Back to Journals » Clinical Pharmacology: Advances and Applications » Volume 17
Triphenylphosphine-Based Mitochondrial Targeting Nanocarriers: Advancing Cancer Therapy
Authors Ali MS, Gowda BHJ, Shukla R , Kesharwani P
Received 7 March 2025
Accepted for publication 29 May 2025
Published 10 June 2025 Volume 2025:17 Pages 119—141
DOI https://doi.org/10.2147/CPAA.S526895
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 4
Editor who approved publication: Professor Arthur E. Frankel
Mohd Shoab Ali,1 BH Jaswanth Gowda,2 Rahul Shukla,3 Prashant Kesharwani1
1Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; 2Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, KA, 575018, India; 3Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER, Raebareli), Raebareli, 226301, India
Correspondence: Prashant Kesharwani, Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India, Tel/Fax +91 7999710141, Email [email protected]
Abstract: Numerous chemotherapeutic drugs are commercially available for cancer treatment; however, their efficacy is often compromised by diminishing therapeutic effectiveness and unpredictable adverse effects. The lack of specific targeting limits their optimal therapeutic potential. Mitochondria are the primary sites of cellular energy production and play a critical role in cell survival and death. Furthermore, numerous studies have found an apparent association between mitochondrial metabolism and carcinogenesis and progression. Therefore, significant attention has been directed toward nanocarriers specifically designed for mitochondrial delivery, aiming to enhance the precision of chemotherapeutic agent transport to these critical organelles. Among these, triphenylphosphonium has emerged as a prominent mitochondrial targeting agent due to its superior targeting capabilities. This approach not only reduces the required drug dosage but also minimizes adverse effects on healthy tissues. This review provides a concise analysis of nanotechnology’s contributions to cancer therapy, emphasizing its potential for targeting at both cellular and sub-cellular levels. Additionally, it delves into mitochondrial targeting, with a particular focus on nanocarriers engineered for efficient mitochondrial drug delivery. Moreover, it focuses on strategies employed by researchers to introduce TPP in nanocarrier systems for mitochondrial delivery and concludes by addressing challenges associated with TPP including hemolytic activity and how researchers mitigate this issue.
Keywords: nanomedicine, cancer, mitochondrial targeting, triphenylphosphine, nanoparticles
Graphical Abstract:
Introduction
Cancer stands as the primary contributor to mortality, causing the loss of over six million lives annually.1,2 The significant percentages of cancer in men exist in the urinary bladder, rectum, colon, bronchus, lung, prostate, and blood whereas the highest cancer prevalence in women occurs in the breast, thyroid, uterine corpus, rectum, colon, bronchus, lung, and blood.3,4 In general, breast and prostate cancer constitute a major portion of women and men.5 Despite substantial investments in personnel and resources to investigate novel cancer treatments, existing clinical procedures have encountered restricted success due to the intricacy, variety, and heterogeneity inherent in the field of oncology.6 The primary challenge arises from the lack of specificity in chemotherapeutic drugs, a factor that frequently limits their clinical effectiveness and gives rise to severe side effects.2,7
Recent progress in the field of nanocarriers enables their passive targeting and accumulation in tumor tissues via the enhanced permeability and retention (EPR) effect.2,8–10 However, studies have shown that a very low percentage of introduced nanocarriers ultimately gather in solid tumor locations, primarily due to various complexities such as nonspecific adherence, off-target effects, and premature release.11,12 Thus, the development of organelle-specific targeting nanocarriers has surfaced as a current focal point within contemporary targeted drug delivery research.13–15
The majority of eukaryotic cells possess mitochondria, crucial cellular structures that control programmed cell death and function as a vital energy reservoir for cellular metabolism.16 Mitochondria initiate the cellular apoptosis pathway through diverse mechanisms, such as the release of cytochrome C, the translocation of apoptotic proteins, and the reduction of mitochondrial membrane potential (MMP).17 Alterations in mitochondrial structure not only disrupt the development, metabolism, and proliferation of tumors but also serve as the catalyst for inducing apoptosis in tumor cells. Nevertheless, the direct targeting of mitochondria using traditional nanocarriers proves to be difficult, majorly due to the complex structure of mitochondria and various other barriers in the tumor environment.18,19 The frequently employed targeting moieties for mitochondria encompass cationic peptides, triphenylphosphonium (TPP), pyridinium, dequalinium (DQA), and so on.20 However, several studies have unveiled the potential of TPP-conjugated nanocarriers as a robust mitochondria-targeted system for cancer treatment (Figure 1).21,22
![]() |
Figure 1 Mitochondria-targeted cancer therapies enabled by nanoplatforms including chemotherapy, photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), sonodynamic therapy (SDT), radiodynamic therapy (RDT), and combined immunotherapy, reproduced from Gao Y, Tong H, Li J, et al. Mitochondria-targeted nanomedicine for enhanced efficacy of cancer therapy. Front Bioeng Biotechnol. 2021;9:720508. Copyright © 2021 Gao, Tong, Li, Li, Huang, Shi and Xia.,23 licensed under CC BY 4.0. |
TPP can easily accumulate in mitochondria after successfully passing through the phospholipid bilayer.22,24 Alkylated TPP cations were initially utilized to investigate the process of linking the oxidative phosphorylation (OXPHOS) with MMP and subsequently, they were used to measure the MMP and it was also found that pharmacophores, antioxidants, and probes were also transported to mitochondria by TPP cations.22 In this context, the present review briefly discusses the role of nanotechnology, the importance of targeted delivery, and the scope of cellular and sub-cellular targeting in cancer treatment. Furthermore, it meticulously delves into the technical aspects of mitochondrial targeting and TPP-conjugated mitochondrial targeting nanocarriers in cancer therapy.
Brief Note on Role of Nanotechnology in Cancer Therapy
Nanoparticles (NPs) have gained popularity in recent years because of their potential to solve issues with chemotherapy medications like stability, solubility, immunogenicity, blood circulation half-time, and diffusivity as well as to improve the targeted delivery of drugs to cancer cells during cancer treatment.25–30 Additionally, NPs offer many benefits for diagnosis and treatment, including easy surface functionalization, a large surface area for carrying therapeutic and imaging payloads, the ability to solubilize hydrophobic drugs, drugs can be protected from biodegradation, and control over size and shape.31 NPs can be developed into a variety of sha pes, including one-dimensional (1D) structures like nanowires and nanotubes, two-dimensional (2D) structures like single-layered graphene and other molecular sheets, three-dimensional (3D) structures like liposomes, micelles, and hydrogels, and zero-dimensional (0D) structures like quantum dots.32 Nanomedicine is fueled by the synergy between NPs and their special adjustable qualities (shape, optical properties, size, surface properties, etc) in contrast to traditional pharmaceutical ingredients.33 Cancers exhibit irregular cell proliferation, which is facilitated through the newly formed blood vessel networks with large gaps between the endothelial cells, making them highly permeable.34,35 NPs are engineered to circulate in the bloodstream for extended periods and to accumulate in tumors, either through the EPR effect or as a result of the affinity between ligands attached to NPs and tumor receptors.36 This is because tumor tissues have different anatomical and physiological features than healthy tissues.37 Among the most crucial and promising elements of cancer treatment is drug distribution. Controlled, prolonged, and predictable release of medicines or biomolecules, external/internal stimulus-responsive site-targeted distribution, and the detection of certain biomolecules that can offer therapeutic feedback are some of the ways that NPs are used in drug delivery systems. Optimal cellular and subcellular delivery can be achieved by accurately controlling drug delivery platforms through changes in their size, shape, surface chemistry, and chemical composition. Doxorubicin-loaded liposomes were approved 25 years ago to treat Kaposi’s sarcoma, and they have also been approved recently to treat ovarian and breast cancer.38 The FDA has so far approved over 20 distinct NPs for use in therapeutic settings.39 The therapeutic advantages of nanotechnology for the focused administration of anticancer agents, nanomedicine may help with unresolved clinical issues, lessen adverse effects, and increase a drug’s bioavailability. Some of the NPs include liposomes, dendrimers, nanostructured lipid carriers, polymer NPs, micelles, gold NPs, mesoporous silica NPs, metal-organic frameworks, and so on.
Among cellular delivery, subcellular delivery gained the attention of researchers all over the world to increase the therapeutic activity of active moiety.40 Mitochondria are essential to the production of energy which leads to the survival of eukaryotic cells. Mitochondria are essential modulators of the intrinsic apoptotic process by controlling the movement of pro-apoptotic proteins from the mitochondrial intermembrane space to the cytosol. Moreover, mitochondria are involved in several non-apoptotic cell death processes, most notably controlled necrosis, or necroptosis.41,42 As per Otto Warburg, the mitochondria of cancerous cells were dysfunctional,43 and due to its key role in cancerous cell progression, its target serves as a promising approach to cancer treatment. For the successful elimination of cancerous cells, therapeutic agents linked with mitochondrial targeting agents should be effectively delivered to mitochondria44 and for that several approaches were designed by researchers for targeting the mitochondria either by small molecules, bioactive molecules, sequences, nanomaterials or mitochondrial biomarkers.
Importance of Targeted Drug Delivery in Cancer Therapy
The goal of targeted drug delivery is to deliver drugs to their intended site of action in high concentrations, regardless of how they are administered.45–50 Therefore, to improve the effectiveness of advanced chemotherapy, either a drug delivery system that can specifically deliver drugs to their intended site of action is needed, or a new target site should be identified.51–54 Targeting drug delivery is preferred above traditional drug delivery systems for four reasons: unsatisfied pharmacotherapeutic (High dose, Adverse effects, Low patient compliance), pharmaceutical (Low solubility, Low stability), pharmacokinetic (Low Half-life, High Volume of Distribution), and pharmacodynamics (Low specificity, Low therapeutic index) performance.55–59 Optimizing drug delivery strategies to target specific areas not only improves therapeutic efficacy but also reduces toxicity associated with large doses and a small therapeutic index.12,55,60–64 Targeting is necessary to address the limits and inherent shortcomings of traditional drug delivery systems.65–70 Furthermore, targeted drug delivery offers promising benefits such as simplified drug-administration processes, reduced drug amount which lowers therapeutic costs, and the ability to quickly boost drug concentration in target compartments without affecting non-target compartments.71–75 In general, targeting was achieved in two ways, active targeting uses specific ligands that bind to specific receptors on cancer cells to deliver drugs more effectively.76–78 The surface of nanocarriers can be made to specifically target cancer cells by attaching targeting ligands to them.79 Chemotherapeutic drugs must be actively targeted by utilizing molecular ligands to increase their cellular uptake and therapeutic efficacy.80,81 The EPR effect is caused by the leaky nature of tumor blood vessels and the poor drainage of fluids from tumors, which leads to the formation of new blood vessels in a rapid and uncontrolled manner.58,82 So, passive targeting utilized this feature of the tumor microenvironment via nanoscale drug carriers to target tumor cells.83 It was also observed that free drug diffuses non-specifically but due to the unique size of nanocarriers, they can penetrate the cancer cell mass with ease through the leaky vasculature via the EPR effect.84–86
Scope of Cellular and Sub-Cellular Targeting in Cancer Therapy
During the pathological transition of a cell from normal to diseased, several changes occur at the macromolecular and morphological levels. These changes include modifications to the cell’s shape and enzymatic profile, alterations to the immediate microenvironment (such as changes in redox properties and pH), and the expression of new molecules at various cellular locations.87,88 Overexpressed markers often require specialized or unique methods for active targeting.64,89,90 The majority of active cellular targeting strategies rely on interactions between ligands and receptors, enzymes and substrates, or antibodies and antigens.91–93 Some cancer cells exhibit an excess of specific surface markers, including somatostatin receptors (SSTRs), vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor 2 (HER2), folate receptor protein, myeloid antigen (CD13), cell adhesion glycoprotein (CD44), programmed death ligand-1 (CD274), myeloid antigen (CD13), and ανβ3 integrin.94,95 The utilization of specific molecules that bind to overexpressed cellular markers has facilitated more effective drug targeting of cancer cells.73,96,97 Cellular targeting is effective at killing cells by delivering the drug solely to the interior of the cell. In contrast, subcellular targeting directs drugs to be released at the subcellular location or targeted organelle (eg, mitochondria, nucleus, endoplasmic reticulum, lysosome, Golgi body).98,99 This has spurred increased research efforts to develop more sophisticated nano-drug designs capable of directing internalized nano-drug complexes to their intracellular target sites.
