Open Access

Mitochondria targeting nano agents in cancer therapeutics (Review)

Retraction in: /10.3892/ol.2020.12101

  • Authors:
    • Xiao‑Ying Zhang
    • Pei-Ying Zhang
  • View Affiliations

  • Published online on: October 21, 2016     https://doi.org/10.3892/ol.2016.5302
  • Pages: 4887-4890
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mitochondria have emerged as noteworthy therapeutic targets as their physiological functions are often altered in pathological conditions such as cancer. The electronic databases of MEDLINE, EMBASE and PubMed were searched for recent studies reporting the importance of mitochondria targeting nanoagents in cancer therapeutics. The concluding remarks of the above papers mostly confirmed the growing potential of these novel nanoagents in the area of anticancer research. Furthermore, numerous studies demonstrated the immense potential of nanocarriers in delivering mitochondria‑acting compounds to their target site. Among the assemblage of nanomaterials, carbon nanotubes (CNTs) are becoming more prominent for drug delivery due to favorable attributes including their unique shape, which promotes cellular uptake, and large aspect ratio that facilitates conjugation of bioactive molecules on their surface. The present review focused on the current view of variable options available in mitochondria‑targeting anticancer therapeutics. It may be concluded that improvements are essential for its establishment as a gold standard therapeutic option especially in the clinical setting.

Introduction

The concept of mitochondrial structural equilibrium is influenced by various metabolic and cellular signals that may significantly affect mitochondrial physiology, and substantially and ultimately impact cellular health (1,2). This concept in the recent research has been exploited to form new drugs for the efficient management of various diseases including diabetes, Alzheimers diseases, Parkinsons diseases and cancer (3,4). Compared to normal cells, mitochondria of cancer cells exhibit a stark difference in terms of structure and function (5). Cancer cells have adapted to a series of metabolic changes in order to achieve better survival regardless of existing conditions such as availability of nutrients. These are detailed in the well-regarded Warburg effect, named after Warburg who first reported this phenomenon in 1927 (6,7). The Warburg effect describes malignant metabolic alterations encompassing elevated glycolysis as well as defects in mitochondrial oxidative respiration (8). This extensive metabolic reprogramming also influences significantly the various catabolic processes of cancer cells including apoptosis, necrosis and autophagy (9).

Despite the numerous therapeutic advantages that may be realized by targeting mitochondria, there are very few examples of mito-active drugs (10). This is reflected by the challenges of delivering compounds to this organelle, which possesses a convoluted and complex membrane structure that prohibits access to its inner space. A well-established approach is to exploit the negative charge of the mitochondrial matrix in order to turn compounds toward mitochondria. This electrochemical gradient facilitates the accumulation of positively charged molecules (cations) in the mitochondrial matrix (11). However, translocation is energetically unfavorable for molecules that are hydrophilic in nature (12). This category of lipophilic and membrane-permeable cations is then able to pass through the hydrophobic interior of the phospholipid bilayer of biological membranes while retaining their overall positive charge. As a result, these lipophilic cations, for example the commonly used mitochondrial fluorescence dyes, such as JC-1 or MitoTracker® dyes, are capable of accumulating at mitochondria (13).

DLCs for targeting to mitochondria

Although a wide range of delocalized lipophilic cations (DLCs) may in principle be utilised for drug targeting to mitochondria, only a few such compounds have been extensively researched for this purpose. The most prominent example is triphenylphosphonium (TPP) cation, a positively charged phosphorus atom surrounded by three hydrophobic phenyl groups that impart an extended hydrophobic surface (14). The TPP moiety has been employed as a probe to explore the mitochondrial membrane potential for over 40 years and consequently its behaviour and interaction with mitochondria are well defined (11). The relative TPP concentration inside the negatively charged membrane compartments increases by one order of magnitude for every 60 mV of negative membrane potential. Due to the active transport of salt ions by membrane-bound pumps, the interior part of the plasma membrane is negatively charged relative to the exterior side. As a result, the plasma membrane potential ranges generally between −30 and −60 mV, which is sufficient to promote up to 10-fold accumulation of TPP inside the cell. Typical mitochondrial membrane potential is up to −180 mV, which facilitates a 1,000-fold accumulation of TPP inside mitochondria (10).

