neurotoxin originally extracted from the marine fish Pardachirus marmoratus. Few studies have reported the anticancer potential of this AMP. Wu et al; [82] reported that pardaxin exhibited antitumour activity against murine fibrosarcoma both in vivo and in vitro. The synthetic peptide, at 25mg/kg, 0.5mg/day inhibit the proliferation of MN-11 cells after 14 d and reduce colony formation in a soft agar assay. By caspase-dependent and reactive oxygen species (ROS)-dependent apoptotic mechanism, the peptide inhibited human fibrosarcoma cells (HT1080) proliferation [83]. Pardaxin induced programmed cell death in HeLa cells but inhibited proliferation of HT1080 cells via a different mechanism [84].

Buforin-IIB & Aurein 1.2

Buforin-IIb is a 21-amino acid peptide derivative of buforin I (a 39-amino acid peptide first isolated from the Asian toad Bufo bufo gargarizan). Both peptides comprise helix-hinged-helix structures derived from histone H2A [85]. Buforin IIb exerts a broad spectrum of anticancer activity; the peptide have been reported to exert cytotoxicity on a remarkably 62 cancer cell lines by explicitly targeting cancer cells through interaction with cell surface gangliosides. It stimulates apoptosis by mitochondria-dependent pathway as authenticated by caspase-9 activation and cytochrome c release to cytosol besides DNA laddering and annexin V-FITC double staining [86]. Aurein 1.2, another peptide of anuran origin showed anticancer effect in 50 out of 60 cancer cell lines tested against by National Cancer Institute (NCI) tumour line testing program [87].

NRC-03 & NRC-07

Pleurocidin-family cationic AMPs – NRC-03 & NRC-07 were shown to be cytotoxic to multiple breast cancer cells including MCF7-TX400 cells that overexpress p-glycoprotein and slow-growing breast adenocarcinoma (SKBR3) cells by binding to the negatively charged molecules on the cancer cells [88]. NRC-03 substantially reduces the median effective concentration EC50 of cisplatin for breast cells implying its use as a chemosensitizer. Both peptides killed breast cancer cells grown as xenografts in non-obese diabetic, severe combined immunodeficiency (NOD SCID) mice strains [88].

AMP containing the CisoDGRC (CDAK) motif displayed cytotoxicity in aminopeptidase (CD13) negative breast cancer cells MCF7 & MDA-MD-231 in vitro. The peptide reduced mitochondrial membrane potential, promoted caspase-3 and inhibited Bcl-2 expression in the breast cancer cell lines. Moreover, the peptide inhibited the progression of the xenograft tumour and the generation of neovascularization in nude mice in vivo [89].

Temporin-1CEa

Temporin-1CEa is a 17-residue amphipathic α-helical AMP isolated from the skin secretion of the Chinese brown frog Rana chensinensis and have been reported to exert rapid cytotoxicity against human breast cancer cell lines [90]. The anticancer mechanisms of the peptide against two human breast cancer cell lines, MCF7 and MDA-MB-23, have been investigated. Temporin-1CEa trigger rapid cell death in breast cancer cells by intracellular Ca2+ leakage, over-generation of reactive oxygen species (ROS), collapse of mitochondrial membrane potential [90]. Further study of temporin-1CEa on ERα negative human breast cancer cell line Bcap-37 yielded the same cytotoxic mechanisms in line with previous report [61].

TH1-5 & TH2-3

Hepcidins are cysteine-rich peptides that have been identified in many vertebrates including humans, amphibians, reptiles and fish [91]. Fish hepcidin have been shown to exert cytotoxicity activity against cancer cells. Tilapia hepcidin TH2-3, a synthetic 20-mer AMP from Oreochromis mossambicus, inhibited the growth and migration of HT1080 in a concentration-dependent manner. The peptide also caused lethal membrane disruption in HT1080 cancer cells. Real-time PCR and migration assay suggest that the peptide possess cytolytic activity and downregulatesthe c-jun gene in cancer cells [92]. TH1-5, another peptide from O. mossambicus inhibited the proliferation of HeLa, HepG2 (human hepatocellular carcinoma), and HT1080 tumour cells by altering membrane disruption and inducing apoptosis at low dose. In addition, TH1-5 also expressed modulation of immune-related genes [52].