Mitochondrial Targeting in Cancer Therapy
The energy-producing organelles known as mitochondria are essential for maintaining metabolic and cellular life functions.100–102 They play a key role in vital physiological functions such as calcium ion control, ATP synthesis, electron transport, reactive oxygen species (ROS) formation, and the start of cell death.103–105 The ATP synthase enzyme is responsible for producing ATP on the inner membrane of mitochondria. Complexes I, III, and IV pump protons from the mitochondrial matrix into the intermembrane gap during the synthesis of ATP.106 The usual mitochondrial membrane potential (about 140 mV) is maintained by pumping out protons from the inner membrane using electrons supplied from reduced coenzymes via the ETC. Figure 2 demonstrates how complexes I, III, and IV are involved in ROS production and calcium ion transport, which in turn opens the mitochondrial transition pores and causes cell death. Normal cells depend on mitochondria for a variety of vital functions, such as energy provision, oxidative phosphorylation, ATP synthesis, reactive oxygen species (ROS) generation, cell cycle regulation, cell differentiation, and programmed cell death. Moreover, mitochondria play a key role in a variety of metabolic processes that affect cell fate, such as the citric acid cycle, gluconeogenesis, steroid production, and beta-oxidation of fatty acids.107–109 Traditional methods that target the mitochondria of cancer cells typically try to harm or impair the function of the mitochondria.110 The growth and survival of cancer cells are significantly influenced by mitochondria, which makes them a desirable target for cancer treatment.111,112 Numerous investigations have demonstrated that the development of cancer cells is significantly influenced by mitochondrial building blocks.111 ATP, ROS, and metabolites for the synthesis of macromolecules are all primarily produced by mitochondria.112 Targeting mitochondria has therefore been shown to be a successful cancer treatment tactic. Numerous authorized medications that provide therapeutic benefits by triggering mechanisms of apoptosis by directly affecting mitochondria emphasize the significance of mitochondria in pharmacology as targets.113,114
![]() |
Figure 2 Schematic illustrating mitochondria as a primary driver and therapeutic target of cancer. The figure depicts the chaotic development of cell death events. The respiratory activities of complexes I, III, and IV cause proton translocation across the inner mitochondrial membrane, resulting in the production of ATP from ADP and Pi. Quinone oxidoreductase reduces coenzyme quinone (Q) after oxidizing the mitochondrial electron transport chain. Reduced Q produces cytochrome C (by complex III), which catalyzes the reduction of molecular oxygen to water (via complex IV). The formation of reactive oxygen species (ROS) by complex I, together with the accumulation of calcium ions, increases the opening of the mitochondrial permeability transition pore (MPTP), leading to programmed cell death, reproduced from Tabish TA, Hamblin MR. Mitochondria-targeted nanoparticles (mitoNANO): an emerging therapeutic shortcut for cancer. Biomater Biosyst. 2021;3:100023.115 copyright 2021, Elsevier. |
Additionally, it has been observed that the mitochondria in cancerous cells exhibit structural and functional differences compared to those in normal cells (Figure 3).116,117 The transmembrane potential (MTP), also known as the MMP, is established across the inner mitochondrial membrane through the negative charges carried by the mitochondrial membrane.118 The variance in membrane potential between mitochondria and the plasma membrane is the basis for achieving mitochondrial targeting. Generally, the plasma membrane potential is 3- to 5-fold lower than the MMP.22,119 Furthermore, it has been noted in several cases that the MMP of cancerous cells is higher than that of healthy cells.120 This biological distinction has served as a basis for the development of mitochondrial targeting compounds, allowing drugs to be preferentially concentrated within the mitochondria of cancerous cells. The capacity to generate highly reactive oxygen species (ROS) and adenosine triphosphate (ATP) through aerobic glycolysis is significantly greater in the mitochondria of cancerous cells than in normal cell mitochondria. This distinction provides an alternative approach for targeting cancer cell mitochondria over normal cell mitochondria.121 By increasing the mitochondrial accumulation of anticancer drugs, drug resistance may be addressed, and therapeutic effectiveness could be enhanced.122 There are two well-known methods for delivering drugs to the mitochondria: the targeting ligand is directly conjugated to drugs, or the targeting ligand is attached to nanocarriers.123 Small compounds that target the mitochondria include rhodamine 123, rhodamine 19, TPP, guanidine salt, dequalinium, tetrachlorotetraethyl benzimidazole carbocyanine iodine, and N-methylpyridineiodide Tetrachloro-1,1′,3,3′-tetraethyl-imidacarbocyanine (5,5′,6,6′) compounds (JC-1).124–126 These compounds can target mitochondria due to their lipid solubility, enabling them to enter both the cell and the mitochondria. The positive charge of these particles allows them to be drawn into the mitochondria under the influence of MMP.127
![]() |
Figure 3 Important distinguishing features of mitochondria of cancer cells from mitochondria of normal cells, reproduced from Mani S, Swargiary G, Tyagi S, et al. Nanotherapeutic approaches to target mitochondria in cancer. Life Sci. 2021;281:119773.,128 copyright 2021, Elsevier. |
Mitochondrial Targeting Agents
Mitochondria targeting agents are chemicals under the special category that have a positive charge and interact with mitochondria. They can be small peptides, molecules, or metal complexes that can accumulate in mitochondria without the need for additional targeting systems. High transmembrane potential and protein import mechanism of mitochondria are the primary reasons through which mitochondria targeting agents target the mitochondria.129–132 Some of the majorly used mitochondrial targeting agents are discussed below.
Dequalinium (DQA)
DQA, or dequalinium, is a lipophilic substance first reported by Weiss et al in 1987. It is made up of two cationic quinoline groups joined by an alkyl chain of ten carbons. It is a cationic, delocalized lipophilic chemical that targets mitochondria. According to Weiss et al, this substance prevented several cancer cell lines from proliferating in vitro and in vivo. According to studies, DQA can cause the generation of ROS by preventing ATP synthesis, which in turn causes cytochrome c expression and a decrease in mitochondrial membrane potential. Ultimately, this mechanism triggers the endogenous apoptosis pathway that is dependent on caspase-3 and 9.133 As a result, DQA’s cytotoxicity restricts its use in numerous biological investigations, including cell biology research and fluorescent probes. But because of its cytotoxicity, DQA is also a great drug delivery ligand for anti-cancer treatment. To transport Dox to mitochondria specifically, studies have coupled DQA chloride with Dox (DQA-Dox). The drug compound DQA-Dox demonstrated substantial cytotoxicity to cancer cells and was mostly observed to accumulate in the mitochondria of MCF-7/ADR cells. Despite this, the low endosome DQA capacity for escape and transfection efficiency restrict its application in delivery that targets the mitochondria.134
Pyridinium
Another type of often used, positively charged, lipophilic mitochondrial targeting group is pyridinium salts. Pyridine salts are frequently modified with ethylenic bonds to create big conjugated systems, in contrast to TPP, which is modified with long alkyl chain links. For the optical detection of mitochondria, the conjugated systems and electron-withdrawing properties of pyridine salt may produce or control molecular luminescence. However, pyridine salts’ lipophilicity prevents them from entering tissues in vivo. Cheng’s team created a human serum albumin-facilitated pyridine salt complex to address this problem. This complex was able to target mitochondria in tumor tissue and cause tumor cell death, leading to a breakthrough in the in vivo use of pyridinium salt.135
Mitochondria Targeting Cationic Peptides
Another method for making mitochondrial targeting easier is a peptide. Peptide-based delivery scaffolds have appealing properties such as high absorption in cells and in vivo, tunability, biocompatibility, and ease of production. Peptide advantages make them extremely versatile for transporting a wide range of chemicals. Peptides that target mitochondria are typically lipophilic and cationic charged, enabling them to interact positively with hydrophobic mitochondrial membranes and take advantage of the negative membrane potential of mitochondria. Peptides did not induce mitochondrial depolarization at millimole concentrations like TPP and pyridinium salt. Additionally, due to their biocompatibility, peptides were better suited for treating illnesses and delivering medications to living organisms.136
Mitochondria Penetrating Peptides
Peptides based on cell penetrating peptide (CPP) that effectively penetrate mitochondrial double membranes are known as mitochondrial penetrating peptides (MPP). The highly negative charge of mitochondrial membranes makes it simple for positively charged peptides to enter mitochondria. However, in contrast to CPP, MPP interacts more with inner mitochondrial membrane (IMM) than plasma membrane because of more hydrophobicity exhibited by IMM. Horton et al created mitochondrial penetrating peptide (MPP) with high cellular uptake and mitochondrial matrix targeting capacity for the first time by striking a balance between cationic amino acids (arginine and lysine) and hydrophobic amino acids (cyclohexylalanine).137 It was shown that MPP might alter the intracellular distribution of medications, causing them to mostly collect in mitochondria rather than the nucleus, when covalently conjugated with anticancer medications such as cisplatin or DOX. Additionally, research using fluorescence quenching and PCR-based assays verified that MPP transport medications to the mitochondrial matrix to harm mtDNA.138–140 Moreover, the mitochondrial accumulation was 11.6 times greater in the connected MPP to HPMA copolymer-DOX conjugation.141 Overall, the research suggested that MPP may be used to preferentially deliver nanosystems and chemotherapeutics to mitochondria.
Triphenylphosphine
The history of triphenylphosphine targeted medication delivery dates back to 1995 when Liberman et al used alkylated TPP cations as mitochondrial probes to evaluate oxidative stress in mitochondria. Following that, Smith’s team fabricated Mito-vite E in 1999, an antioxidant that efficiently protected mitochondria from oxidative stress. Other noteworthy advances included MitoQ10 (combines coenzyme Q10 with TPP) which demonstrated selective oxidative stress protection.142–145 In 2002, Fantin et al developed F16-TPP to target mitochondria and fluoresced after aggregating in mitochondria and killing multiple-tumor cells.
Therefore, the discovery of TPP is considered one of the most successful examples of using lipophilic cations to target mitochondria.24 TPP, a delocalized lipophilic cation, is typically employed for mitochondrial targeting because it accumulates on the negatively charged mitochondrial membrane146,147 (Figure 4). The three lipophilic phenyl groups surround the positively charged phosphorus atom that makes up TPP, enhancing its combination.148 This structure also allows for the addition of numerous functional molecules and the formation of a positive charge that is delocalized, enabling it to pass through the non-polar membrane of the mitochondria.149 TPP increases the accumulation of drugs in the mitochondrial matrix by 100–500 times by reducing the activation energy during the MMP process. Additionally, TPP leads to the generation of harmful ROS and the release of apoptosis activators, ultimately resulting in mitochondria-mediated apoptotic cell death.150–152 Drugs modified with lipophilic cations, such as TPP, can effectively accumulate in mitochondria because they can penetrate the phospholipid membrane of mitochondria due to their lipophilicity. This lipophilicity is attributed to the phenyl group and the delocalization of positive charge on the phosphorus atom.153 Consequently, the mitochondrial targeting vector TPP group is frequently incorporated into anticancer medications to reduce their toxicity.22,154 Many bioactive molecules have been conjugated with TPP to facilitate their mitochondrial targeting. For instance, when doxorubicin (DOX) was chemically linked to TPP, it demonstrated a strong ability to target mitochondria. This led to the induction of apoptosis, or programmed cell death, in tumor cells through the mitochondrial pathway. The combination proved to be more effective in damaging mitochondria compared to using DOX alone.149 Combining TPP with nanoformulations can also enhance mitochondrial targeting. For instance, liposomes loaded with paclitaxel (PTX) and modified with the lipophilic cation TPP can effectively target mitochondria in cancer cells155 (Table 1). Apart from the advantages, the main disadvantage associated with TPP due to its strong positive is hemolysis. This problem was overcome by the researchers after utilizing strategies like stimuli-responsive nanocarriers which expose TPP only at the desired location. For example, TPP-DOX was specifically released only in the tumor microenvironment via pH-responsive BSA-based nanocarrier due to which premature interaction of TPP with normal cells was inhibited. Similarly, chitosan nanoparticle coated by HA enables its active uptake through CD44 receptors and this coat masks the positive charge of TPP and is released in acidic tumor microenvironment.156 Moreover, another approach was done by Zhang et al in which they embedded TPP in the lipidic bilayer of PEGylated liposomes which gives stealth behavior in the bloodstream and shows mitochondrial targeting after internalization. These strategies overcome the limitations associated with positive TPP charge which ensures safety with significant therapeutic efficacy.