The most extensively investigated example showing enhanced therapeutic efficacy conferred by conjugation with TPP for mitochondrial targeting is MitoQ (15). Another example, gamitrinib, is the mitochondrial-targeted Hsp90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) via TPP or 1–4 tandem repeats of cyclic guanidinium (16). The construct exhibited profound improvement in efficacy compared to 17-AAG with excellent selectivity for cancer cells/tissues in vitro and in vivo. Accordingly, gamitrinib is currently being evaluated in clinical settings (17).

Another group of DLCs that have been examined for their application in directing compounds to mitochondria is the rhodamine group of fluorescent compounds. Rhodamine compounds have been used as histological stains and some of the members have been utilized in investigating the energy state of isolated mitochondria (18).

Peptide-based sequence for targeting to mitochondria

Peptide-based sequences form another prominent branch of mitochondrial-targeting moiety apart from DLCs. Peptide-based mitochondrial molecular transporters possess some advantages over DLCs, including better biocompatibility and relatively straightforward synthesis with amide coupling. These sequences are normally derived from cytosolic synthesized proteins destined for trafficking to mitochondria (19). In adition to the 13 proteins encoded by mtDNA, the remaining mitochondrial proteins are ciphered in the nuclear genome and synthesized in the cytosol. Sorting to mitochondria is facilitated through the N-terminal mitochondrial targeting sequence (MTS), in which Arg, Ala, or Ser are abundant, while negatively charged amino acids such as Asp or Glu are rare (20). Despite the weak consensus between different MTSs, they consist of alternating hydrophobic amino acids and positively charged amino acids that form amphiphilic α-helix, which is crucial for the import of nascent proteins into mitochondria (20). Nevertheless, these naturally occurring MTSs are lengthy and their application in targeted delivery is limited by challenges in generating conjugates that are soluble and cell permeable (4). In view of this, several synthetic peptides have been designed and reported to have mitochondrial targeting activity. Notably, some of them also have intrinsic pharmacological properties as well. For example, the Szeto-Schiller (SS) peptide antioxidants is a class of short peptide antioxidants that also possess the ability to penetrate into IMM (21).

Nanocarriers and mitochondrial delivery

A therapeutic compound may be selectively delivered to mitochondria via covalent conjugation with a mitochondrial-targeting moiety. Nevertheless, this strategy is inevitably accompanied by limitations. For example, DLCs are ineffective in directing large polar cargo and intrinsically toxic to mitochondria at high concentrations. In addition, the covalent conjugation of bioactive compounds with DLCs entails modification of the structure, which may lead to alteration of chemical properties and thus the resulting activity. As a result, an alternative DS that can overcome these limitations, i.e., developing a mitochondrial-targeting carrier to incorporate the drug without modification, is highly demanded.

In light of this, great attention has been recently devoted to the development of a DS that is able to: i) Cross several biological barriers; ii) protect bioactive agents from premature deactivation; and iii) elevate intracellular bioavailability of the drugs at their target site (22). One of the emerging DS is represented by nanocarriers, i.e., nano-sized materials (possessing at least one dimension in the range of 1–100 nm) that can incorporate/encapsulate therapeutic compounds and be functionalized to carry multiple moieties such as imaging agents or targeting ligands at their surface. Nanocarriers, encompassing natural and synthetic polymer conjugates, polymeric nanoparticles, lipid-based carriers (such as liposomes and micelles), dendrimers, carbon nanotubes (CNTs), gold, platinum and titanium oxide nanoparticles (including nano-shells and nano-cages), have been subjected to intensive research for potential therapeutic applications (23).