Epinecidin-1 and Piscidin-1

Epinecidin-1 is asynthetic 21-mer peptide originally gotten from the grouper Epinephelus coioides. It displayed cytotoxic activity against several cancerous cell lines including A549, HT1080, HepG2, HeLa, human kidney cell (WS-1), mouse fibroblast cell (NIH3T3), murine hepatocyte cell (AML-12), human hepatic tumor-derived cell (HA59T/VGH) and mouse macrophage from Abelson murine leukaemia virus-induced tumour (RAW264.7) [54]. Epinecidin-1 increased the cytotoxicity of these cancer lines in a dose and time-dependent manner, which suggest that cell death occurred by membrane disruption [54]. Again it inhibited the proliferation of human leukemia cells U937 and induce apoptosis in response to cytokine reproduction [93]. Piscidin-1 is a 22-residue peptide with a cationic α-helical structure isolated from the mast cells of hybrid striped bass Morone saxatilis x M. chrysops [94]. Lin et al. showed that Piscidin-1 at low doses induce both apoptosis and necrosis in HT1080 cells. It also triggers a necrotic cell death pathway in a short period of high-dose treatment. In addition, piscidin-1 also inhibits the migration of HT1080 cells in a dose-dependent manner [94].

A9K & GW-H1

Resembling conventional surfactants, A9K is a short designed amphiphilic peptide that comprises a hydrophilic lysine residue as the C-terminus, followed by nine consecutive hydrophobic alanine residues [95]. The peptide exhibited high selectivity and dual modes of antitumour effects on the cancerous cells by membrane cell disruption and induction of cell apoptosis. It inhibited the growth of HeLa and human promyelocytic leukemia cells (HL60). One unique feature of this synthetic peptide is that it is not degraded by protease [95]. GW-H1 is a synthetic cationic amphipathic AMP with α-helix conformation. It inhibited the viability of three hepatocellular carcinoma cell line (J5, Huh7 and Hep3B) [64]. GW-H1 exerts high selective cytotoxicity against J5 cell line via caspase-dependent apoptosis. Also, in vivo it inhibited the growth of J5 xenografts in nude mice suggesting its potential antitumour capacity [64].

TsAP-1 & TsAP-2

TsAP-1 and TsAP-2 are two novel peptides obtained from the venom of the yellow Brazilian scorpion Tityus serrulatus. Both peptides are 17-mer amidated linear and non-disulfide bridged peptides. [58]. Lysine residue substitution of these two peptides enhanced their potency against five human cancer cell lines namely human squamous adenocarcinoma cell line (NCI-HI57), human lung adenocarcinoma cell line (NCI-H838), human androgen-independent prostate adenocarcinoma cell line (PC-3), human glioblastoma cell line (U251) and MCF7. TsAP-2 is more potent of the two; has IC50 values ranging between 0.83 and 2.0µM [58].

Lactoferricin

Lactoferrricin is a 25-amino acid AMP obtained by acid-pepsin hydrolysis of mammalian lactoferrin [57]. The amphipathic cationic peptide has been reported to be cytotoxic to murine and human cancer cell lines including leukemia, fibrosarcoma, various carcinomas and neuroblastoma cells [96–99]; this cytotoxicity occur at concentrations that do not significantly affect normal cells such as normal fibroblasts, lymphocytes, epithelial cells, endothelial cells or erythrocytes [98,100]. In vivo, LfcinB also exerts potent antitumour effect on mice inoculated with L5178-ML25 murine lymphoma and B16-BL6 murine melanoma cells [96]. Limitations of this peptide as a potential anticancer therapeutic is its susceptibility to enzymatic digestion as well as inactivation by anionic serum components.

Gomesin

Gomesin is a potent 18-amino residue AMP isolated from haemocytes of the spider Acanthoscurria gomesiana; its anticancer activity both in vitro and in vivo has been tested [60]. Gomesin exerted direct cytotoxicity on murine and human tumour cells in vitro with an estimated IC50 for the murine melanoma B16F10-Nex2 being 3.5µM and <10 µM for the human tumour cell lines (HeLa, SKBR3, LS180). In vivo, local treatment with gomesin-containing cream significantly increased the survival time of murine melanoma-challenged mice as shown by delayed growth [60].