![]() |
Figure 4 TPP conjugated substances are uptaken by cells via plasma and mitochondria membrane potentials. Created in BioRender. Gowda b.h., J. (2025) https://BioRender.com/ qxll1ii. |
![]() |
Table 1 Investigations on TPP-Based Mitochondrial Targeting Nanocarriers for Cancer Therapy |
Based on literature, it can be inferred that TPP is superior over DQA, pyridinium, and cationic peptides in terms of mitochondrial targeting. Despite the luminescent property of pyridinium salts, it suffers from low tumor tissue penetration due to its excessive lipophilicity. While DQA has a limitation like poor transfection efficiency and low endosomal escape. However, TPP offers significant accumulation in mitochondria due to its optimal ratio of delocalized positive charge and lipophilicity. Moreover, enzymatic degradation is the major concern associated with cationic peptides in the biological environment unlike TPP that is chemically stable, and deeply penetrates mitochondria across various cells. These properties make TPP an efficient and consistent approach for focused mitochondrial delivery.
Evaluation Methods of TPP Targeting
Here, we discussed the methods used to evaluate the mitochondrial targeting ability by TPP which is further elaborated in the next section. One of the widely used techniques is confocal laser scanning microscopy in which mitochondria specialized dyes like MitoTracker Red or Green were used which allows the researchers to visualize the accumulation of TPP-functionalized cargo or nanoparticle within subcellular organelle mitochondria. Another technique is flow cytometry which can quantitatively assess the degree of cellular and subcellular uptake of therapeutic agent or nanocarrier via fluorescence intensity. For assessment of mitochondrial uptake, researchers first performed subcellular fractionation via High-Performance Liquid Chromatography to isolate mitochondria and then analyzed the accumulation of therapeutic cargo or nanoparticle within it. Another approach is the decrement of mitochondrial membrane potential that leads to the disruption of mitochondria which implies effective mitochondrial targeting and for that confirmation stainings like Rhodamine 123 or JC-1 are employed by the researchers. For example, after colocalized MitoTracker with TD@MB, mitochondrial delivery of DOX was validated,164 and in another research, fluorescence intensity indicates the substantial uptake of PTX-TPP loaded HSA-Apt nanoparticle in mitochondria.165
Triphenylphosphine-Based Mitochondrial Targeting Nanocarriers in Cancer Therapy
TPP can be introduced into the nanoparticles either by physically or chemically on the surface of nanoparticle or it can be conjugated chemically with the therapeutic agent and then load into the nanoparticle (Table 2). This review comprehensively explores both the approaches.
![]() |
Table 2 Investigation the Physicochemical Properties and TPP Incorporation Strategies of Nanoparticles |
Nanoparticle Surface Modification with TPP
Polymer dots (PDs) used as nanocarriers have several advantages, such as biocompatibility and high hydrophilicity. Therefore, they can be employed to address the issue of drug hydrophobicity by increasing the solubility of drugs once they are loaded into PDs.169,170 Due to the varying levels of ROS and glutathione (GSH), diselenide bonds can selectively target cancer cells and release chemotherapeutic drugs only in a cancerous environment. This prevents the release of chemotherapeutic drugs in normal cells, thereby reducing the side effects of cancer treatment.171 By utilizing diselenide bonds, researchers have synthesized redox-responsive PDs, allowing for controlled drug release in response to redox stimuli at targeted sites.172,173 In this lane, Kim and teammates formulated ROS and GSH-responsive PDs as a nanocarrier with TPP for mitochondrial targeting to control the release of PTX (PDs-TPP-PTX) in malignant cells as demonstrated in (Figure 5). MTT assay was done by using HeLa and PC-3 (cancerous cells) and CHO-K1 (normal cells) and the results revealed that on increasing the concentrations of PDs-TPP all three cell lines showed high cell viability which confirms that PDs-TPP is not able to kill cancer cells. When the concentration of (PDs-TPP-PTX) was increased, cell viability in HeLa and PC-3 cells decreased significantly. However, cell viability in CHO-K1 cells remained relatively high even at high concentrations of (PDs-TPP-PTX). These findings demonstrated that this approach was capable of controlling the release of PTX in cancer cells while minimizing undesired release in normal cells. A cellular uptake study showed that both HeLa and PC-3 cells exhibited bright fluorescence after 3 h of PDs-TPP uptake, which became even brighter after 6 h. In contrast, CHO-K1 cells showed low fluorescence intensity even after 6 h of uptake. It was concluded that PDs-TPP occurred more frequently in cancer cells because of the cleavage of diselenide bonds in higher levels of ROS and GSH.157
![]() |
Figure 5 PDs-TPP (PTX) synthesis, the mechanism of selective PTX release, and mitochondrial targeting in cancer cells are shown schematically. Image reproduced from Kim SG, Robby AI, Lee BC, Lee G, Park SY. Mitochondria-targeted ROS- and GSH-responsive diselenide-crosslinked polymer dots for programmable paclitaxel release. J Ind Eng Chem. 2021;99:98–106. Copyright (2021), with permission from Elsevier.157 |
A recent study conducted by Gong et al developed a nanoscale drug delivery system (DDS) targeting mitochondria and responsive to GSH stimuli. The DDS is based on a compound called TPP, which incorporates a disulfide linker (“S-S”), and fluorescent carbon nanodots (CDs). The DDS can deliver the anticancer medication camptothecin (CPTN) to mitochondria. After evaluating cytotoxicity studies on HeLa, HepG2, A549, MCF-7, and 4T1 cells it was found that TPP-CDs-S-CPTN possessed remarkable in vitro antineoplastic activity. On comparing CDs-S-CPTN and TPP-CDs-S-CPTN, the latter possessed higher cytotoxicity which makes TPP-CDs-S-CPTN a good therapeutic nano drug delivery system. This is because TPP-CDs-S-CPTN released a sufficient amount of CPTN by the breakage of disulfide linkage after being administered into cancerous cells. Moreover, prominent cytotoxicity was induced in normal LO2 cells by naked drug CPTN, whereas much lower cytotoxicity was induced in normal LO2 cells with the same drug concentration by TPP-CDs-C-CPTN, TPP-CDs-S-CPTN, and CDs-S-CPTN. This is because TPP-CDs-S-CPTN and CDs-S-CPTN released a sufficient amount of CPTN in cancerous cells rather than healthy cells. After all, cancerous cells have higher levels of intracellular GSH than normal cells and at last, it was also found that cytotoxicity in healthy as well as cancerous cells was not shown by CDs and TPP-CDs, which indicates that CDs are a good drug carrier. To assess the biological toxicity organ indices, spleen coefficient, body weight, and tissue slices were analyzed in mice 28 days after injection of TPP-CDs-S-CPTN, TPP-CDs-C-CPTN, CDs-S-CPTN, TPP-CDs-S and normal saline in the tail by intravenous injection for five times. Observed results indicate that the developed formulation has no potential biotoxicity and is a good candidate for mitochondrial-targeted drug delivery.158
A study conducted by Biswas et al developed a novel approach using polyethylene glycol-phosphatidylethanolamine (PEG-PEA) conjugated with TPP (TPP-PEG-PEA) and subsequently incorporated it into the liposomal lipid bilayer for the delivery of PTX. Mitochondrial targeting was investigated by Rhodamine-PE-tagged liposomes and MTG-stained mitochondria. It was found that TPP-PEG-PEA modified liposomes (TPP-PEG-PEA-L) have significantly more Rhodamine-PE fluorescence colocalization within the mitochondria than plain liposomes. A cytotoxicity study was done on 4T1 cells and it was found (TPP-PEG-PEA-L-PTX) significantly reduced the viability of cell than plain liposomal PTX. Similar results were obtained after evaluating the efficacy of anti-tumor that TPP-PEG-PEA-L-PTX induced a much more inhibitory growth than plain liposomal PTX.155
TPP bromide, hyaluronic acid-d-α-tocopherol succinate-(4-carboxymethyl) (TPP-TS-HUA) NPs designed by Lee et al were employed for the administration of lapatinib (LAP) in the management of triple-negative breast cancer (TNBC). Designed NPs were examined using a mouse model with MDA-MB-231 tumors for determining its tumor targeting capability which demonstrated that Cy5.5-HUA-TS-TPP/LAP NPs exhibited higher fluorescence signal than Cy5.5-HUA-TS/LAP NPs in the image obtained through ex-vivo scanning of excised tumor. The possible reason could be that HUA-TS-TPP/LAP NPs appear to have smaller particle size and unimodal size distribution over HUA-TS/LAP NPs which contribute to the enhancing the accumulation of NPs in the tumor. The results from the cellular uptake study which was done on MDA-MB-231 cells and determined by fluorescence detector Cy5.5 found that the fluorescence intensity on an average of Cy5.5-TPP-TS-HUA/LAP is greater than Cy5.5-TS-HUA/LAP NPs. The possible reason could be the presence of TPP on the exterior facade of TS-HUA/LAP NPs which seems to enhance the efficiency of cellular accumulation of TPP-TS-HUA/LAP NPs over TS-HA/LAP NPs. The effectiveness of LAP NPs in combating tumors was evaluated using an MDA-MB-231 tumor xenograft mouse model. After H&E staining of tissue removed from a tumor, it was found that HUA-TS-TPP/LAP NPs induced a higher degree of apoptosis within a significant portion of the tumor mass compared to HUA-TS/LAP NPs. The possible reason could be a combination of factors, including the passive targeting strategy of the EPR effect, the active targeting approach involving interactions with HUA-CD44 receptors, the influence of TPP and TS on mitochondrial targeting and disruption, and the pharmacological effects of LAP.159
Photodynamic therapy (PDT) uses a photosensitizer (PS) molecule that is activated by near-infrared (NIR) light to generate ROS which ultimately damages mitochondria, leading to cell death.174,175 IR780 is a lipophilic dye that can generate oxygen when exposed to NIR laser light and it also preferentially accumulates in tumors, making it a potential therapeutic agent for cancer. Photothermal therapy (PTT) converts light radiation directly into heat energy that can kill cells, even without the need for oxygen. This makes it a promising new treatment for cancer and other diseases.176,177 In this context, Ruttala et al, after combining PTT and PDT, developed a robust nano-platform system consisting of the TPP moiety for the mitochondrial targeting of cancerous cells. They developed a new type of NP that is made up of a polymer lipid hybrid core, a zinc-copper oxide NP shell, and TPP molecules (TPP-Z-I-PNPs) as demonstrated in (Figure 6). Flow cytometry study showed that TPP-Z-I-PNPs notably uptake by HT-29 and A549 cells but TPP-Z-I-PNPs showed good uptake than Z-I-PNPs. This is because TPP-conjugated NP systems have good interaction with the cell membrane as it is negatively charged. In determining the mitochondrial targeting ability of TPP-loaded nanosystems, it was found that malignant cells’ mitochondria have a high concentration of nanosystems. Phototherapy of TPP-Z-I-PNPs was evaluated after evaluating the photothermal effect of zinc copper oxide NPs and the photodynamic effect of IR780 and they were determined after exposure to NIR laser. The analysis demonstrates that after 5 min of NIR irradiation, the temperature of zinc copper oxide NPs increased from 25° C to 56.8° C, and similarly after a short exposure of NIR IR780 temperature increased to 54.9° C. There is also a temperature that rises rapidly to 61.8° C by TPP-Z-I-PNPs due to the photodynamic and photothermal nature of IR780 and zinc-copper oxide NPs. It was concluded that the death of cells happened solely in the region that was irradiated, whereas cells beyond the laser region continued alive. An HT-29-bearing xenograft model was used to assess anticancer effectiveness in vivo by using ZNP (+NIR), ZNP (-NIR), Z-I-PNPs, TPP-Z-I-PNPs (+NIR), and TPP-Z-I-PNPs (-NIR) and it was found that maximum tumor regression showed by TPP-ZC-PNPs (+NIR) because of the synergistic potential of PTT and PDT.160
![]() |
Figure 6 TPP-conjugated-ZnCuO NPs (ZNP) and IR780-loaded polymer-lipid hybrid NPs (TPP-Z-I-PNPs), which target mitochondria, are shown schematically. Image reproduced from Ruttala HB, Ramasamy T, Ruttala RRT, et al. Mitochondria-targeting multi-metallic ZnCuO nanoparticles and IR780 for efficient photodynamic and photothermal cancer treatments. J Mater Sci Technol. 2021;86:139–150, Copyright (2021), with permission from Elsevier.160 |
TPP-Conjugated Therapeutics
Through conjugation of the PS pyropheophorbide-a (PPa) with the mitochondrial targeting group TPP via a biodegradable ROS-responsive thioketal linkage, Huang et al developed and synthesized a PS (TPP-TKl-PPa). TPP-TKl-PPa can self-assemble into NPs because of its amphiphilic nature. These NPs can then further incorporate GSH-depleting agents (DEM) to create co-administration nanoplatforms for highly efficacious PDT. To investigate the TPP-modified nanomaterials may target mitochondria or not MitoTracker Green and Hoechst 33342 were used to stain mitochondria and cell nuclei. The results indicate that MDA-MB-231 cells treated with TPP-TKl-PPa/DEM NPs and TPP-TKl-PPa NPs showed abundant colocalization yellow fluorescent signals which indicates that preferably targeting mitochondria was achieved by using TPP. After being treated with various samples, the ability of MDA-MB-231 breast cancer cells to produce intracellular ROS was assessed by measuring the DCF fluorescence intensity of the DCFH-DA fluorescent probe. It was found that in MDA-MB-231 cells (control), PPa-treated, and DEM-treated cells negligible green fluorescence was observed which indicates that PPa or DEM alone might not have a therapeutic effect without PDT whereas green fluorescence was observed in TPP-TKl-PPa/DEM NPs and TPP-TKl-PPa NPs which indicates that under the exposure of light irradiation, a considerable amount of ROS was produced. A study on MDA-MB-231 cells treated with various samples both with and without light irradiation was conducted to ascertain cell apoptosis and findings suggest that apoptosis was negligible in the case of without light irradiation whereas for 2 min with light irradiation, the highest (94.4%) apoptosis rate was observed in case of TPP-TKl-PPa/DEM NPs. This is because of the presence of PPa and DEM which exhibited greater phototoxicity due to oxidative stress and PDT working together.161
