1Assistant Professor, University College of Pharmaceutical Sciences, Guru Kashi University, Talwandi Sabo, Bathinda, Punjab, India
2Associate Professor, Department of Pharmaceutics, Siddharth College of Pharmacy, Mudhol, Bagalkot, Karnataka, India
3Assistant Professor, Department of Microbiology, Dinabandhu Andrews College, Baishnabghata, South 24 Parganas, Kolkata, West Bengal, India
4Associate Professor, Department of Pharmacy, Institute of Pharmacy, Vikram University, Ujjain, Madhya Pradesh, India
5Assistant Professor, Department of Pharmaceutics, Sardar Patel College of Pharmacy,Vidyanagar - Vadtal Road,Bakrol, Dist& Ta-Anand, Gujarat,India
6Assistant Professor, Department of Pharmacy, Chhatrapati Shivaji College of Pharmacy, Deori, Maharashtra, India
7Associate Professor & Dean, Faculty of Pharmacy, Guru Kashi University, Bhatinda, Punjab, India
8Assistant Professor, Department of Pharmacy, Shri Venkateshwara University, Gajraula, Uttar Pradesh, India
*9Assistant Professor, Himalayan School of Pharmaceutical Sciences, Swami Rama Himalayan University, Jolly Grant, Dehradun, India
Rare mitochondrial and peroxisomal disorders represent a group of devastating, genetically inherited metabolic diseases characterized by organelle dysfunction, disrupted cellular homeostasis, and profound clinical heterogeneity. Current treatment strategies are largely palliative, with conventional pharmacotherapies demonstrating limited efficacy due to poor intracellular targeting and systemic toxicity. Organelle-specific drug delivery has emerged as a transformative approach in precision medicine, offering enhanced therapeutic precision, reduced off-target effects, and improved bioavailability at the subcellular level. This review explores the pathophysiological landscape of mitochondrial and peroxisomal disorders, highlighting the molecular underpinnings and clinical manifestations of key syndromes such as MELAS, Leigh syndrome, Zellweger spectrum disorders, and X-linked adrenoleukodystrophy. We systematically discuss organelle-targeting strategies, including lipophilic cations, mitochondria-penetrating peptides, peroxisomal targeting signals, and ligand-conjugated nanocarriers. Furthermore, we evaluate nanocarrier platforms such as liposomes, dendrimers, polymeric nanoparticles, and biomimetic systems that have been engineered for organelle-directed therapies. Preclinical and clinical progress in targeting these organelles using antioxidant drugs, gene delivery vectors, and enzyme-loaded nanoparticles is examined, along with existing translational and regulatory challenges. Finally, the review delves into emerging trends such as CRISPR/Cas-based genome editing, artificial organelles, and AI-driven nanomedicine design, outlining a forward-looking roadmap for organelle-specific therapeutics in ultra-rare disease contexts.
Rare mitochondrial and peroxisomal disorders represent a class of genetically inherited metabolic diseases characterized by defects in organelle function that critically impair cellular homeostasis and energy metabolism. Although individually rare, collectively these disorders affect thousands worldwide, with mitochondrial diseases estimated to occur in approximately 1 in 4,300 individuals (Gorman et al., 2016), while peroxisomal disorders like Zellweger spectrum disorders are reported with an incidence of 1 in 50,000 to 1 in 100,000 live births (Braverman et al., 2016). These conditions stem from mutations in either mitochondrial DNA (mtDNA) or nuclear genes encoding mitochondrial or peroxisomal proteins, leading to clinical heterogeneity that ranges from neurodegeneration and developmental delays to multi-organ failure (Wanders et al., 2020; Rahman, 2020).
Despite advancements in molecular diagnostics and disease classification, therapeutic options for these disorders remain limited and primarily supportive rather than curative. Conventional pharmacological interventions face major challenges due to poor bioavailability, lack of specificity, and limited ability to cross subcellular barriers to reach dysfunctional organelles (Vafai & Mootha, 2012). Additionally, systemic drug administration often leads to off-target effects and cumulative toxicity, further complicating long-term disease management (Wallace, 2013).