BMAP-27 & BMAP-28

Bovine myeloid antimicrobial peptides (BMAP) are α-helical cationic peptides of the cathelicidin family. Despite their narrow therapeutic range, few studies have evidenced that they possess cytotoxic activity on certain human leukemic cells by apoptotic induction [101–103]. At high doses, they cause the hemolysis of benign erythrocytes and leukocytes [101,103].

Tachyplesin

Isolated from the haemocytes of the horseshoe crab Tachypleus tridentatus, the 17-amino acid peptide is characterized by two antiparallel β-sheets rigidly held in place by two disulphide bonds. Its antitumour mechanism involve a multistep model in which the peptide binds to hyaluronan on the surfaces of target cells and forms a membrane attack complex (MAC) at the final stage. This complex consequently disrupt the plasma membrane integrity and thereby kill the target cells [55]. Preferential expression of hyaluronan on the surface of malignant and endothelial cells involved in cancer vascularization suggests that the peptide inhibit the proliferation of cells relative to other non-tumorigenic cell lines that express less of hyaluronan on their surfaces [104]. Tachyplesin has been shown to inhibit malignant growth in the presence of normal serum even against cells that over express the multiple-drug resistant gene [55]. A synthetic version of tachyplesin conjugated to the integrin homing domain RGD blocked the growth of tumor cells both in vitro and in vivo [104]. RGD-tachyplesin has been shown to inhibit the proliferation of both cultured malignant and endothelial cells and reduced the colony formation of TSU prostate cancer cells. In vivo, it has been reported to inhibit the growth of neoplasms via induction of apoptosis in both malignant and endothelial cells evidenced by activation of several caspases in both the mitochondrial and Fas-dependent pathways  [55].

CONCLUSION

Anticancer antimicrobial peptides hold promise to yielding novel drugs for the treatment of cancer that could replace the traditional neoplastic agents or used in combination therapy. Several studies have documented the remarkable activities of these molecules in several cancers; this is evident in the number of the ACPs in the database and documented literature. Nevertheless, further studies into this emerging field of anticancer peptide need to be embraced. More in vitro studies need to be conducted correlating with the effects reported on numerous cancer cell lines. Understanding the detailed and precise mechanisms of this class of agents and structure-activity relationship will provide a knowledge platform for answering some of unanswered questions about AMPs and designing superior peptides. Instability and proteolytic degradation of the peptides need to be further studied as bypassing this ‘hostility’ allow these agents to exert their full therapeutic potentials and perhaps decipher some activities yet unknown.