Another interesting investigation by Battogtokh and teammates used PDT for the development of a mitochondrial-targeted PS. They used pheophorbide-a (PheA) as a porphyrin-derivative PS and carboxybutyl TPP for mitochondrial targeting, both are conjugated via carbodiimide linkage and then further TPP-PheA compound was added to folate-cholesteryl albumin (FA-cho-BSA) NPs to improve biocompatibility. TPP-PheA@FA-cho-BSA NP and TPP-PheA showed stronger fluorescence intensity than free PheA and TPP-PheA@cho-BSA NPs in cellular uptake study and further this red and green fluorescence overlapped, and turned into yellow which indicates that TPP-PheA after passed through the cells, localized on the mitochondria. After performing ROS quantity and mitochondrial membrane disruption, it was found that on Hela cells TPP-PheA@cho-BSA produced 1.5, TPP- PheA@FA-cho-BSA produced 2.7 and TPP-PheA produced 3.6 folds more than that of PheA. On determining mitochondrial membrane disruption, it was found that PheA-treated cells and control cells showed red fluorescence signals but TPP-PheA and TPP-PheA@FA-cho-BSA NP showed green fluorescence. Based on fluorescence, it was concluded that PheA-treated cells and control cells have greater MMP whereas, low MMP was seen in TPP-PheA@FA-cho-BSA NP and TPP-PheA-treated cells. In vitro phototoxicity of TPP-PheA@FA-cho-BSA NP and TPP-PheA was performed against HeLa cells and it was found that both of them are significantly much more than free PheA and PheA@FA-chol-BSA. This is because of the presence of TPP with mitochondrial localization. In vivo imaging and biodistribution study was done with the help of fluorescence microscopy sections of frozen tumor and it was found that low fluorescence intensity from PheA after intravenously administered PheA@FA-cho-BSA whereas red fluorescence signals of PheA obtained from TPP-PheA@FA-chol-BSA which indicates greater intracellular uptake of TPP-PheA. Antitumor efficacy in vivo was determined by calculating the therapeutic effectiveness from the tumor mass in formulation-injected mice and control mice on day 14 and it was found that TPP-PheA@FA-chol-BSA NP showed the highest tumor Inhibition (82.9%), PheA@FA-chol-BSA-NP group showed 45% tumor inhibition value and lastly, PheA treated showed only 30% tumor inhibition value.162
In a non-invasive procedure called sonodynamic therapy (SDT), fatal ROS are created using ultrasound (US) and sonosensitizers.178,179 It has many advantages such as good therapeutic accuracy, deeper tissue penetration, and minimal skin damage.180,181 A study conducted by Cao et al used SDT to treat breast cancer. They developed extracellular vesicles (EVs) based nano sensitizers by using Chlorin e6 (Che6) as a sonosensitizer, linked to TPP moiety that targets the mitochondria and for achieving effective and focused anticancer activity piperlonguime (PL) which is cancer-specific chemotherapeutic and pro-oxidant was further co-encapsulated into EVs as demonstrated in (Figure 7). After performing cellular uptake studies on MC-7 cells good cellular intake levels of both TPP-Che6 and EV (TPP-Che6) was observed but comparatively EV (TPP-Che6) is better than TPP-Che6 because it facilitated cellular internalization via ligand-receptor connections or EV fusion directly with cell membranes. To determine the mitochondrial absorption of TPP-Che6, Che6, and EV (TPP-Che6), Mander’s overlap coefficient was calculated and more Mander’s overlap coefficient of TPP-Che6 and EV (TPP-Che6) than the Che6 group was observed. This is because of the mitochondria-targeting properties of TPP and it is also clear from these data that TPP-Che6 effectively targeted MCF-7 cells mitochondria. To exhibit ROS production from TPP-Che6 by US exposure, ROS production levels in EV (TPP-Che6/PL), EV (TPP-Che6), TPP-Che6, and free Che6 were determined in a system without cells. The US irradiation significantly raised the ROS production levels of EV (TPP-Che6/PL), EV (TPP-Che6), and TPP-Che6 demonstrating the sonodynamic action of TPP-Che6. Upon US exposure, EV (TPP-Che6/PL) and EV (TPP-Che6) both generated much more ROS than TPP-Ce6. This was because entrapped TPP-Ce6 in EVs protects it from degradation, which allows it to generate more ROS. Intracellular ROS levels in MCF-7 cells treated with TPP-Che6-loaded EV samples after exposure to the US were measured using DCF-DA dye. TPP-Che6 was capable of producing more ROS than Ce6. The possible reason could be that TPP-Che6 has a higher rate of mitochondrial absorption than Che6. In vivo, tumor accumulation efficiency was evaluated by real-time fluorescence imaging after samples were injected intravenously into mice having MCF-7 tumors. EV (TPP-Che6/PL)-administered mice’s tumors exhibited fluorescence till 24 h after intravenous administration. However, TPP-Che6 treated mice’s tumors fluorescence almost vanished after 24 h of intravenous administration. This is because TPP-Che6 encapsulation in Evs enhanced TPP-Che6 accumulation within the tumor through EPR effects. To evaluate the in vivo toxicity, the chief organs of mice given TPP-Che6, EV (TPP-Che6), and EV (TPP-Che6/PL) did not show any significant pathological changes when exposed to US radiation on H&E staining images which indicates the high biosafety of EV loaded with TPP-Ce6.163
![]() |
Figure 7 (A) TPP-Che6 synthetic scheme. (B) Schematic representation of combination chemo-sonodynamic therapy (chemo-SDT) employing biocompatible extracellular vesicles (EVs) containing pro-oxidant piperlongumine (PL) and mitochondria-targeting TPP-Che6. The release of TPP-Che6 and PL from the EVs was activated by ultrasonic (US) irradiation after EV (TPP-Che6/PL) was internalized into breast cancer cells by endocytosis. Under US irradiation, the released TPP-Che6 effectively builds up in the mitochondria and produces reactive oxygen species (ROS). The ROS causes mitochondrial damage, which leads to apoptosis. Through the excessive production of intracellular ROS caused by the released PL, mitochondria-targeted sonodynamic cancer therapy in conjunction with chemotherapy is made possible. Image reproduced from Nguyen Cao TG, Truong Hoang Q, Hong EJ, et al. Mitochondria-targeting sonosensitizer-loaded extracellular vesicles for chemo-sonodynamic therapy. J Control Release. 2023;354:651–663, Copyright (2023), with permission from Elsevier.163 |
Chemotherapy medications can be used as neoadjuvant and adjuvant therapy for the management of certain cancers to slow tumor growth and trigger apoptosis in tumor cells.182 Chemodynamic therapy amplifies the anti-tumor effect by using the Fenton reaction to produce the “OH” group highly in tumors.183 Generally, this nanomaterial liberates metal ions (like vanadium ion, copper ion, manganese ion, ferrous ion, chromium ion, and so on), which convert hydrogen peroxide into high OH in the slightly acidic environment of the tumor. Ultimately, it leads to cell death and the inhibition of tumor growth.184 By combining chemotherapy and chemodynamic therapy, Zhong et al prepared a pH-responsive nanoformulation TPP-DOX@MnBSA (TD@MnB) by using albumin from bovine serum (BSA) as a carrier and co-loading TPP-doxorubicin (TPP-DOX), and manganese ions (Mn). The results obtained from cytotoxicity assay in vitro showed that TPP-DOX can efficiently eliminate MCF-7/ADR cells that are more resistant to DOX than DOX alone. Additionally, the fact that TD@MnB is capable of killing drug-sensitive and drug-resistant MCF-7 cells indicates that it has a greater ability to eliminate actual heterogeneous tumors. After performing a cellular uptake study, it was concluded that DOX@MnB and TD@MnB cellular uptake are remarkably more than free DOX. This is because of the presence of albumin which can be more effectively absorbed by tumor cells, which demand large amounts of nutrients. TPP-DOX is more widely uptaken by the tumor cells than DOX because the positive charge of the lipophilic cation TPP in TPP-DOX interacts with the negatively charged cell membranes to facilitate TPP-DOX uptake. The results obtained from the transwell experiment which was done to calculate the inhibition of cell migration are that TD@MnB, TPP-DOX, DOX@MnB, and DOX all are suitable for inhibiting tumor cell migration, but still, TD@MnB and TPP-DOX are much more efficient than free DOX and DOX@MnB. This is due to the reason that the capacity of free DOX to prevent tumor metastasis is less than that of mitochondrial targeting TPP-DOX. After performing in vivo biodistribution of TD@MnB, TPP-DOX, and DOX on tumor-bearing mouse model by injecting an equivalent dose of DOX, the drug distribution in the kidney, lung, spleen, liver, heart, and tumor tissues was examined at 12 and 24 h, and it was concluded that the final formulation TD@MnB which consists of TPP has relative enrichment of tumor locations and longer system circulation time compared to free DOX.164
An investigation by Manesh and teammates involved the conjugation of the PTX molecule to TPP through a pH-sensitive ester bond for the delivery of anticancer drugs to mitochondria. The Hydroxyl group of PTX and the carboxylic group of TPP are responsible for the formation of Prodrug (PTX-TPP). Further, human serum albumin (HSAb) NPs modified with Aptamer (Ap) MUC1 were loaded with the PTX-TPP prodrug to create a selective multi-targeting system that is targeted specifically to mitochondria. To determine the cellular uptake and mitochondrial targeting, FITC-labeled samples have been applied to MCF-7 cells (FITC-TPP, FITC-HSAb NPs, FITC-TPP-HSAb NPs, and FITC-TPP-HSAb-Ap NP) then continued to be treated by DNA stain DAPI and MTred. Results showed that FITC-TPP-HSAb-Ap NPs are highly cellularly absorbed and localized in the mitochondria. The potential cause could be that the TPP lipophilic cation causes mitochondrial localization and cellular absorption could be improved by modifying HSAb-NPs with an Ap targeting agent. On determining apoptotic activity, results revealed that the apoptotic rate induced by PTX-TPP-HSAb-Ap NPs is higher as compared to PTX-TPP-HSAb NPs, TPP-HSAb, PTX-TPP, PTX, and Ap NPs (blank targeting NPs). The possible reason could be that aptamer-modified NPs with PTX-TPP can give MCF-7 cells sufficient PTX concentration at the mitochondrial site. The results from the cytotoxicity study when performed on MUC1 under expressing MDA-MB-435 cells and MUC1 overexpressing MCF-7 cell revealed more cytotoxicity of PTX-TPP with IC50 value 60.4 nmol when compared to plain PTX. This is because plasma membrane potential allows TPP-conjugated PTX to accumulate in the cytoplasm through the extracellular environment. The final formulation TPP-HSAb-Ap NPs demonstrated greater cytotoxicity in MCF-7 cells which is due to overexpressing of MUC1 that resulted in higher cellular uptake via receptor-mediated endocytotic pathway whereas in contrast, MDA-MB-435 cells do not express MUC1.165
Another interesting investigation by Arafa et al utilized a core-shell nanoparticulate system that targeted sequentially for the management of breast cancer. At first, cellular level targeting by the nanoparticulate system and then further subcellular level targeting was achieved. Cellular-level targeting is made possible by using hyaluronic acid (HUA), a specific CD44 receptor ligand, and with the help of TPP subcellular mitochondrial targeting was achieved. DOX-loaded NPs, DOX-TPP conjugate-loaded NPs, and free DOX were used in MCF-7 breast cancer cells to examine the cytotoxicity concentration of HUA-coated core-shell NPs. The analysis demonstrates that the cytotoxicity of NPs was considerably increased by increasing the HUA content. This is because of the improved cellular uptake based on CD-44 receptors. On further comparison, it was confirmed that after TPP conjugation, cell viability was significantly reduced due to the ability to target the mitochondria of tumor cells. Solid Ehrlich carcinoma (SEC) bearing mice were used to evaluate the anti-tumor efficacy of DOX-TPP and their loaded NPs, and the result was that both DOX-TPP and their loaded NPs significantly reduced the volume of the tumor. Based on the above experiments and results, it was concluded that the core-shell NPs developed for treating MCF-7 breast cancer cell lines exhibited potent anti-tumor activity, leading to apoptosis and cell cycle arrest.156
Different research studies have been explored based on mitochondrial-targeted drug delivery by TPP and they all have shown promising therapeutic outcomes in cancer treatment. Although all these researches aim is to kill cancerous cells by damaging their mitochondria, they used different materials, methods, and targeting approaches. For instance, EV(TPP-Ce6/PL) utilized extracellular vesicles to co-administer pro-oxidant and sonosensitizer to enhance chemotherapy and sonodynamic therapy by synergistic generation of ROS. On the contrary, another system is TD@MB nanoparticles which enhance ROS levels through a Fenton-like reaction which is catalyzed by manganese and its incorporated chemotherapeutic drug was released in a pH-responsive manner. On the other hand, a dual-targeting strategy was employed by another nanosystem (PTX-TPP-HSA-Apt NPs) through MUC1 aptamer for cancerous cells and TPP for mitochondria. Due to this cytotoxicity was enhanced by 18 times than free paclitaxel. Similarly, chitosan nanoparticles coated by HUA utilized CD44 receptors to target breast cancerous cells in a controlled and pH-sensitive release. Furthermore, triple-negative breast cancer which is very hard to treat was treated by nanosystem HUA-TS-TPP/LPT NPS. Another prominent example is TPP-PEG-modified liposomes which demonstrate significant antitumor activity and considerable mitochondrial accumulation. Conclusively, all the strategies signify the potential of mitochondrial targeting through TPP. Moreover, treatment effectiveness was significantly enhanced after combining it with different approaches as we have discussed like aptamer guided, ROS amplification, and so on. This comparative evaluation highlights the therapeutic advantages, targeting mechanisms, and different design strategies which provide us a comprehensive insight into how these studies differ and complement one another.