A favourable attribute of the nanocarrier as effective DS for cancer therapy is the ability to passively accumulate at tumour tissues due to an enhanced permeability and retention (EPR) effect. Accordingly, the newly formed tumour vessels for supplying nutrients to rapidly growing tumour mass are usually abnormal in architecture with poor alignment and wide fenestrations. In addition, it is well recognized that tumour tissues lack effective lymphatic drainage (24). This allows nanocarriers to preferentially accumulate at the tumour sites (24). Such a property has been harnessed for drug delivery to neoplastic tissues, i.e., passive targeting. Additionally, due to the high surface area-to-volume ratio of the nanocarrier, it is possible to achieve high ligand density of targeting moiety on the surface for selective targeting (25). Thus, nanocarriers potentially increase local drug concentration at the tumour site by carrying drug molecules to the tumour and releasing them only in response to altered physical conditions specific to the tumour microenvironment. Thus, in the context of targeting mitochondria for cancer therapy, the development of nanocarriers to carry mito-active agents for improved targeting and bioavailability at mitochondria is of great interest.

Polymer-based nanocarriers directed to mitochondria

The engineering of mitochondrial-targeting polymeric nanoparticles demonstrated the flexible application of this nanocarrier for therapeutics against diseases such as cancer, diabetes, and Alzheimers disease (26). Notably, FDA-approved poly(lactic-co-glycolic acid) (PLGA) and polyethylene glycol (PEG)-based materials were used to synthesize the nanoparticle, providing a distinctive edge in terms of biocompatibility. This accentuates the potential of advancing the nanoparticle to clinical settings in the near future. The mitochondrial-targeted PLGA-b-PEG-TPP copolymer was synthesized by conjugating TPP at the terminal end of PEG. The polymeric nanoparticle was assembled by a polymer-blending technology via the nanoprecipitation method (27), which is capable of fine-tuning the resulting size and surface charges for optimal mitochondrial localization.

Liposome-like nanocarriers for mitochondrial targeting

One of the most extensively reviewed mitochondrial-targeted nanocarriers is dequalinium-based liposome-like vesicles (DQAsomes) (28). Dequalinium is a molecule with two symmetrical cationic charge centres separated by a hydrophobic carbon chain. DQAsome is made from the self-assembly of dequalinium chloride into vesicle-like aggregates in an aqueous suspension. This cationic colloidal DS formed is converted to DQAsome/DNA complexes by interaction with DNA. DQAplexes were shown to enter cells via endocytosis (29). The advantage of the DQAsome is the ability to evade endosomal compartments.

Carbon nanotube and mitochondria

Given the versatility and effectiveness of CNTs in delivering various drugs, the role of CNTs in mitochondrial medicine has been investigated. The application of CNTs in organellar targeting is currently at its infancy stage. In an exploratory study, fluorescein isothiocyanate (FITC)-labelled PL-PEG (PL-PEG-FITC)-functionalized SWCNTs (SWCNT-PL-PEG-FITC) were serendipitously found to be localized at mitochondria in tumour and normal cells (HeLa, MCF-7, ASTC-a-1, COS-7 and EVC304) in a mitochondrial transmembrane potential-dependent manner (30). By contrast, in RAW264.7 murine macrophage, SWCNT-PL-PEG-FITC was observed to be mainly distributed in lysosomes instead of mitochondria, possibly due to lysosomal trafficking following phagocytosis. Notably, when SWCNTs were functionalized with molecules that are normally internalized by receptor-mediated endocytosis (FA and bovine serum albumin), the entire constructs were found to distribute to lysosomes (31). However, the SWCNTs would travel to mitochondria ensuing direct membrane penetration when the functionalized moiety was a small molecule chemical compound (FITC and PI). Since SWCNTs are a potent NIR-absorbing nanomaterial for the thermal destruction of tumour cells (31), this specific mitochondrial localization property was capitalized. It was observed that SWCNT-PL-PEG induced apoptosis in HeLa cells via mitochondrial damages after irradiation with NIR laser.