To address these limitations, organelle-specific drug delivery systems have emerged as a transformative strategy, particularly with the rise of nanotechnology-enabled precision therapeutics. These systems exploit the unique biochemical and structural properties of mitochondria and peroxisomes to facilitate targeted drug accumulation, thereby improving efficacy while minimizing systemic side effects (Ramasamy et al., 2020). For example, nanocarriers functionalized with mitochondria-targeting moieties such as triphenylphosphonium (TPP?) or peroxisomal targeting signals (PTS) can selectively deliver bioactive agents to their intended subcellular destinations (Pathak et al., 2014). This review aims to provide a comprehensive overview of organelle-specific drug delivery systems in the context of rare mitochondrial and peroxisomal disorders. It discusses the molecular pathology of these conditions, evaluates current and emerging nanocarrier platforms for targeted therapy, and explores translational advancements and challenges in the field. By bridging insights from molecular biology, materials science, and pharmacology, this review highlights the growing relevance of subcellular targeting in the era of precision nanotherapeutics.
II. Mitochondrial and Peroxisomal Disorders: Pathophysiological Landscape
A. Mitochondrial Disorders
Mitochondrial disorders are a group of clinically heterogeneous diseases caused by mutations in either mitochondrial DNA (mtDNA) or nuclear DNA that encodes mitochondrial proteins. mtDNA mutations are maternally inherited and often affect genes involved in oxidative phosphorylation, while nuclear gene mutations follow Mendelian inheritance patterns and can impact mitochondrial structure, function, or biogenesis (Gorman et al., 2016; Vafai & Mootha, 2012).
Common mitochondrial disorders include:
The pathophysiology is primarily driven by impaired oxidative phosphorylation leading to inadequate ATP production, excessive generation of reactive oxygen species (ROS), and subsequent oxidative damage to proteins, lipids, and DNA (Wallace, 2013). These biochemical abnormalities often culminate in multi-system involvement, particularly affecting energy-demanding organs such as the brain, muscles, and heart.
B. Peroxisomal Disorders
Peroxisomal disorders encompass a range of genetic diseases caused by dysfunction of peroxisomes—organelles involved in lipid metabolism, hydrogen peroxide detoxification, and bile acid synthesis (Wanders et al., 2020). These disorders are broadly classified into:
ZSDs manifest with craniofacial abnormalities, hypotonia, seizures, and profound developmental delays, often resulting in early mortality. In contrast, X-ALD is characterized by the accumulation of very-long-chain fatty acids (VLCFAs) in tissues, leading to inflammatory demyelination and adrenal insufficiency (Wiesinger et al., 2013). Dysregulation of peroxisomal β-oxidation and plasmalogen biosynthesis in these conditions impairs cellular membrane integrity and results in neurodegeneration, systemic inflammation, and hepatic dysfunction (Ferdinandusse et al., 2016).
Table 1. Representative Mitochondrial and Peroxisomal Disorders and Their Key Features
Disorder |
Organelle |
Genetic Cause |
Pathophysiology |
Clinical Features |
MELAS |
Mitochondria |
mtDNA (e.g., m.3243A>G) |
Impaired oxidative phosphorylation |
Stroke-like episodes, myopathy |
Leigh Syndrome |
Mitochondria |
Nuclear/mtDNA |
Complex I/IV deficiency |
Psychomotor regression, lactic acidosis |
LHON |
Mitochondria |
mtDNA (e.g., ND1, ND4) |
ROS accumulation, optic nerve atrophy |
Vision loss, optic neuropathy |
Zellweger Spectrum Disorders |
Peroxisomes |
PEX genes (e.g., PEX1) |
Defective peroxisome biogenesis |
Craniofacial dysmorphism, hypotonia |
X-linked Adrenoleukodystrophy |
Peroxisomes |
ABCD1 |
VLCFA accumulation, demyelination |
Adrenal insufficiency, neurodegeneration |
Figure 1: Flowchart of Classification and Pathogenesis inherited organelle disorders
III. Organelle-Specific Targeting Strategies
Targeting drugs directly to mitochondria or peroxisomes represents a transformative strategy in treating organelle-specific disorders. This approach enhances therapeutic efficacy, minimizes systemic toxicity, and aligns with the principles of precision medicine. Recent advances in nanotechnology have facilitated the design of delivery platforms that exploit organelle-specific physicochemical and biochemical features.