Conclusion
Recently, there has been increased focus on employing nanocarriers tailored for mitochondria to deliver chemotherapeutic agents with precision to these cellular organelles. Triphenylphosphonium stands out among these carriers for its remarkable targeting prowess, promising precise delivery. This approach not only reduces drug dosage but also mitigates the indiscriminate side effects on normal tissue. Hence, this review succinctly examines the pivotal role of nanotechnology, underscores the importance of targeted delivery, and explores the potential benefits of targeting at both cellular and sub-cellular levels in cancer treatment. Furthermore, it delves deeply into mitochondrial targeting, particularly emphasizing nanocarriers engineered for mitochondrial delivery, especially those coupled with triphenylphosphonium, within the realm of cancer therapy.
Mitochondrial targeting with TPP is emerging as an innovative approach in cancer management. This review describes various types of TPP-conjugated NPs developed by numerous researchers like polymer lipid nanoparticles, carbon nanodots, extracellular vesicles, and albumin-based nanoparticles. Intracellular uptake and mitochondrial accumulation were enhanced by these TPP-functionalized nanosystems and induced apoptosis by the formation of ROS and disruption of the mitochondrial membrane. One of the major concerns associated with TPP is its cytotoxicity but this happens only at high doses, it was evident from in-vivo results after animal models being tested by TPP-derivative Mito-VitE, TPMP, and MitoQ and it was further proved by the commercial availability of MitoQ. The other challenge with TPP is hemolysis because of its positive charge and researchers mitigate this issue by encapsulating it into nanocarriers, because of which it only interacts with mitochondria after reaches to desired location. It was found that the non-specificity of cells by chemotherapeutic agents was overcome by using the targeting moiety, which enhances the accumulation of the NPs on tumor cells and decreases the dose, ultimately reducing the adverse effects associated with the drug. Ultra-high cytotoxicity in cancer cell lines was achieved by using mitochondrial-targeted NPs rather than non-targeted NPs. In vivo studies also confirmed that tumor growth inhibition is greater in the case of mitochondrial-targeted NPs than in non-targeted NPs. Hence, it can be inferred that NPs utilizing TPP for mitochondrial targeting hold significant promise in treating diverse cancer types. Their notable attributes include forming connections with chemotherapy drugs, displaying responsiveness to pH changes, supporting PTT, and ensuring targeted drug delivery to combat MDR. This review furnishes an up-to-date and comprehensive understanding of mitochondrial targeting using TPP-conjugated NPs for cancer treatment, serving as a valuable resource for scientists and researchers in this domain. Research should be advanced in the future by focusing on developing novel derivatives of TPP and multifunctional nanocarriers that respond to cancerous microenvironments more efficiently and integrate both therapeutic and diagnostic. Moreover, it is imperative to emphasize the necessity for additional clinical evidence and research before these interventions can be introduced into the market. Conclusively, TPP-based mitochondrial targeting offers a promising approach to next-generation cancer treatment.
Disclosure
There is no conflict of interest and disclosures associated with the work.
References
1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11–30. doi:10.3322/caac.21166
2. Gowda BHJ, Ahmed MG, Almoyad MA, et al. Nanosponges as an emerging platform for cancer treatment and diagnosis. Adv Funct Mater. 2023:2307074. doi:10.1002/ADFM.202307074
3. Hani U, Gowda BHJ, Haider N, et al. Nanoparticle-based approaches for treatment of hematological malignancies: a comprehensive review. AAPS PharmSciTech. 2023;24:1–24. doi:10.1208/s12249-023-02670-0
4. Gowda BHJ, Ahmed MG, Alshehri SA, et al. The cubosome-based nanoplatforms in cancer therapy: seeking new paradigms for cancer theranostics. Environ Res. 2023;237:116894. doi:10.1016/j.envres.2023.116894
5. Siegel RL, Miller KD, Jemal A. Cancer statistics. CA. 2018;68:7–30.
6. Hani U, Jaswanth Gowda BH, Siddiqua A, et al. Herbal approach for treatment of cancer using curcumin as an anticancer agent: a review on novel drug delivery systems. J Mol Liq. 2023;390:123037. doi:10.1016/j.molliq.2023.123037
7. Ahamed J, Jaswanth Gowda BH, Almalki WH, et al. Recent advances in nanoparticle-based approaches for the treatment of brain tumors: opportunities and challenges. Eur Polym J. 2023;193:112111. doi:10.1016/j.eurpolymj.2023.112111
8. Banazadeh M, Behnam B, Ganjooei NA, et al. Curcumin-based nanomedicines: a promising avenue for brain neoplasm therapy. J Drug Deliv Sci Technol. 2023;89:105040. doi:10.1016/j.jddst.2023.105040
9. Gowda BHJ, Ahmed MG, Chinnam S, et al. Current trends in bio-waste mediated metal/metal oxide nanoparticles for drug delivery. J Drug Deliv Sci Technol. 2022;71:103305. doi:10.1016/j.jddst.2022.103305
10. Narayana S, et al. Recent advances in ocular drug delivery systems and targeting VEGF receptors for management of ocular angiogenesis: a comprehensive review. Futur J Pharm Sci. 2021;71(7):1–21.
11. Zeng L, et al. Advancements in nanoparticle-based treatment approaches for skin cancer therapy. Mol Cancer. 2023;221(22):1–50.
12. Khan MS, Gowda BHJ, Nasir N, et al. Advancements in dextran-based nanocarriers for treatment and imaging of breast cancer. Int J Pharm. 2023;643:123276. doi:10.1016/j.ijpharm.2023.123276
13. Yang J, Griffin A, Qiang Z, Ren J. Organelle-targeted therapies: a comprehensive review on system design for enabling precision oncology. Signal Transduct Target Ther. 2022;71(7):1–27.
14. Li Z, Zou J, Chen X. In response to precision medicine: current subcellular targeting strategies for cancer therapy. Adv Mater. 2023;35:2209529. doi:10.1002/adma.202209529
15. Khan AA, Allemailem KS, Almatroudi A, et al. Novel strategies of third level (Organelle-specific) drug targeting: an innovative approach of modern therapeutics. J Drug Deliv Sci Technol. 2021;61:102315. doi:10.1016/j.jddst.2020.102315
16. Dhiman A, Handa M, Ruwali M, et al. Recent trends of natural based therapeutics for mitochondria targeting in Alzheimer’s disease. Mitochondrion. 2022;64:112–124. doi:10.1016/j.mito.2022.03.006
17. Khan MS, Jaswanth Gowda BH, Almalki WH, et al. Unravelling the potential of mitochondria-targeted liposomes for enhanced cancer treatment. Drug Discov Today. 2024;29:103819. doi:10.1016/j.drudis.2023.103819
18. Dubey SK, et al. Microparticulate and nanotechnology mediated drug delivery system for the delivery of herbal extracts. J Biomater Sci Polym. 2022:1–24. doi:10.1080/09205063.2022.2065408
19. Hani U, et al. Novel drug delivery systems as an emerging platform for stomach cancer therapy. Pharm. 2022;14:157614,1576.
20. Battogtokh G, Cho YY, Lee JY, et al. Mitochondrial-targeting anticancer agent conjugates and nanocarrier systems for cancer treatment. Front Pharmacol. 2018;9:400259. doi:10.3389/fphar.2018.00922
21. Hoye AT, Davoren JE, Wipf P, Fink MP, Kagan VE. Targeting mitochondria. Acc Chem Res. 2008;41:87–97. doi:10.1021/ar700135m
22. Zielonka J, Joseph J, Sikora A, et al. Mitochondria-targeted triphenylphosphonium-based compounds: syntheses, mechanisms of action, and therapeutic and diagnostic applications. Chemical Reviews. 2017;117:10043–10120. doi:10.1021/acs.chemrev.7b00042
23. Gao Y, Tong H, Li J, et al. Mitochondria-targeted nanomedicine for enhanced efficacy of cancer therapy. Front Bioeng Biotechnol. 2021;9:720508. doi:10.3389/fbioe.2021.720508
24. Wang JY, Li JQ, Xiao YM, Fu B, Qin ZH. Triphenylphosphonium (TPP)-based antioxidants: a new perspective on antioxidant design. ChemMedChem. 2020;15:404–410. doi:10.1002/cmdc.201900695
25. Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Control Release. 2015;200:138–157. doi:10.1016/j.jconrel.2014.12.030
26. Chandra J, Hasan N, Nasir N, et al. Nanotechnology-empowered strategies in treatment of skin cancer. Environ Res. 2023;235:116649. doi:10.1016/j.envres.2023.116649
27. Rahiman N, Markina YV, Kesharwani P, Johnston TP, Sahebkar A. Curcumin-based nanotechnology approaches and therapeutics in restoration of autoimmune diseases. J Control Release. 2022;348:264–286. doi:10.1016/j.jconrel.2022.05.046
28. Gowda BHJ, et al. Nanoparticle-based therapeutic approaches for wound healing: a review of the state-of-the-art. Mater Today Chem. 2023;27:101319.