CONFLICT OF INTERESTS

The authors declare no conflict of interest

REFERENCES

  1. Xiao X, Wang P, Lin W, Jia J, Chou K. iAMP-2L: A two-level multi-label classifier for identifying antimicrobial peptides and their functional types. Anal Biochem 2013;436:168–77.
  2. Seo M, Won H, Kim J, Mishig-ochir T, Lee B. Antimicrobial peptides for therapeutic applications: a review. Mol 2012;17:12276–86.
  3. Zasloff M. Antimicrobial peptides of multicellular organisms. Nat 2002;415:389–95.
  4. Pushpanathan M, Gunasekaran P, Rajendhran J. Antimicrobial Peptides: versatile biological properties. Int J Pept 2013;2013:1–15.
  5. Kamysz W, Okrój M, Łukasiak J. Novel properties of antimicrobial peptides. Acta Biochim Pol 2003;50:461–9.
  6. Pasupuleti M, Schmidtchen A, Malmsten M. Antimicrobial peptides: key components of the innate immune system. Crit Rev Biotechnol 2012;32:143–71.
  7. Wang G. Human antimicrobial peptides and proteins. Pharm 2014;7:545–94.
  8. Li Y, Xiang Q, Zhang Q, Huang Y, Su Z. Overview on the recent study of antimicrobial peptides: origins, functions, relative mechanisms and application. Pept 2012;37:207–15.
  9. Gajski G, Garaj-Vrhovac V. Melittin: a lytic peptide with anticancer properties. Environ Toxicol Pharmacol 2013;36:697–705.
  10. Aerts AM, Franåois IEJA, Cammue BPA, Thevissen K. Review The mode of antifungal action of plant, insect and human defensins. Cell Mol Life Sci 2008;65:2069–79.
  11. Václav Č, Bém R. Lucifensins, the insect defensins of biomedical importance: the story behind the maggot therapy. Pharm 2014;7:251–64.
  12. Koo JC, Lee SY, Chun HJ, Cheong YH, Choi JS, Kawabata SI, et al. Two hevein homologs isolated from the seed of Pharbitis nil L. exhibit potent antifungal activity. Biochim Biophys Acta-Protein Struct Mol Enzymol 1998;1382:80–90.
  13. Orrù S, Scaloni A, Giannattasio M, Urech K, Pucci P, Schaller G. Amino acid sequence, S-S bridge arrangement and distribution in plant tissues of thionins from Viscum album. Biol Chem 1997;378:989–96.
  14. Sharma S, Verma HN, Sharma NK. Cationic bioactive peptide from the seeds of Benincasa hispida. Int J Pept 2014;2014:1–12.
  15. Huang PH, Chen JY, Kuo CM. Three different hepcidins from tilapia, Oreochromis mossambicus: analysis of their expressions and biological functions. Mol Immunol 2007;44:1922–34.
  16. Buonocore F, Randelli E, Casani D, Picchietti S, Belardinelli MC, de Pascale D, et al. A piscidin-like antimicrobial peptide from the icefish Chionodraco hamatus (Perciformes: Channichthyidae): molecular characterization, localization and bactericidal activity. Fish Shellfish Immunol 2012;33:1183–91.
  17. Cho JH, Sung BH, Kim SC. Buforins: histone H2A-derived antimicrobial peptides from toad stomach. Biochim Biophys Acta 2009;1788:1564–9.
  18. Conlon JM, Mechkarska M, Lukic ML, Flatt PR. Potential therapeutic applications of multifunctional host-defense peptides from frog skin as anti-cancer, anti-viral, immunomodulatory, and anti-diabetic agents. Pept 2014;57:67–77.
  19. Coronado MA, Georgieva D, Buck F, Gabdoulkhakov AH, Ullah A, Spencer PJ, et al. Purification, crystallization and preliminary X-ray diffraction analysis of crotamine, a myotoxic polypeptide from the Brazilian snake Crotalus durissus terrificus. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012;68:1052–4.
  20. Wang Y, Hong J, Liu X, Yang H, Liu R, Wu J, et al. Snake cathelicidin from Bungarus fasciatus is a potent peptide antibiotics. PLoS One 2008;3:e3217.
  21. Stegemann C, Kolobov A, Leonova YF, Knappe D, Shamova O, Ovchinnikova TV, et al. Isolation, purification and de novo sequencing of TBD-1, the first beta-defensin from leukocytes of reptiles. Proteomics 2009;9:1364–73.