A. Mitochondrial Targeting Approaches
1. Lipophilic Cations (e.g., TPP?-Conjugated Drugs)
Triphenylphosphonium (TPP?) is a delocalized lipophilic cation that can selectively accumulate in the mitochondrial matrix, driven by the negative membrane potential of the inner mitochondrial membrane (Murphy & Smith, 2007). TPP? has been used to deliver a range of antioxidants, chemotherapeutics, and imaging agents directly to mitochondria, enhancing both bioavailability and organelle-specific action (Smith et al., 2012).
2. Mitochondria-Penetrating Peptides (MPPs)
MPPs are short peptides engineered to possess both positive charge and moderate lipophilicity, enabling them to translocate across mitochondrial membranes. They are capable of delivering cargo such as nucleic acids, proteins, and small molecules with high mitochondrial selectivity (Horton et al., 2008). Examples include Szeto-Schiller (SS) peptides and synthetic amphipathic carriers that preserve mitochondrial membrane integrity while facilitating uptake.
3. pH/Redox-Sensitive Nanoparticles
The mitochondrial microenvironment, particularly its slightly alkaline pH and redox state, can be exploited for selective drug release. Nanocarriers loaded with pH- or glutathione-responsive linkers degrade preferentially within mitochondria, ensuring that the drug payload is activated only at the target site (Yousaf et al., 2021). This strategy is especially valuable for delivering ROS-sensitive drugs or mitochondrial enzyme modulators.
B. Peroxisomal Targeting Approaches
1. Peroxisomal Targeting Signals (PTS1/PTS2)
Endogenous protein import into peroxisomes is mediated by peroxisomal targeting signals. PTS1 is a C-terminal tripeptide (Ser-Lys-Leu), while PTS2 is an N-terminal nonapeptide. These sequences can be engineered onto therapeutic proteins or enzymes to facilitate their direct import into peroxisomes via the PEX receptor system (Braverman et al., 2016; Purdue & Lazarow, 2001).
2. Ligand-Conjugated Nanoparticles
Targeting peroxisomes with ligand-conjugated nanoparticles remains an emerging field. Ligands that bind selectively to peroxisomal membrane receptors or associated enzymes may serve as anchors for nanoparticles, directing their intracellular trafficking (Weller et al., 2013). Peptides mimicking peroxisomal import signals or receptor-specific aptamers are being explored to enhance targeting specificity.
3. Enzyme-Loaded Nanocarriers
To address single-enzyme deficiencies in peroxisomal disorders, nanocarriers have been developed to encapsulate and deliver functional enzymes such as acyl-CoA oxidase or peroxisomal catalase. These platforms are often designed with endosomal escape capabilities and peroxisomal targeting motifs, ensuring the bioactive enzyme reaches the correct subcellular compartment (Reddy et al., 2011).
Table 2. Organelle-Targeting Strategies and Their Mechanistic Attributes
Strategy |
Organelle |
Mechanism of Targeting |
Therapeutic Application |
TPP?-Conjugated Compounds |
Mitochondria |
Membrane potential-driven accumulation |
Antioxidant delivery in mitochondrial diseases |
Mitochondria-Penetrating Peptides |
Mitochondria |
Lipophilicity and cationic charge |
Delivery of proteins and nucleic acids |
Redox-Sensitive Nanoparticles |
Mitochondria |
Redox-responsive release mechanisms |
Controlled drug activation |
PTS1/PTS2 Signal Tags |
Peroxisomes |
Receptor-mediated import |
Enzyme replacement therapies |
Ligand-Conjugated Nanoparticles |
Peroxisomes |
Ligand–receptor interactions |
Emerging peroxisomal targeting |
Enzyme-Loaded Nanocarriers |
Peroxisomes |
Nanocarrier-based enzyme delivery |
Correction of metabolic deficiencies |
IV. Nanocarrier Platforms for Organelle-Specific Delivery
Nanocarrier platforms serve as precision tools for delivering therapeutic agents directly to intracellular organelles such as mitochondria and peroxisomes. Their structural diversity, biocompatibility, and functional tunability enable the incorporation of targeting moieties, responsive release mechanisms, and diagnostic functionalities—key attributes for treating rare metabolic disorders characterized by subcellular dysfunction.