The design and synthesis of CNTs for active mitochondrial targeting has been reported recently (19). In that study, MWCNTs were functionalized covalently with an established MTS derived from the N-terminal region of subunit VIII of human cytochrome c oxidase. The unique mitochondrial localizing property of the MTS-MWCNTs, which was fluorescently labelled with a sulforhodamine B probe, was validated in RAW264.7 and HeLa cells using wide-field epifluorescence microscopy, confocal microscopy, and TEM (19). The colocalization of the conjugates with mitochondria was further confirmed by the presence of MWCNTs in isolated mitochondria observed under TEM. Different from the findings of Zhou et al (30), whereby the active design of CNTs for mitochondrial targeting was unnecessary, in the present study, targeting peptide was essential to direct CNTs to mitochondria.

Concerns regarding CNT-based drug DS

Interaction of CNTs with biological species has not been comprehensively elucidated, as it has been observed to be a threat to the environment as well as human health. The interest of developing CNTs for biomedical applications means it is imperative to investigate and scrutinize the impact of CNTs especially on human. Over the past decades, numerous studies have been published on the interactions between CNTs with cells in vitro in terms of uptake, intracellular distribution, potential expulsion and even destruction/metabolism (3235). However, the large number of dissimilarities existing between CNT batches employed in different research groups impose challenges in comparisons and the drawing of conclusions of the findings.

The nature of the functional molecules on CNTs surface also plays a determining role in the resulting cytotoxicity of CNTs construct. Several other independent groups also reported that CNTs coated by biological molecules, such as DNA, amphiphilic helical peptides, and serum proteins, were non-toxic to cells (36). Nevertheless, widespread concerns persist on the issue that CNTs are particularly strong in structure. In addition, it is generally recognized that large and pristine CNTs, which are poorly suspendable in aqueous solution, may form bundles and aggregates that induce inflammation and granuloma formation (37). On the other hand, such toxicities are not commonly observed with smaller and individualized CNTs (38). Furthermore, functionalization of CNTs with well-established biocompatible moieties, such as PEG, has conferred reduced in vivo toxicity after being i.v. injected into animals compared to the pristine counterparts (39). Nevertheless, issues of the effects of CNTs on reproductive functions and immune responses have only been partially addressed (40,41). Thus, it is essential to systematically investigate the impact of CNTs to living systems in all aspects before advancing CNTs into the clinical stage as a drug carrier.

Conclusions

It can therefore be determined that use of mitochondria targeting against cancer is promising. However, many improvements are required for proper and more efficient clinical use.

References

1 

Xiong H, Du S, Ni J, Zhou J and Yao J: Mitochondria and nuclei dual-targeted heterogeneous hydroxyapatite nanoparticles for enhancing therapeutic efficacy of doxorubicin. Biomaterials. 94:70–83. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Chen H and Chan DC: Mitochondrial dynamics - fusion, fission, movement, and mitophagy - in neurodegenerative diseases. Hum Mol Genet. 18(R2): R169–R176. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Schapira AH: Mitochondrial diseases. Lancet. 379:1825–1834. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Fulda S, Galluzzi L and Kroemer G: Targeting mitochondria for cancer therapy. Nat Rev Drug Discov. 9:447–464. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Wallace DC: Mitochondria and cancer. Nat Rev Cancer. 12:685–698. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Warburg O, Wind F and Negelein E: The metabolism of tumors in the body. J Gen Physiol. 8:519–530. 1927. View Article : Google Scholar : PubMed/NCBI

7 

Warburg O: On the origin of cancer cells. Science. 123:309–314. 1956. View Article : Google Scholar : PubMed/NCBI

8 

Galluzzi L, Larochette N, Zamzami N and Kroemer G: Mitochondria as therapeutic targets for cancer chemotherapy. Oncogene. 25:4812–4830. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Mathew R, Karantza-Wadsworth V and White E: Role of autophagy in cancer. Nat Rev Cancer. 7:961–967. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Yousif LF, Stewart KM and Kelley SO: Targeting mitochondria with organelle-specific compounds: strategies and applications. ChemBioChem. 10:1939–1950. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Liberman EA, Topaly VP, Tsofina LM, Jasaitis AA and Skulachev VP: Mechanism of coupling of oxidative phosphorylation and the membrane potential of mitochondria. Nature. 222:1076–1078. 1969. View Article : Google Scholar : PubMed/NCBI