  22. Zhang G, Sunkara LT. Avian antimicrobial host defense peptides: from biology to therapeutic applications. Pharm 2014;7:220–47.
  23. Wu J, Gao B, Zhu S. The fungal defensin family enlarged. Pharm 2014;7:866–80.
  24. Xiong YQ, Hady WA, Deslandes A, Rey A, Fraisse L, Kristensen HH, et al. Efficacy of NZ2114, a novel plectasin-derived cationic antimicrobial peptide antibiotic, in experimental endocarditis due to methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother 2011;55:5325–30.
  25. Gyawali R, Ibrahim SA. Natural products as antimicrobial agents. Food Control 2014;46:412–29.
  26. Lee DG, Hahm K-S, Park Y, Kim H-Y, Lee W, Lim S-C, et al. Functional and structural characteristics of anticancer peptide Pep27 analogues. Cancer Cell Int 2005;5:21.
  27. McManus AM, Dawson NF, Wade JD, Carrington LE, Winzor DJ, Craik DJ. Three-dimensional structure of RK-1:a novel alpha-defensin peptide. Biochem 2000;39:15757–64.
  28. Jenssen H, Hamill P, Hancock REW. Peptide antimicrobial agents. Clin Microbiol Rev 2006;19:491–511.
  29. Hancock REW, Sahl H-G. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat Biotechnol 2006;24:1551–7.
  30. Yang P, Ramamoorthy A, Chen Z. Membrane orientation of MSI-78 measured by sum frequency generation vibrational spectroscopy. Langmuir 2011;27:7760–7.
  31. Brogden KA. Antimicrobial peptides: pore formers or metabolic inhibitors in bacteria? Nat Rev Microbiol 2005;3:238–50.
  32. Pouny Y, Rapaport D, Mor A, Nicolas P, Shai Y. Interaction of antimicrobial dermaseptin and its fluorescently labeled analogues with phospholipid membranes. Biochem 1992;31:12416–23.
  33. Zhang L, Rozek A, Hancock RE. Interaction of cationic antimicrobial peptides with model membranes. J Biol Chem 2001;276:35714–22.
  34. Silva PM, Gonçalves S, Santos NC. Defensins: antifungal lessons from eukaryotes. Front Microbiol 2014;5:1–17.
  35. Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med 2006;3:2011–30.
  36. Papo N, Shai Y. Host defense peptides as new weapons in cancer treatment. Cell Mol Life Sci 2005;62:784–90.
  37. Gaspar D, Veiga AS, Castanho MARB. From antimicrobial to anticancer peptides. A review. Front Microbiol 2013;4:294.
  38. Oren Z, Shai Y. Mode of action of linear amphipathic alpha-helical antimicrobial peptides. Biopolymers 1998;47:451–63.
  39. Shai Y. Mechanism of the binding, insertion and destabilization of phospholipid bilayer membranes by alpha-helical antimicrobial and cell non-selective membrane-lytic peptides. Biochim Biophys Acta-Biomembr 1999;1462:55–70.
  40. Mai JC, Mi Z, Kim SH, Ng B, Robbins PD. A proapoptotic peptide for the treatment of solid tumors. Cancer Res 2001;61:7709–12.
  41. Hwang C, Heath EI. Angiogenesis inhibitors in the treatment of prostate cancer. J Hematol Oncol 2010;3:1–12.
  42. Starzec A, Vassy R, Martin A, Lecouvey M, Di Benedetto M, Crépin M, et al. Antiangiogenic and antitumor activities of peptide inhibiting the vascular endothelial growth factor binding to neuropilin-1. Life Sci 2006;79:2370–81.
  43. Wu D, Gao Y, Chen L, Qi Y, Kang Q, Wang H, et al. Anti-tumor effects of a novel chimeric peptide on S180 and H22 xenografts bearing nude mice. Pept 2010;31:850–64.
  44. Wu D, Gao Y, Qi Y, Chen L, Ma Y, Li Y. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett 2014;351:13–22.
  45. Schweizer F. Cationic amphiphilic peptides with cancer-selective toxicity. Eur J Pharmacol 2009;625:190–4.
  46. Li J-T, Zhang J-L, He H, Ma Z-L, Nie Z-K, Wang Z-Z, et al. Apoptosis in human hepatoma HepG2 cells induced by corn peptides and its anti-tumor efficacy in H22 tumor bearing mice. Food Chem Toxicol 2013;51:297–305.