A. Liposomes and Dendrimers
Liposomes are phospholipid bilayer vesicles capable of encapsulating hydrophilic and hydrophobic drugs. Through surface modification with targeting ligands like triphenylphosphonium (TPP?) or peroxisomal peptides, liposomes can achieve organelle specificity (Bozzuto & Molinari, 2015). Dendrimers, with their highly branched structure and modifiable surface groups, offer multivalency for conjugating both therapeutic and targeting agents. They have demonstrated success in mitochondrial targeting via conjugation with delocalized lipophilic cations and in delivering antioxidant enzymes for peroxisomal restoration (Tomalia et al., 2012). In rare metabolic diseases such as Zellweger spectrum disorders and mitochondrial encephalopathies, liposome and dendrimer-based carriers show promise for overcoming enzyme mislocalization and drug inefficacy (Wang et al., 2020).
B. Polymeric Nanoparticles
Polymeric nanoparticles fabricated from biocompatible and biodegradable polymers like poly(lactic-co-glycolic acid) (PLGA) and PEG-PLGA copolymers are widely utilized in controlled drug delivery. These nanoparticles can be engineered to release drugs in response to pH, redox potential, or enzymatic activity, matching the microenvironment of target organelles (Danhier et al., 2012). Their versatility allows incorporation of mitochondrial or peroxisomal targeting sequences and the delivery of nucleic acids, enzyme replacements, and small molecules for sustained therapeutic effects in disorders like MELAS or X-linked adrenoleukodystrophy (Xu et al., 2016).
C. Inorganic Nanoparticles
Gold nanoparticles (AuNPs) and mesoporous silica nanoparticles (MSNs) offer unique optical and structural properties, respectively, suitable for theranostic applications. Their surfaces can be functionalized with organelle-specific ligands and imaging agents, enabling simultaneous drug delivery and diagnostic tracking (Wang et al., 2014). AuNPs conjugated with mitochondrial targeting sequences have been employed to deliver apoptosis modulators in neurodegenerative models, while MSNs loaded with enzymes or gene-editing tools are under investigation for correcting metabolic defects in peroxisomal disorders (Chen et al., 2013).
D. Hybrid and Biomimetic Nanocarriers
Hybrid nanocarriers combine organic and inorganic elements for enhanced functionality. Biomimetic nanocarriers, such as cell membrane-coated nanoparticles and exosome-based systems, mimic native cellular interactions and evade immune clearance (Fang et al., 2018). Exosomes, being naturally secreted vesicles, possess intrinsic targeting capabilities and have shown potential in organelle-specific delivery by surface engineering with peptides or aptamers (El Andaloussi et al., 2013). In preclinical studies, exosome-loaded antioxidant enzymes and cell membrane-camouflaged nanoparticles have successfully targeted dysfunctional mitochondria and peroxisomes, indicating translational potential for rare genetic diseases (Jiang et al., 2020).
Table 3. Nanocarrier Platforms for Organelle-Specific Therapeutics
Nanocarrier Type |
Key Features |
Organelle Targeting Mechanism |
Application in Rare Disorders |
Liposomes |
Biocompatible, lipid bilayer |
Ligand-modified surface (e.g., TPP?, peptides) |
Mitochondrial and peroxisomal enzyme delivery |
Dendrimers |
Highly branched, modifiable surface |
Multivalent ligand conjugation |
Antioxidant delivery in mitochondrial diseases |
PLGA/PEG-PLGA NPs |
Biodegradable, controlled release |
pH/redox-sensitive linkers with targeting moieties |
Drug/gene therapy in metabolic syndromes |
Gold & Silica NPs |
Optical properties, high surface area |
Surface functionalization with targeting peptides |
Theranostics in organelle dysfunction |
Cell Membrane-Coated NPs |
Immune evasion, prolonged circulation |
Membrane-mediated homotypic targeting |
Personalized nanotherapy in peroxisomal disorders |
Exosome-Based Nanocarriers |
Natural origin, inherent targeting |
Engineered for organelle-specific ligands |
Enzyme/gene delivery in inherited disorders |
V. Organelle-Targeted Therapeutics: Preclinical and Clinical Advances
Organelle-specific therapeutics are progressing from bench to bedside with promising preclinical data and emerging clinical trials targeting mitochondrial and peroxisomal dysfunctions. However, challenges remain in translation due to biological complexity, delivery limitations, and regulatory hurdles.