12 

Murphy MP and Smith RA: Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu Rev Pharmacol Toxicol. 47:629–656. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Ross MF, Kelso GF, Blaikie FH, James AM, Cochemé HM, Filipovska A, Da Ros T, Hurd TR, Smith RA and Murphy MP: Lipophilic triphenylphosphonium cations as tools in mitochondrial bioenergetics and free radical biology. Biochemistry (Mosc). 70:222–230. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Cortes LA, Castro L, Pesce B, Maya JD, Ferreira J, Castro- Castillo V, Parra E, Jara JA and López-Muñoz R: Novel gallate triphenylphosphonium derivatives with potent antichagasic activity. PLoS One. 10:e01368522015. View Article : Google Scholar : PubMed/NCBI

15 

Gruber J, Fong S, Chen CB, Yoong S, Pastorin G, Schaffer S, Cheah I and Halliwell B: Mitochondria-targeted antioxidants and metabolic modulators as pharmacological interventions to slow ageing. Biotechnol Adv. 31:563–592. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Kang BH, Plescia J, Song HY, Meli M, Colombo G, Beebe K, Scroggins B, Neckers L and Altieri DC: Combinatorial drug design targeting multiple cancer signaling networks controlled by mitochondrial Hsp90. J Clin Invest. 119:454–464. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Kang BH, Siegelin MD, Plescia J, Raskett CM, Garlick DS, Dohi T, Lian JB, Stein GS, Languino LR and Altieri DC: Preclinical characterization of mitochondria-targeted small molecule hsp90 inhibitors, gamitrinibs, in advanced prostate cancer. Clin Cancer Res. 16:4779–4788. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Johnson LV, Walsh ML and Chen LB: Localization of mitochondria in living cells with rhodamine 123. Proc Natl Acad Sci USA. 77:990–994. 1980. View Article : Google Scholar : PubMed/NCBI

19 

Battigelli A, Russier J, Venturelli E, Fabbro C, Petronilli V, Bernardi P, Da Ros T, Prato M and Bianco A: Peptide-based carbon nanotubes for mitochondrial targeting. Nanoscale. 5:9110–9117. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Emanuelsson O, Nielsen H, Brunak S and von Heijne G: Predicting subcellular localization of proteins based on their N-terminal amino acid sequence. J Mol Biol. 300:1005–1016. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Szeto HH: Cell-permeable, mitochondrial-targeted, peptide antioxidants. AAPS J. 8:E277–E283. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Jean SR, Ahmed M, Lei EK, Wisnovsky SP and Kelley SO: Peptide-mediated delivery of chemical probes and therapeutics to mitochondria. Acc Chem Res. 49:1893–1902. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R and Langer R: Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2:751–760. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Iyer AK, Khaled G, Fang J and Maeda H: Exploiting the enhanced permeability and retention effect for tumor targeting. Drug Discov Today. 11:812–818. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Pastorin G: Crucial functionalizations of carbon nanotubes for improved drug delivery: a valuable option? Pharm Res. 26:746–769. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Marrache S and Dhar S: Engineering of blended nanoparticle platform for delivery of mitochondria-acting therapeutics. Proc Natl Acad Sci USA. 109:16288–16293. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Kolishetti N, Dhar S, Valencia PM, Lin LQ, Karnik R, Lippard SJ, Langer R and Farokhzad OC: Engineering of self-assembled nanoparticle platform for precisely controlled combination drug therapy. Proc Natl Acad Sci USA. 107:17939–17944. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Yamada Y and Harashima H: Mitochondrial drug delivery systems for macromolecule and their therapeutic application to mitochondrial diseases. Adv Drug Deliv Rev. 60:1439–1462. 2008. View Article : Google Scholar : PubMed/NCBI