  47. Xue Z, Liu Z, Wu M, Zhuang S, Yu W. Effect of rapeseed peptide on DNA damage and apoptosis in Hela cells. Exp Toxicol Pathol 2010;62:519–23.
  48. Prasanthi K, Lily Y. Apoptosis signaling pathway and resistance to apoptosis in breast cancer stem cells. In: George GC, Lai PBS(eds) Apoptosis in carcinogenesis and Chemotherapy. Springer Science & Business Media; 2009.
  49. Fang XY, Chen W, Fan JT, Song R, Wang L, Gu YH, et al. Plant cyclopeptide RA-V kills human breast cancer cells by inducing mitochondria-mediated apoptosis through blocking PDK1-AKT interaction. Toxicol Appl Pharmacol 2013;267:95–103.
  50. Liu J-J, Lin M, Yu J-Y, Liu B, Bao J-K. Targeting apoptotic and autophagic pathways for cancer therapeutics. Cancer Lett 2011;300:105–14.
  51. Fernandez DI, Sani M, Miles AJ, Wallace BA, Separovic F. Membrane defects enhance the interaction of antimicrobial peptides, aurein 1.2 versus caerin 1.1. BBA-Biomembr 2013;1828:1863–72.
  52. Chang W-T, Pan C-Y, Rajanbabu V, Cheng C-W, Chen J-Y. Tilapia (Oreochromis mossambicus) antimicrobial peptide, hepcidin 1-5, shows antitumor activity in cancer cells. Pept 2011;32:342–52.
  53. Aoki W, Ueda M. Characterization of antimicrobial peptides toward the development of novel antibiotics. Pharm 2013;6:1055–81.
  54. Lin W-J, Chien Y-L, Pan C-Y, Lin T-L, Chen J-Y, Chiu S-J, et al. Epinecidin-1, an antimicrobial peptide from fish (Epinephelus coioides) which has an antitumor effect like lytic peptides in human fibrosarcoma cells. Pept 2009;30:283–90.
  55. Chen J, Xu X, Underhill CB, Yang S, Wang L. Tachyplesin activates the classic complement pathway to kill tumor cells. Cancer Res 2005;65:4614–22.
  56. Huang C-Y, Huang H-Y, Forrest MD, Pan Y-R, Wu W-J, Chen H-M. Inhibition effect of a custom peptide on lung tumors. PLoS One 2014;9:e109174.
  57. Pepe G, Tenore GC, Mastrocinque R, Stusio P, Campiglia P. Potential anticarcinogenic peptides from bovine milk. J Amino Acids 2013;2013:939804.
  58. Guo X, Ma C, Du Q, Wei R, Wang L, Zhou M, et al. Two peptides, TsAP-1 and TsAP-2, from the venom of the Brazilian yellow scorpion, Tityus serrulatus: evaluation of their antimicrobial and anticancer activities. Biochim 2013;95:1784–94.
  59. Conlon JM, Mechkarska M. Host-defense peptides with therapeutic potential from skin secretions of frogs from the family pipidae. Pharm 2014;7:58–77.
  60. Rodrigues EG, Dobroff ASS, Cavarsan CF, Paschoalin T, Nimrichter L, Mortara RA, et al. Effective topical treatment of subcutaneous murine B16F10-NEX2 melanoma by the antimicrobial peptide gomesin. Neoplasia 2008;10:61–8.
  61. Wang C, Zhou Y, Li S, Li H, Tian L, Wang H, et al. Anticancer mechanisms of temporin-1CEa, an amphipathic α-helical antimicrobial peptide, in Bcap-37 human breast cancer cells. Life Sci 2013;92:1004–14.
  62. Vad BS, Bertelsen K, Johansen CH, Pedersen JM, Skrydstrup T, Nielsen NC, et al. Pardaxin permeabilizes vesicles more efficiently by pore formation than by disruption. Biophys J 2010;98:576–85.
  63. Kim J-K, Lee S-A, Shin S, Lee J-Y, Jeong K-W, Nan YH, et al. Structural flexibility and the positive charges are the key factors in bacterial cell selectivity and membrane penetration of peptoid-substituted analog of Piscidin1. Biochim Biophys Acta 2010;1798:1913–25.
  64. Chen Y-LS, Li J-H, Yu C-Y, Lin C-J, Chiu P-H, Chen P-W, et al. Novel cationic antimicrobial peptide GW-H1 induced caspase-dependent apoptosis of hepatocellular carcinoma cell lines. Pept 2012;36:257–65.
  65. Bechinger B. Structure and functions of channel-forming peptides: Magainins, cecropins, melittin and alamethicin. J Membr Biol 1997;156:197–211.
  66. Huang Y-B, He L-Y, Jiang H-Y, Chen Y-X. Role of helicity on the anticancer mechanism of action of cationic-helical peptides. Int J Mol Sci 2012;13:6849–62.