A. Experimental Models for Evaluation
Robust and representative disease models are essential to assess the efficacy and organelle-targeting precision of nanotherapeutics. In vitro models include immortalized cell lines and CRISPR-engineered cells harboring mitochondrial or peroxisomal mutations. Patient-derived organoids offer 3D structural fidelity and genotype-specific drug response prediction (Lancaster & Knoblich, 2014). In vivo models such as transgenic mice and zebrafish are commonly used for evaluating biodistribution, pharmacokinetics, and therapeutic index in systemic disorders like Leigh syndrome or X-linked adrenoleukodystrophy (Meyer et al., 2017).
B. Notable Preclinical Studies
Several organelle-targeted therapeutics have demonstrated efficacy in preclinical models:
These findings support the translational potential of subcellular delivery platforms in rare metabolic diseases.
C. Clinical Trials and Translational Gaps
Some organelle-targeted compounds have entered clinical evaluation, especially in the context of mitochondrial diseases:
However, the translation of peroxisome-targeted therapies remains limited due to:
Bridging these gaps necessitates integrative approaches combining omics, high-throughput screening, and regulatory innovations.
Table 4. Organelle-Targeted Therapeutics: Preclinical and Clinical Progress
Therapeutic Type |
Agent/Platform |
Target Organelle |
Disease Model |
Outcome Highlights |
Stage |
Mitochondrial Antioxidant |
MitoQ, SkQ1 |
Mitochondria |
Rodent models, neuronal cells |
Reduced ROS, improved bioenergetics |
Preclinical/Phase II |
Peptide Therapy |
Elamipretide (SS-31) |
Mitochondria |
Barth syndrome, PMM |
Increased ATP, cardiac function |
Phase II/III |
Nucleic Acid Delivery |
mtDNA/siRNA nanocarriers |
Mitochondria/Peroxisome |
Cellular and murine models |
Enzyme restoration, metabolic correction |
Preclinical |
Organoid Drug Testing |
Exosome-loaded MitoQ |
Mitochondria |
Patient-derived organoids |
Enhanced drug response fidelity |
Preclinical |
VI. Challenges and Considerations
Despite the promising advancements in organelle-specific nanotherapeutics for mitochondrial and peroxisomal disorders, numerous scientific, clinical, and regulatory hurdles must be overcome for successful translation. This section highlights key obstacles in design, delivery, safety, scalability, and policy implementation.
A. Barriers to Intracellular and Organelle Delivery
One of the foremost challenges in nanotherapeutic delivery is overcoming the cellular membrane barrier and achieving endosomal escape. After cellular uptake—primarily via clathrin-mediated or caveolae-dependent endocytosis—nanocarriers often get entrapped in endosomes, leading to lysosomal degradation of the payload (Varkouhi et al., 2011). Further, targeting subcellular organelles like mitochondria and peroxisomes necessitates precise navigation through the cytoplasmic space, often hindered by cytoskeletal confinement and organelle crowding (Yamashita et al., 2020).
B. Off-Target Effects and Safety Concerns
The use of cationic lipids, dendrimers, or metal-based nanoparticles often poses risks of cytotoxicity, inflammatory responses, and immunogenicity. For example, excessive mitochondrial accumulation of TPP?-linked drugs can disrupt membrane potential and mitochondrial integrity (Mukhopadhyay et al., 2022). Moreover, unintended interactions with non-target cells or organelles could exacerbate off-target effects, especially in systemic delivery systems. Long-term safety and biodistribution profiling remain inadequately characterized for most organelle-targeting nanocarriers.