29. Sanjana A, Ahmed MG, Gowda BHJ, Surya S. Formulation and characteristic evaluation of tacrolimus cubosomal gel for vitiligo. J Dispersion Sci Technol. 2022. doi:10.1080/01932691.2022.2139716
30. Damiri F, Gowda BJ, Andra S, et al. Chitosan nanocomposites as Scaffolds for bone tissue regeneration. In: Chitosan Nanocomposites: Bionanomechanical Applications; 2023:377–394. doi:10.1007/978-981-19-9646-7_16
31. Kamaly N, Xiao Z, Valencia PM, Radovic-Moreno AF, Farokhzad OC. Targeted polymeric therapeutic nanoparticles: design, development and clinical translation. Chem Soc Rev. 2012;41:2971–3010. doi:10.1039/c2cs15344k
32. Tabish TA, Abbas A, Narayan RJ. Graphene nanocomposites for transdermal biosensing. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2021;13:e1699. doi:10.1002/wnan.1699
33. Mitchell MJ, Jain RK, Langer R. Engineering and physical sciences in oncology: challenges and opportunities. Nat Rev Cancer. 2017;17:659–675. doi:10.1038/nrc.2017.83
34. Hasan N, Imran M, Sheikh A, et al. Cannabis as a potential compound against various malignancies, legal aspects, advancement by exploiting nanotechnology and clinical trials. J Drug Target. 2022;30:709–725. doi:10.1080/1061186X.2022.2056188
35. Kumari S, Choudhary PK, Shukla R, Sahebkar A, Kesharwani P. Recent advances in nanotechnology based combination drug therapy for skin cancer. J Biomater Sci Polym Ed. 2022;1–34. doi:10.1080/09205063.2022.2054399
36. Sristi, Fatima M, Sheikh A, et al. Recent advancement on albumin nanoparticles in treating lung carcinoma. J Drug Target. 2023;31:486–499. doi:10.1080/1061186X.2023.2205609
37. Nazir S, Hussain T, Ayub A, Rashid U, MacRobert AJ. Nanomaterials in combating cancer: therapeutic applications and developments. Nanomed Nanotechnol Biol Med. 2014;10:19–34.
38. Ferrari M. Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer. 2005;5:161–171. doi:10.1038/nrc1566
39. Tabish TA, Narayan RJ. Mitochondria-targeted graphene for advanced cancer therapeutics. Acta Biomater. 2021;129:43–56. doi:10.1016/j.actbio.2021.04.054
40. Ali MS, Jha SK, Gupta G, et al. Advancements in 5-fluorouracil-Loaded liposomal nanosystems: a comprehensive review on recent innovations in nanomedicine for cancer therapy. J Drug Deliv Sci Technol. 2024;96:105730. doi:10.1016/j.jddst.2024.105730
41. Golstein P, Kroemer G. Cell death by necrosis: towards a molecular definition. Trends Biochem Sci. 2007;32:37–43. doi:10.1016/j.tibs.2006.11.001
42. Liu Z-G, Jiao D. Necroptosis, tumor necrosis and tumorigenesis. Cell Stress. 2019;4:1–8. doi:10.15698/cst2020.01.208
43. Warburg O. On the origin of cancer cells. Science. 1956;123:309–314. doi:10.1126/science.123.3191.309
44. Kroemer G. Mitochondrial control of apoptosis: an introduction. Biochem Biophys Res Commun. 2003;304:433–435. doi:10.1016/S0006-291X(03)00614-4
45. Torchilin VP. Targeted pharmaceutical nanocarriers. AAPS J. 2007;9:128–147. doi:10.1208/aapsj0902015
46. Bhagwat GS, Athawale RB, Gude RP, et al. Formulation and development of transferrin targeted solid lipid nanoparticles for breast cancer therapy. Front Pharmacol. 2020;11. doi:10.3389/fphar.2020.614290
47. Patel HK, Gajbhiye V, Kesharwani P, Jain NK. Ligand anchored poly(propyleneimine) dendrimers for brain targeting: comparative in vitro and in vivo assessment. J Colloid Interface Sci. 2016;482:142–150. doi:10.1016/j.jcis.2016.07.047
48. Kesharwani P, Tekade RK, Jain NK. Formulation development and in vitro – in vivo assessment of the fourth-generation Ppi dendrimer as a cancer-targeting vector. Nanomedicine. 2014;9:2291–2308. doi:10.2217/nnm.13.210
49. Singh S, Numan A, Maddiboyina B, et al. The emerging role of immune checkpoint inhibitors in the treatment of triple-negative breast cancer. Drug Discov Today. 2021;26:1721–1727. doi:10.1016/j.drudis.2021.03.011
50. Amjad MW, Amin MCIM, Katas H, et al. In vivo antitumor activity of folate-conjugated cholic acid-polyethylenimine micelles for the codelivery of doxorubicin and siRNA to colorectal adenocarcinomas. Mol Pharm. 2015;12:4247–4258. doi:10.1021/acs.molpharmaceut.5b00827
51. Kurmi BD, Kayat J, Gajbhiye V, Tekade RK. Lung Targeting. Drug Deliv. 2010;781–794.
52. Chadar R, Kesharwani P. Nanotechnology-based siRNA delivery strategies for treatment of triple negative breast cancer. Int J Pharm. 2021;605:120835. doi:10.1016/j.ijpharm.2021.120835
53. Tekade RK, Tekade M, Kesharwani P, D’Emanuele A. RNAi-combined nano-chemotherapeutics to tackle resistant tumors. Drug Discov Today. 2016;21:1761–1774. doi:10.1016/j.drudis.2016.06.029
54. Kaur H, Kesharwani P. Advanced nanomedicine approaches applied for treatment of skin carcinoma. J Control Release. 2021;337:589–611. doi:10.1016/j.jconrel.2021.08.003
55. Rani K, Paliwal S. A review on targeted drug delivery: its entire focus on advanced therapeutics and diagnostics. Sch J Appl Med Sci. 2014;2:328–331.
56. Patnaik S, Gorain B, Padhi S, et al. Recent update of toxicity aspects of nanoparticulate systems for drug delivery. Eur J Pharm Biopharm. 2021;161:100–119. doi:10.1016/j.ejpb.2021.02.010
57. Hasan N, Imran M, Kesharwani P, et al. Intranasal delivery of Naloxone-loaded solid lipid nanoparticles as a promising simple and non-invasive approach for the management of opioid overdose. Int J Pharm. 2021;599:120428. doi:10.1016/j.ijpharm.2021.120428
58. Dongsar TT, Dongsar TS, Abourehab MAS, Gupta N, Kesharwani P. Emerging application of magnetic nanoparticles for breast cancer therapy. Eur Polym J. 2023;187:111898. doi:10.1016/j.eurpolymj.2023.111898
59. Sheikh A, Md S, Kesharwani P. Aptamer grafted nanoparticle as targeted therapeutic tool for the treatment of breast cancer. Biomed Pharmacother. 2022;146:112530. doi:10.1016/j.biopha.2021.112530
60. Tripathi PK, Gorain B, Choudhury H, Srivastava A, Kesharwani P. Dendrimer entrapped microsponge gel of dithranol for effective topical treatment. Heliyon. 2019;5:e01343. doi:10.1016/j.heliyon.2019.e01343
61. Kesharwani P, Mishra V, Jain NK. Validating the anticancer potential of carbon nanotube-based therapeutics through cell line testing. Drug Discov Today. 2015;20:1049–1060. doi:10.1016/j.drudis.2015.05.004
62. Gowda BHJ, Ahmed MG, Sahebkar A, et al. Stimuli-responsive microneedles as a transdermal drug delivery system: a demand-supply strategy. Biomacromolecules. 2022;23:1519–1544. doi:10.1021/ACS.BIOMAC.1C01691
63. Luong D, Kesharwani P, Killinger BA, et al. Solubility enhancement and targeted delivery of a potent anticancer flavonoid analogue to cancer cells using ligand decorated dendrimer nano-architectures. J Colloid Interface Sci. 2016;484:33–43. doi:10.1016/j.jcis.2016.08.061
64. Afshari AR, Sanati M, Mollazadeh H, et al. Nanoparticle-based drug delivery systems in cancer: a focus on inflammatory pathways. Semin Cancer Biol. 2022;86:860–872. doi:10.1016/J.SEMCANCER.2022.01.008
65. Mishra N, et al. Targeted drug delivery: a review. Am J PharmTech Res. 2016;6.
66. Fatima M, Sheikh A, Hasan N, et al. Folic acid conjugated poly(amidoamine) dendrimer as a smart nanocarriers for tracing, imaging, and treating cancers over-expressing folate receptors. Eur Polym J. 2022;170:111156. doi:10.1016/j.eurpolymj.2022.111156
67. Dubey SK, Kali M, Hejmady S, et al. Recent advances of dendrimers as multifunctional nano-carriers to combat breast cancer. Eur J Pharm Sci. 2021;164:105890. doi:10.1016/j.ejps.2021.105890
68. Choudhari M, et al. Evolving new-age strategies to transport therapeutics across the blood-brain-barrier. Int J harm. 2021;120351. doi:10.1016/j.ijpharm.2021.120351
69. Gawde KA, Kesharwani P, Sau S, et al. Synthesis and characterization of folate decorated albumin bio-conjugate nanoparticles loaded with a synthetic curcumin difluorinated analogue. J Colloid Interface Sci. 2017;496:290–299. doi:10.1016/j.jcis.2017.01.092
70. Kesharwani P, Tekade RK, Jain NK. Dendrimer generational nomenclature: the need to harmonize. Drug Discovery Today. 2015;20:497–499. doi:10.1016/j.drudis.2014.12.015
71. Gujral S, Khatri S. A review on basic concept of drug targeting and drug carrier system. Int J Adv Pharmacy Biol Chem. 2013;2(1):1–7.
72. Kesharwani P, Gajbhiye V, Tekade RK, Jain NK. Evaluation of dendrimer safety and efficacy through cell line studies. Curr Drug Targets. 2011;12:1478–1497. doi:10.2174/138945011796818135
73. Sanati M, Afshari AR, Kesharwani P, Sukhorukov VN, Sahebkar A. Recent trends in the application of nanoparticles in cancer therapy: the involvement of oxidative stress. J Control Release. 2022;348:287–304. doi:10.1016/j.jconrel.2022.05.035
74. Sheikh A, Kesharwani P. An insight into aptamer engineered dendrimer for cancer therapy. Eur Polym J. 2021;159:110746. doi:10.1016/j.eurpolymj.2021.110746
75. Jain A, et al. Recent advances in galactose-engineered nanocarriers for the site-specific delivery of siRNA and anticancer drugs. Drug Discov Today. 2018;23:960–973. doi:10.1016/j.drudis.2017.11.003
76. Qin W, Chandra J, Abourehab MAS, et al. New opportunities for RGD-engineered metal nanoparticles in cancer. Mol Cancer. 2023;22:87. doi:10.1186/s12943-023-01784-0
77. Kesharwani P, et al. Gold nanoparticles and gold nanorods in the landscape of cancer therapy. Mol Cancer. 2023;221(22):1–31.
78. Sanati M, Afshari AR, Aminyavari S, et al. RGD-engineered nanoparticles as an innovative drug delivery system in cancer therapy. J Drug Deliv Sci Technol. 2023;84:104562. doi:10.1016/j.jddst.2023.104562
79. Kesharwani P, Sheikh A, Abourehab MAS, Salve R, Gajbhiye V. A combinatorial delivery of survivin targeted siRNA using cancer selective nanoparticles for triple negative breast cancer therapy. J Drug Deliv Sci Technol. 2023;80:104164. doi:10.1016/j.jddst.2023.104164
80. Psimadas D, Georgoulias P, Valotassiou V, Loudos G. Molecular Nanomedicine Towards Cancer. J Pharm Sci. 2012;101:2271–2280. doi:10.1002/jps.23146
81. Steichen SD, Caldorera-Moore M, Peppas NA. A review of current nanoparticle and targeting moieties for the delivery of cancer therapeutics. Eur J Pharm Sci. 2013;48:416–427. doi:10.1016/j.ejps.2012.12.006
82. Dongsar TT, Dongsar TS, Gupta N, et al. Emerging potential of 5-Fluorouracil-loaded chitosan nanoparticles in cancer therapy. J Drug Deliv Sci Technol. 2023;82:104371. doi:10.1016/j.jddst.2023.104371
83. Gajbhiye KR, et al. Lipid polymer hybrid nanoparticles: a custom-tailored next-generation approach for cancer therapeutics. Mol Cancer. 2023;221(22):1–44.
84. Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor Agent Smancs. Cancer Res. 1986;46:6387–6392.