  67. Huang Y-B, Wang X-F, Wang H-Y, Liu Y, Chen Y. Studies on mechanism of action of anticancer peptides by modulation of hydrophobicity within a defined structural framework. Mol Cancer Ther 2011;10:416–26.
  68. Suttmann H, Retz M, Paulsen F, Harder J, Zwergel U, Kamradt J, et al. Antimicrobial peptides of the cecropin-family show potent antitumor activity against bladder cancer cells. BMC Urol 2008;8:5.
  69. Wu J-M, Jan P-S, Yu H-C, Haung H-Y, Fang H-J, Chang Y-I, et al. Structure and function of a custom anticancer peptide, CB1a. Pept 2009;30:839–48.
  70. Lehmann J, Retz M, Sidhu SS, Suttmann H, Sell M, Paulsen F, et al. Antitumor activity of the antimicrobial peptide magainin II against bladder cancer cell lines. Eur Urol 2006;50:141–7.
  71. Miyazaki Y, Aoki M, Yano Y, Matsuzaki K. Interaction of antimicrobial peptide magainin 2 with gangliosides as a target for human cell binding. Biochem 2012;51:10229–35.
  72. Soballe PW, Maloy WL, Myrga ML, Jacob LS, Herlyn M. Experimental local therapy of human melanoma with lytic magainin peptides. Int J Cancer 1995;60:280–4.
  73. Ohsaki Y, Gazdar AF, Chen H, Johnson BE. Antitumor activity of magainin analogues against human lung cancer cell lines. Cancer Res 1992;52:3534–8.
  74. Baker MA, Maloy WL, Zasloff M, Jacob LS. Anticancer efficacy of magainin2 and analogue peptides. Cancer Res 1993;53:3052–7.
  75. Bessalle R, Kapitkovsky A, Gorea A, Shalit I, Fridkin M. All-D-magainin: Chirality, antimicrobial activity and proteolytic resistance. FEBS Lett 1990;274:151–5.
  76. Moon D-O, Park S-Y, Heo M-S, Kim K-C, Park C, Ko WS, et al. Key regulators in bee venom-induced apoptosis are Bcl-2 and caspase-3 in human leukemic U937 cells through downregulation of ERK and Akt. Int Immunopharmacol 2006;6:1796–807.
  77. Slaninová J, Mlsová V, Kroupová H, Alán L, Tůmová T, Monincová L, et al. Toxicity study of antimicrobial peptides from wild bee venom and their analogs toward mammalian normal and cancer cells. Pept 2012;33:18–26.
  78. Sørensen OE, Follin P, Johnsen AH, Calafat J, Sandra Tjabringa G, Hiemstra PS, et al. Human cathelicidin, hCAP-18, is processed to the antimicrobial peptide LL-37 by extracellular cleavage with proteinase 3. Blood 2001;97:3951–9.
  79. Okumura K, Itoh A, Isogai E, Hirose K, Hosokawa Y, Abiko Y, et al. C-terminal domain of human CAP18 antimicrobial peptide induces apoptosis in oral squamous cell carcinoma SAS-H1 cells. Cancer Lett 2004;212:185–94.
  80. Li X, Li Y, Han H, Miller DW, Wang G. Solution structures of human ll-37 fragments and NMR-based identification of a minimal membrane-targeting antimicrobial and anticancer region. J Am Chem Soc 2006;128:5776–85.
  81. Johansson J, Gudmundsson GH, Rottenberg ME, Berndt KD, Agerberth B. Conformation-dependent antibacterial activity of the naturally occurring human peptide LL-37. J Biol Chem 1998;273:3718–24.
  82. Wu S-P, Huang T-C, Lin C-C, Hui C-F, Lin C-H, Chen J-Y. Pardaxin, a fish antimicrobial peptide, exhibits antitumor activity toward murine fibrosarcoma in vitro and in vivo. Mar Drugs 2012;10:1852–72.
  83. Huang T-C, Lee J-F, Chen J-Y. Pardaxin, an antimicrobial peptide, triggers caspase-dependent and ROS-mediated apoptosis in HT-1080 cells. Mar Drugs 2011;9:1995–2009.
  84. Hsu JC, Lin LC, Tzen JTC, Chen JY. Pardaxin-induced apoptosis enhances antitumor activity in HeLa cells. Pept 2011;32:1110–6.
  85. Al-Benna S, Shai Y, Jacobsen F, Steinstraesser L. Oncolytic activities of host defense peptides. Int J Mol Sci 2011;12:8027–51.
  86. Lee HS, Park CB, Kim JM, Jang SA, Park IY, Kim MS, et al. Mechanism of anticancer activity of buforin IIb, a histone H2A-derived peptide. Cancer Lett 2008;271:47–55.