C. Manufacturing and Scalability
Translation from lab-scale to clinical-grade nanotherapeutics is complicated by batch-to-batch variability, low encapsulation efficiency, and difficulties in maintaining stability during storage and transport. For rare diseases, this is compounded by the high cost of GMP (Good Manufacturing Practice) production and the lack of large patient cohorts to justify investment (Ventola, 2017). Ensuring reproducibility and scalability while preserving functional targeting remains a major challenge for clinical translation.
D. Ethical and Regulatory Hurdles
The emerging field of personalized nanomedicine raises ethical considerations regarding equitable access, data privacy, and off-label applications. For rare diseases, regulatory pathways are complex, especially when organelle-targeted therapies lack conventional pharmacodynamic biomarkers or validated surrogate endpoints. Policies such as the Orphan Drug Act facilitate incentives but need refinement to accommodate the nanoscale and personalized nature of these interventions (Choudhury & Maiti, 2020).
VII. Future Perspectives and Emerging Trends
The landscape of organelle-specific drug delivery is evolving rapidly, driven by innovations in genome editing, synthetic biology, AI-based drug design, and the growing recognition of unmet needs in ultra-rare disorders. Future developments promise not only to overcome current limitations but also to redefine the precision nanotherapeutic paradigm.
A. CRISPR/Cas and Organelle Genome Editing
Emerging tools such as mitoCRISPR and DddA-derived cytosine base editors offer potential for correcting mitochondrial DNA (mtDNA) mutations directly within the organelle (Lee et al., 2021). For peroxisomes, although editing is less explored, CRISPR/Cas9 systems targeting nuclear-encoded peroxins may help restore peroxisomal biogenesis in Zellweger spectrum disorders.
B. Artificial Organelles and Organelle Engineering
Bioengineered artificial organelles, including peroxisome-mimicking nano-reactors and mitochondrial bioreactors, aim to compensate for dysfunctional organelles (van Oppen et al., 2020). These synthetic systems can be customized for drug catalysis, ROS detoxification, or ATP generation, offering a futuristic therapeutic niche.
C. Multi-organelle Targeting Strategies
Given the inter-organelle communication in metabolic disorders, there is growing interest in designing nanocarriers that simultaneously target mitochondria and peroxisomes (e.g., dual-ligand systems or fusion peptide-guided delivery). This multi-target paradigm can enhance therapeutic outcomes in disorders involving co-regulated pathways (Smith et al., 2023).
D. AI and Machine Learning in Drug Design
AI-driven platforms are revolutionizing target prediction, nanocarrier design, and delivery optimization. Deep learning algorithms now facilitate the prediction of organelle-targeting sequences and drug-biomolecule interactions (Gómez-Bombarelli et al., 2018). Such platforms can reduce time and cost for rare disease drug development.
E. Precision Nanotherapeutics in Ultra-Rare Diseases
With fewer than 1 in 50,000 individuals affected, ultra-rare mitochondrial and peroxisomal disorders are often overlooked in mainstream drug pipelines. Precision nanotherapeutics offer tailored interventions even with limited patient cohorts, especially when supported by n-of-1 trials and compassionate use frameworks (Nguyen & Isakov, 2022).
VIII. CONCLUSION
Organelle-specific drug delivery represents a frontier in nanomedicine, particularly for rare mitochondrial and peroxisomal disorders where systemic therapies have failed to yield substantial efficacy. This review has illustrated the pathophysiological context, targeting strategies, nanocarrier platforms, and the state of clinical translation while acknowledging existing challenges. Looking forward, the integration of CRISPR/Cas systems, synthetic organelles, and AI-driven design will play pivotal roles in transforming therapeutic outcomes. A coordinated push involving researchers, clinicians, regulatory agencies, and patient advocacy groups is essential to transition these innovations from bench to bedside.
REFERENCES
Devinder Kumar Maheshwari, Shankar Gavaroji, Tanmay Ghosh, Narendra Mandoria, Ankur Patel, Dimpal Jaydev Yesansure, Manoj Kumar Katual, Yash Srivastav, Raghav Dixit*, Organelle-Specific Drug Delivery Systems in the Treatment of Rare Mitochondrial and Peroxisomal Disorders: A Frontier in Precision Nanotherapeutics, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 5, 566-577. https://doi.org/10.5281/zenodo.15335812