85. Sciences M, Monsky WL, Yuan F. Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc Natl Acad Sci USA. 1998;95:4607–4612. doi:10.1073/pnas.95.8.4607
86. Hashizume H, Baluk P, Morikawa S, et al. Openings between defective endothelial cells explain tumor vessel leakiness. Am J Pathol. 2000;156:1363–1380. doi:10.1016/S0002-9440(10)65006-7
87. Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009;30:1073–1081. doi:10.1093/carcin/bgp127
88. Zheng CJ, Ji Z, Cao Z, Ji ZL, Cao ZW, Chen YZ. Therapeutic targets: progress of their exploration and investigation of their therapeutic targets: progress of their exploration. Pharmacol Rev. 2006;58:259–279. doi:10.1124/pr.58.2.4
89. Bae YH, Park K. Targeted drug delivery to tumors: myths, reality and possibility. J Control Release. 2011;153:198–205. doi:10.1016/j.jconrel.2011.06.001
90. Singh N, Handa M, Singh V, Kesharwani P, Shukla R. Lymphatic targeting for therapeutic application using nanoparticulate systems. J Drug Target. 2022;30:1017–1033. doi:10.1080/1061186X.2022.2092741
91. Boosting cancer therapy with organelle-targeted nanomaterials. ACS Appl Mater Interf. 2019. doi:10.1021/acsami.9b01370
92. Boonstra MC, de Geus SWL, Prevoo HAJM, et al. Selecting targets for tumor imaging: an overview of cancer-associated membrane proteins. Biomarkers in Cancer. 2016:119–133. doi:10.4137/BIC.S38542
93. Masood F. Polymeric nanoparticles for targeted drug delivery system for cancer therapy. Mater Sci Eng C. 2015;60:569–578. doi:10.1016/j.msec.2015.11.067
94. Jahan ST, Sadat SMA, Walliser M, Haddadi A. Targeted therapeutic nanoparticles: an immense promise to fight against cancer. J Drug Delivery. 2017;2017:1–24. doi:10.1155/2017/9090325
95. Mahdavi Gorabi A, Sadat Ravari M, Sanaei M-J, et al. Immune checkpoint blockade in melanoma: advantages, shortcomings and emerging roles of the nanoparticles. Int Immunopharmacol. 2022;113:109300. doi:10.1016/j.intimp.2022.109300
96. Arab A, Nikpoor AR, Asadi P, et al. Virus-Like Nanoparticles (VLPs) based technology in the development of breast cancer vaccines. Process Biochem. 2022;123:44–51. doi:10.1016/J.PROCBIO.2022.10.020
97. Rehman U, Parveen N, Sheikh A, et al. Polymeric nanoparticles-siRNA as an emerging nano-polyplexes against ovarian cancer. Colloids Surfaces B Biointerfaces. 2022;218:112766. doi:10.1016/j.colsurfb.2022.112766
98. Rajendran L, Knölker H, Simons K. Subcellular targeting strategies. Nat Rev Drug Discov. 2010;9:29–42. doi:10.1038/nrd2897
99. Ma X, Gong N, Zhong L, Sun J, Liang X. Biomaterials future of nanotherapeutics: targeting the cellular sub-organelles. Biomaterials. 2016;97:10–21. doi:10.1016/j.biomaterials.2016.04.026
100. Hu C, Huang Y, Li L. Drp1-dependent mitochondrial fission plays critical roles in physiological and pathological progresses in mammals. Int J Mol Sci. 2017;18:144. doi:10.3390/ijms18010144
101. He D, Ma Z, Xue K, Li H. Juxtamembrane 2 mimic peptide competitively inhibits mitochondrial trafficking and activates ROS-mediated apoptosis pathway to exert anti-tumor effects. Cell Death Dis. 2022;13:264. doi:10.1038/s41419-022-04639-6
102. Leduc-Gaudet J-P, Hussain SNA, Barreiro E, Gouspillou G. Mitochondrial dynamics and mitophagy in skeletal muscle health and aging. Int J Mol Sci. 2021;22:8179. doi:10.3390/ijms22158179
103. Norambuena A, Sun X, Wallrabe H, et al. SOD1 mediates lysosome-to-mitochondria communication and its dysregulation by amyloid-β oligomers. Neurobiol Dis. 2022;169:105737. doi:10.1016/j.nbd.2022.105737
104. Guimarães NSS, Ramos VS, Prado-Souza LFL, et al. Rosemary (Rosmarinus officinalis L.) glycolic extract protects liver mitochondria from oxidative damage and prevents acetaminophen-induced hepatotoxicity. Antioxidants. 2023;12:628. doi:10.3390/antiox12030628
105. Duarte-Hospital C, Tête A, Brial F, et al. Mitochondrial dysfunction as a hallmark of environmental injury. Cells. 2021;11:110. doi:10.3390/cells11010110
106. Smith RAJ, Hartley RC, Cochemé HM, Murphy MP. Mitochondrial pharmacology. Trends Pharmacol Sci. 2012;33:341–352. doi:10.1016/j.tips.2012.03.010
107. Miriyala S, et al. Novel role of 4-hydroxy-2-nonenal in AIFm2-mediated mitochondrial stress signaling. Free Radic Biol Med. 2016;91:68–80.
108. Pu W, Gu Z. Incompatibilities between common mtDNA variants in human disease. Proc Natl Acad Sci USA. 2023;120:e2217452119. doi:10.1073/pnas.2217452119
109. Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Role of cardiolipin in mitochondrial function and dynamics in health and disease: molecular and pharmacological aspects. Cells. 2019;8:728. doi:10.3390/cells8070728
110. Lopez J, Tait SWG. Mitochondrial apoptosis: killing cancer using the enemy within. Br J Cancer. 2015;112:957–962. doi:10.1038/bjc.2015.85
111. Pustylnikov S, Costabile F, Beghi S, Facciabene A. Targeting mitochondria in cancer: current concepts and immunotherapy approaches. Transl Res. 2018;202:35–51. doi:10.1016/j.trsl.2018.07.013
112. Wen S, Zhu D, Huang P. Targeting cancer cell mitochondria as a therapeutic approach. Future Med Chem. 2013;5:53–67. doi:10.4155/fmc.12.190
113. Claus C, Manssen L, Hübner D, et al. Activation of the mitochondrial apoptotic signaling platform during Rubella Virus infection. Viruses. 2015;7:6108–6126. doi:10.3390/v7122928
114. Capasso A, Cerchia C, Di Giovanni C, et al. Ligand-based chemoinformatic discovery of a novel small molecule inhibitor targeting CDC25 dual specificity phosphatases and displaying in vitro efficacy against melanoma cells. Oncotarget. 2015;6:40202–40222. doi:10.18632/oncotarget.5473
115. Tabish TA, Hamblin MR. Mitochondria-targeted nanoparticles (mitoNANO): an emerging therapeutic shortcut for cancer. Biomater Biosyst. 2021;3:100023. doi:10.1016/j.bbiosy.2021.100023
116. Biasutto L, Dong LF, Zoratti M, Neuzil J. Mitochondrially targeted anti-cancer agents. Mitochondrion. 2010;10:670–681. doi:10.1016/j.mito.2010.06.004
117. Peng YB, Zhao ZL, Liu T, et al. A multi-mitochondrial anticancer agent that selectively kills cancer cells and overcomes drug resistance. ChemMedChem. 2017;12:250–256. doi:10.1002/cmdc.201600538
118. Zorova LD, et al. Mitochondrial membrane potential. Anal Biochem. 2017. doi:10.1016/j.ab.2017.07.009
119. Zhang XY, Zhang P. Mitochondria targeting nano agents in cancer therapeutics. Oncology Letters. 2016;12:4887–4890. doi:10.3892/ol.2016.5302
120. Dairkee SH, Hackett AJ. Differential retention of rhodamine 123 by breast carcinoma and normal human mammary tissue. Breast Cancer Res Treatment. 1991;18:57–61. doi:10.1007/BF01975444
121. Wallace DC. Mitochondria and cancer: warburg addressed mitochondria and cancer: warburg addressed. In: Cold Spring Harbor Symposia on Quantitative Biology; 2005:363–374. doi:10.1101/sqb.2005.70.035
122. Neuzil J, Dong L, Rohlena J, Truksa J, Ralph SJ. Mitochondrion Classi fi cation of mitocans, anti-cancer drugs acting on mitochondria. MITOCH. 2012. doi:10.1016/j.mito.2012.07.112
123. Wang H, Xu W. Biochemical and biophysical research communications mito-methyl coumarin, a novel mitochondria-targeted drug with great antitumor potential was synthesized. Biochem Biophys Res Commun. 2017;489:1–7. doi:10.1016/j.bbrc.2017.05.116
124. Chazotte B. Labeling mitochondria with TMRM or TMRE protocol labeling mitochondria with TMRM or TMRE. Cold Spring Harbor Protocols. 2012;2012. doi:10.1101/pdb.prot5641
125. Perelman A, Wachtel C, Cohen M, et al. JC-1: alternative excitation wavelengths facilitate mitochondrial membrane potential cytometry. Cell Death Disease. 2012;3:e430–7. doi:10.1038/cddis.2012.171
126. Cunha-oliveira T, Rego AC, Cardoso SM, et al. Mitochondrial dysfunction and caspase activation in rat cortical neurons treated with cocaine or amphetamine. Brain Research. 2006;89.
127. Murphy MP. Targeting lipophilic cations to mitochondria. Biochimica et Biophysica Acta. 2008;1777:1028–1031. doi:10.1016/j.bbabio.2008.03.029
128. Mani S, Swargiary G, Tyagi S, et al. Nanotherapeutic approaches to target mitochondria in cancer. Life Sci. 2021;281:119773. doi:10.1016/j.lfs.2021.119773
129. Ilmi R, Tseriotou E, Stylianou P, et al. A novel conjugate of Bis[((4-bromophenyl)amino)quinazoline], a EGFR-TK ligand, with a Fluorescent Ru(II)-Bipyridine complex exhibits specific subcellular localization in mitochondria. Mol Pharm. 2019;16:4260–4273. doi:10.1021/acs.molpharmaceut.9b00608
130. Weissig V, Torchilin VP. Towards mitochondrial gene therapy: dQAsomes as a strategy. J Drug Target. 2001;9:1–13. doi:10.3109/10611860108995628
131. Li Q, Huang Y. Mitochondrial targeted strategies and their application for cancer and other diseases treatment. J Pharm Investig. 2020;50:271–293. doi:10.1007/s40005-020-00481-0
132. Wu J, Li J, Wang H, Liu C-B. Mitochondrial-targeted penetrating peptide delivery for cancer therapy. Expert Opin Drug Deliv. 2018;15:951–964. doi:10.1080/17425247.2018.1517750
133. Sancho P, Galeano E, Nieto E, Delgado MD, García-Pérez AI. Dequalinium induces cell death in human leukemia cells by early mitochondrial alterations which enhance ROS production. Leuk Res. 2007;31:969–978. doi:10.1016/j.leukres.2006.11.018
134. Song Y, Liu D-Z, Cheng Y, et al. Dual subcellular compartment delivery of doxorubicin to overcome drug resistant and enhance antitumor activity. Sci Rep. 2015;5:16125. doi:10.1038/srep16125
135. Qian K, Chen H, Qu C, et al. Mitochondria-targeted delocalized lipophilic cation complexed with human serum albumin for tumor cell imaging and treatment. Nanomedicine. 2020;23:102087. doi:10.1016/j.nano.2019.102087
136. Todaro B, Ottalagana E, Luin S, Santi M. Targeting peptides: the new generation of targeted drug delivery systems. Pharmaceutics. 2023;15:1648. doi:10.3390/pharmaceutics15061648
137. Somsri S, Mungthin M, Klubthawee N, et al. A mitochondria-penetrating peptide exerts potent anti-plasmodium activity and localizes at parasites’ mitochondria. Antibiot. 2021;10.