29 

DSouza GG, Rammohan R, Cheng SM, Torchilin VP and Weissig V: DQAsome-mediated delivery of plasmid DNA toward mitochondria in living cells. J Control Release. 92:189–197. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Zhou F, Xing D, Wu B, Wu S, Ou Z and Chen WR: New insights of transmembranal mechanism and subcellular localization of noncovalently modified single-walled carbon nanotubes. Nano Lett. 10:1677–1681. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Moon HK, Lee SH and Choi HC: In vivo near-infrared mediated tumor destruction by photothermal effect of carbon nanotubes. ACS Nano. 3:3707–3713. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Ali-Boucetta H, Al-Jamal KT, Müller KH, Li S, Porter AE, Eddaoudi A, Prato M, Bianco A and Kostarelos K: Cellular uptake and cytotoxic impact of chemically functionalized and polymer-coated carbon nanotubes. Small. 7:3230–3238. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Mao H, Kawazoe N and Chen G: Uptake and intracellular distribution of collagen-functionalized single-walled carbon nanotubes. Biomaterials. 34:2472–2479. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Holt BD, Dahl KN and Islam MF: Cells take up and recover from protein-stabilized single-wall carbon nanotubes with two distinct rates. ACS Nano. 6:3481–3490. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Kagan VE, Konduru NV, Feng W, Allen BL, Conroy J, Volkov Y, Vlasova II, Belikova NA, Yanamala N, Kapralov A, et al: Carbon nanotubes degraded by neutrophil myeloperoxidase induce less pulmonary inflammation. Nat Nanotechnol. 5:354–359. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Chin SF, Baughman RH, Dalton AB, Dieckmann GR, Draper RK, Mikoryak C, Musselman IH, Poenitzsch VZ, Xie H and Pantano P: Amphiphilic helical peptide enhances the uptake of single-walled carbon nanotubes by living cells. Exp Biol Med (Maywood). 232:1236–1244. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Ali-Boucetta H, Nunes A, Sainz R, Herrero MA, Tian B, Prato M, Bianco A and Kostarelos K: Asbestos-like pathogenicity of long carbon nanotubes alleviated by chemical functionalization. Angew Chem Int Ed Engl. 52:2274–2278. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Wick P, Manser P, Limbach LK, Dettlaff-Weglikowska U, Krumeich F, Roth S, Stark WJ and Bruinink A: The degree and kind of agglomeration affect carbon nanotube cytotoxicity. Toxicol Lett. 168:121–131. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Yang ST, Wang X, Jia G, Gu Y, Wang T, Nie H, Ge C, Wang H and Liu Y: Long-term accumulation and low toxicity of single-walled carbon nanotubes in intravenously exposed mice. Toxicol Lett. 181:182–189. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Campagnolo L, Massimiani M, Palmieri G, Bernardini R, Sacchetti C, Bergamaschi A, Vecchione L, Magrini A, Bottini M and Pietroiusti A: Biodistribution and toxicity of pegylated single wall carbon nanotubes in pregnant mice. Part Fibre Toxicol. 10:212013. View Article : Google Scholar : PubMed/NCBI

41 

Salvador-Morales C, Flahaut E, Sim E, Sloan J, Green ML and Sim RB: Complement activation and protein adsorption by carbon nanotubes. Mol Immunol. 43:193–201. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 12 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang XY and Zhang XY: Mitochondria targeting nano agents in cancer therapeutics (Review) Retraction in /10.3892/ol.2020.12101. Oncol Lett 12: 4887-4890, 2016
APA
Zhang, X., & Zhang, X. (2016). Mitochondria targeting nano agents in cancer therapeutics (Review) Retraction in /10.3892/ol.2020.12101. Oncology Letters, 12, 4887-4890. https://doi.org/10.3892/ol.2016.5302
MLA
Zhang, X., Zhang, P."Mitochondria targeting nano agents in cancer therapeutics (Review) Retraction in /10.3892/ol.2020.12101". Oncology Letters 12.6 (2016): 4887-4890.
Chicago
Zhang, X., Zhang, P."Mitochondria targeting nano agents in cancer therapeutics (Review) Retraction in /10.3892/ol.2020.12101". Oncology Letters 12, no. 6 (2016): 4887-4890. https://doi.org/10.3892/ol.2016.5302