  87. Rozek T, Wegener KL, Bowie JH, Olver IN, Carver JA, Wallace JC, et al. The antibiotic and anticancer active aurein peptides from the Australian Bell Frogs Litoria aurea and Litoria raniformis: The solution structure of aurein 1.2. Eur J Biochem 2000;267:5330–41.
  88. Hilchie AL, Doucette CD, Pinto DM, Patrzykat A, Douglas S, Hoskin DW. Pleurocidin-family cationic antimicrobial peptides are cytolytic for breast carcinoma cells and prevent growth of tumor xenografts. Breast Cancer Res 2011;13:R102.
  89. Hou L, Zhao X, Wang P, Ning Q, Meng M, Liu C. Antitumor activity of antimicrobial peptides containing CisoDGRC in CD13 negative breast cancer cells. PLoS One 2013;8:e53491.
  90. Wang C, Tian LL, Li S, Li HB, Zhou Y, Wang H, et al. Rapid cytotoxicity of antimicrobial peptide tempoprin-1cea in breast cancer cells through membrane destruction and intracellular calcium mechanism. PLoS One 2013;8:e60462.
  91. Masso-Silva JA, Diamond G. Antimicrobial peptides from fish. Pharm 2014;7:265–310.
  92. Chen J-Y, Lin W-J, Lin T-L. A fish antimicrobial peptide, tilapia hepcidin TH2-3, shows potent antitumor activity against human fibrosarcoma cells. Pept 2009;30:1636–42.
  93. Chen J-Y, Lin W-J, Wu J-L, Her GM, Hui C-F. Epinecidin-1 peptide induces apoptosis which enhances antitumor effects in human leukemia U937 cells. Pept 2009;30:2365–73.
  94. Lin H-J, Huang T-C, Muthusamy S, Lee J-F, Duann Y-F, Lin C-H. Piscidin-1, an antimicrobial peptide from fish (hybrid striped bass Morone saxatilis x M. chrysops), induces apoptotic and necrotic activity in HT1080 cells. Zoolog Sci 2012;29:327–32.
  95. Xu H, Chen CX, Hu J, Zhou P, Zeng P, Cao CH, et al. Dual modes of antitumor action of an amphiphilic peptide A(9)K. Biomaterials 2013;34:2731–7.
  96. Yoo Y-C, Watanabe R, Koike Y, Mitobe M, Shimazaki K, Watanabe S, et al. Apoptosis in human leukemic cells induced by lactoferricin, a bovine milk protein-derived Peptide: involvement of reactive oxygen species. Biochem Biophys Res Commun 1997;237:624–8.
  97. Tone Eliassen L, Berge G, Sveinbjørnsson B, Svendsen JS, Vorland LH, Rekdal Ø. Evidence for a direct antitumor mechanism of action of bovine lactoferricin. Anticancer Res 2002;22:2703–10.
  98. Mader JS, Salsman J, Conrad DM, Hoskin DW. Bovine lactoferricin selectively induces apoptosis in human leukemia and carcinoma cell lines. Mol Cancer Ther 2005, 4:612–24.
  99. Eliassen LT, Berge G, Leknessund A, Wikman M, Lindin I, Løkke C, et al. The antimicrobial peptide, Lactoferricin B, is cytotoxic to neuroblastoma cells in vitro and inhibits xenograft growth in vivo. Int J Cancer 2006;119:493–500.
  100. Furlong SJ, Mader JS, Hoskin DW. Lactoferricin-induced apoptosis in estrogen-nonresponsive MDA-MB-435 breast cancer cells is enhanced by C6 ceramide or tamoxifen. Oncol Rep 2006;15:1385–90.
  101. Risso A, Zanetti M, Gennaro R. Cytotoxicity and apoptosis mediated by two peptides of innate immunity. Cell Immunol 1998;189:107–15.
  102. Risso A, Braidot E, Sordano MC, Vianello A, Macrı F, Skerlavaj B, et al. BMAP-28, an antibiotic peptide of innate immunity, induces cell death through opening of the mitochondrial permeability transition pore. Mol Cell Biol 2002;22:1926–35.
  103. Skerlavaj B, Gennaro R, Bagella L, Merluzzi L, Risso A, Zanetti M. Biological characterization of two novel cathelicidin-derived peptides and identification of structural requirements for their antimicrobial and cell lytic activities. J Biol Chem 1996;271:28375–81.
  104. Chen Y, Xu X, Hong S, Chen J, Liu N, Underhill CB, et al. RGD-tachyplesin inhibits tumor growth. Cancer Res 2001;61:2434–8.