138. Chamberlain GR, Tulumello DV, Kelley SO. Targeted delivery of doxorubicin to mitochondria. ACS Chem Biol. 2013;8:1389–1395. doi:10.1021/cb400095v
139. Fonseca SB, Pereira M, Mourtada R, et al. Rerouting chlorambucil to mitochondria combats drug deactivation and resistance in cancer cells. Chem Biol. 2011;18:445–453. doi:10.1016/j.chembiol.2011.02.010
140. Wisnovsky SP, Wilson J, Radford R, et al. Targeting mitochondrial DNA with a platinum-based anticancer agent. Chem Biol. 2013;20:1323–1328. doi:10.1016/j.chembiol.2013.08.010
141. Zhou M, Li L, Li L, et al. Overcoming chemotherapy resistance via simultaneous drug-efflux circumvention and mitochondrial targeting. Acta Pharm Sin B. 2019;9:615–625. doi:10.1016/j.apsb.2018.11.005
142. Smith RA, Porteous CM, Coulter CV, Murphy MP. Selective targeting of an antioxidant to mitochondria. Eur J Biochem. 1999;263:709–716. doi:10.1046/j.1432-1327.1999.00543.x
143. Kelso GF, Porteous CM, Coulter CV, et al. Selective targeting of a redox-active ubiquinone to mitochondria within cells: antioxidant and antiapoptotic properties. J Biol Chem. 2001;276:4588–4596. doi:10.1074/jbc.M009093200
144. Apostolova N, Victor VM. Molecular strategies for targeting antioxidants to mitochondria: therapeutic implications. Antioxid Redox Signal. 2015;22:686–729. doi:10.1089/ars.2014.5952
145. Liberman EA, Topaly VP, Tsofina LM, Jasaitis AA, Skulachev VP. Mechanism of coupling of oxidative phosphorylation and the membrane potential of mitochondria. Nature. 1969;222:1076–1078. doi:10.1038/2221076a0
146. Kang EB, In I, Lee KD, Park SY. Mitochondria-targeted fluorescent carbon nano-platform for NIR-triggered hyperthermia and mitochondrial inhibition. J Ind Eng Chem. 2017;55:224–233. doi:10.1016/j.jiec.2017.06.053
147. Wang H, Zhang F, Wen H, et al. Tumor- and mitochondria-targeted nanoparticles eradicate drug resistant lung cancer through mitochondrial pathway of apoptosis. J Nanobiotechnology. 2020;18(8).
148. Lampidis TJ, Hasin Y, Weiss MJ, Chen LB. Selective killing of carcinoma cells ‘in vitro’ by lipophilic-cationic compounds: a cellular basis. Biomed Pharmacother. 1985;39:220–226.
149. Kalinovich AV, Mattsson CL, Youssef MR, et al. Mitochondria-targeted dodecyltriphenylphosphonium (C12 TPP) combats high-fat-diet-induced obesity in mice. Int J Obes. 2016;40:1864–1874. doi:10.1038/ijo.2016.146
150. D’Souza GGM, Wagle MA, Saxena V, Shah A. Approaches for targeting mitochondria in cancer therapy. Biochim Biophys Acta - Bioenerg. 2011;1807:689–696. doi:10.1016/j.bbabio.2010.08.008
151. Pathania D, Millard M, Neamati N. Opportunities in discovery and delivery of anticancer drugs targeting mitochondria and cancer cell metabolism. Adv Drug Deliv Rev. 2009;61:1250–1275. doi:10.1016/j.addr.2009.05.010
152. Croll TI, O’Connor AJ, Stevens GW, Cooper-White JJ. A blank slate? Layer-by-layer deposition of hyaluronic acid and chitosan onto various surfaces. Biomacromolecules. 2006;7:1610–1622. doi:10.1021/bm060044l
153. James AM, Sharpley MS, Manas A-RB, et al. Interaction of the mitochondria-targeted antioxidant MitoQ with phospholipid bilayers and ubiquinone oxidoreductases. J Biol Chem. 2007;282:14708–14718. doi:10.1074/jbc.M611463200
154. Huang M, Myers CR, Wang Y, You M. Mitochondria as a novel target for cancer chemoprevention: emergence of mitochondrial-targeting agents. Cancer Prev Res. 2021;14:285–306. doi:10.1158/1940-6207.CAPR-20-0425
155. Biswas S, Dodwadkar NS, Deshpande PP, Torchilin VP. Liposomes loaded with paclitaxel and modified with novel triphenylphosphonium-PEG-PE conjugate possess low toxicity, target mitochondria and demonstrate enhanced antitumor effects in vitro and in vivo. J Control Release. 2012;159:393–402. doi:10.1016/j.jconrel.2012.01.009
156. Arafa KK, Smyth HDC, El-Sherbiny IM. Mitotropic triphenylphosphonium doxorubicin-loaded core-shell nanoparticles for cellular and mitochondrial sequential targeting of breast cancer. Int J Pharm. 2021;606:120936. doi:10.1016/j.ijpharm.2021.120936
157. Kim SG, Robby AI, Lee BC, Lee G, Park SY. Mitochondria-targeted ROS- and GSH-responsive diselenide-crosslinked polymer dots for programmable paclitaxel release. J Ind Eng Chem. 2021;99:98–106. doi:10.1016/j.jiec.2021.04.016
158. Gong X, Wang Z, Zhang L, et al. A novel carbon-nanodots-based theranostic nano-drug delivery system for mitochondria-targeted imaging and glutathione-activated delivering camptothecin. Colloids Surfaces B Biointerfaces. 2022;218:112712. doi:10.1016/j.colsurfb.2022.112712
159. Lee SY, Cho HJ. Mitochondria targeting and destabilizing hyaluronic acid derivative-based nanoparticles for the delivery of Lapatinib to triple-negative breast cancer. Biomacromolecules. 2019;20:835–845. doi:10.1021/acs.biomac.8b01449
160. Ruttala HB, Ramasamy T, Ruttala RRT, et al. Mitochondria-targeting multi-metallic ZnCuO nanoparticles and IR780 for efficient photodynamic and photothermal cancer treatments. J Mater Sci Technol. 2021;86:139–150. doi:10.1016/j.jmst.2021.01.035
161. Huang Z, Li D, Guo F, et al. Mitochondria-targeted photosensitizer based nanoplatform loading glutathione inhibitor for enhanced breast cancer photodynamic therapy. Colloids Surfaces B Biointerfaces. 2022;220:112956. doi:10.1016/j.colsurfb.2022.112956
162. Battogtokh G, Ko YT. Mitochondrial-targeted photosensitizer-loaded folate-albumin nanoparticle for photodynamic therapy of cancer. Nanomed Nanotechnol Biol Med. 2017;13:733–743. doi:10.1016/j.nano.2016.10.014
163. Nguyen Cao TG, Truong Hoang Q, Hong EJ, et al. Mitochondria-targeting sonosensitizer-loaded extracellular vesicles for chemo-sonodynamic therapy. J Control Release. 2023;354:651–663. doi:10.1016/j.jconrel.2023.01.044
164. Zhong X, Bao X, Zhong H, et al. Mitochondrial targeted drug delivery combined with manganese catalyzed Fenton reaction for the treatment of breast cancer. Int J Pharm. 2022;622:121810. doi:10.1016/j.ijpharm.2022.121810
165. Esfandyari-Manesh M, Mohammadi A, Atyabi F, et al. Enhancement mitochondrial apoptosis in breast cancer cells by paclitaxel-triphenylphosphonium conjugate in DNA aptamer modified nanoparticles. J Drug Deliv Sci Technol. 2019;54:101228. doi:10.1016/j.jddst.2019.101228
166. Qin Y, Wang Z, Wang X, et al. Therapeutic effect of multifunctional celastrol nanoparticles with mitochondrial alkaline drug release in breast cancer. Mater Today Adv. 2023;17:100328. doi:10.1016/j.mtadv.2022.100328
167. Chen H, Fang Z, Song M, Liu K. Mitochondrial targeted hierarchical drug delivery system based on HA-modified liposomes for cancer therapy. Eur J Med Chem. 2022;241:114648. doi:10.1016/j.ejmech.2022.114648
168. Manuscript A, Low TCP, Mitochondria T, Enhanced D, Effects A. NIH Public Access. 2013;159:393–402.
169. Mazrad ZAI, Phuong PTM, Choi CA, et al. pH/Redox-Triggered Photothermal Treatment for Cancer Therapy Based on a Dual-Responsive Cationic Polymer Dot. ChemMedChem. 2018;13:2437–2447. doi:10.1002/cmdc.201800538
170. Choi CA, Lee JE, Mazrad ZAI, et al. Redox- and pH-responsive fluorescent carbon nanoparticles-MnO2-based FRET system for tumor-targeted drug delivery in vivo and in vitro. J Ind Eng Chem. 2018;63:208–219. doi:10.1016/j.jiec.2018.02.017
171. Zhai S, Hu X, Hu Y, Wu B, Xing D. Visible light-induced crosslinking and physiological stabilization of diselenide-rich nanoparticles for redox-responsive drug release and combination chemotherapy. Biomaterials. 2017;121:41–54. doi:10.1016/j.biomaterials.2017.01.002
172. Tian Y, Lei M, Yan L, An F. Diselenide-crosslinked zwitterionic nanogels with dual redox-labile properties for controlled drug release. Polym Chem. 2020;11:2360–2369. doi:10.1039/D0PY00004C
173. Mollazadeh S, Mackiewicz M, Yazdimamaghani M. Recent advances in the redox-responsive drug delivery nanoplatforms: a chemical structure and physical property perspective. Mater Sci Eng C. 2021;118:111536. doi:10.1016/j.msec.2020.111536
174. Wilk KA, Zielińska K, Pietkiewicz J, et al. Photo-oxidative action in MCF-7 cancer cells induced by hydrophobic cyanines loaded in biodegradable microemulsion-templated nanocapsules. Int J Oncol. 2012;41:105–116. doi:10.3892/ijo.2012.1458
175. Zhang C, Wang S, Xiao J, et al. Sentinel lymph node mapping by a near-infrared fluorescent heptamethine dye. Biomaterials. 2010;31:1911–1917. doi:10.1016/j.biomaterials.2009.11.061
176. Ruttala HB, Ko YT. Liposomal co-delivery of curcumin and albumin/paclitaxel nanoparticle for enhanced synergistic antitumor efficacy. Colloids Surfaces B Biointerfaces. 2015;128:419–426. doi:10.1016/j.colsurfb.2015.02.040
177. Cheng X, Sun R, Yin L, et al. Light-triggered assembly of gold nanoparticles for photothermal therapy and photoacoustic imaging of tumors in vivo. Adv Mater. 2017;29. doi:10.1002/adma.201604894
178. Lin X, Song J, Chen X, Yang H. Ultrasound-activated sensitizers and applications. Angew Chemie - Int Ed. 2020;59:14212–14233. doi:10.1002/anie.201906823
179. Wang X, Zhong X, Bai L, et al. Ultrafine Titanium Monoxide (TiO 1+ x) Nanorods for Enhanced Sonodynamic Therapy. J Am Chem Soc. 2020;142:6527–6537. doi:10.1021/jacs.9b10228
180. Son S, Kim JH, Wang X, et al. Multifunctional sonosensitizers in sonodynamic cancer therapy. Chem Soc Rev. 2020;49:3244–3261. doi:10.1039/C9CS00648F
181. Wang X, Wang X, Yue Q, et al. Liquid exfoliation of TiN nanodots as novel sonosensitizers for photothermal-enhanced sonodynamic therapy against cancer. Nano Today. 2021;39:101170. doi:10.1016/j.nantod.2021.101170
182. Del Mastro L, De Placido S, Bruzzi P, et al. Fluorouracil and dose-dense chemotherapy in adjuvant treatment of patients with early-stage breast cancer: an open-label, 2 × 2 factorial, randomised Phase 3 trial. Lancet. 2015;385:1863–1872. doi:10.1016/S0140-6736(14)62048-1
183. Zhang C, Bu W, Ni D, et al. Synthesis of iron nanometallic glasses and their application in cancer therapy by a localized Fenton reaction. Angew Chemie – Int Ed. 2016;55:2101–2106. doi:10.1002/anie.201510031
184. Tang Z, Liu Y, He M, Bu W. Chemodynamic therapy: tumour microenvironment-mediated Fenton and Fenton-like reactions. Angew Chemie - Int Ed. 2019;58:946–956. doi:10.1002/anie.201805664
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

Camptothecin-Loaded and Manganese Dioxide-Coated Polydopamine Nanomedicine Used for Magnetic Resonance Imaging Diagnosis and Chemo-Photothermal Therapy for Lung Cancer
Su M, Chen Y, Jia L, Zhang Z
International Journal of Nanomedicine 2022, 17:6687-6705
Published Date: 28 December 2022
Advancing Photodynamic Therapy with Nano-Conjugated Hypocrellin: Mechanisms and Clinical Applications
Rajan SS, Chandran R, Abrahamse H
International Journal of Nanomedicine 2024, 19:11023-11038
Published Date: 1 November 2024
Development of Cytolytic Iridium-Complexed Octaarginine Peptide Albumin Nanomedicine for Hepatocellular Carcinoma Treatment
Sun X, Wang D, Chang S, Yin L, Zhang H, Ji S, Fei H, Jin Y
International Journal of Nanomedicine 2025, 20:2395-2409
Published Date: 25 February 2025