aims to selectively deliver therapeutic agents to specific sites in the body. This approach minimizes systemic exposure and reduces side effects by exploiting differences between healthy and diseased tissues, such as overexpressed receptors or altered physiological conditions.

Targeted delivery improves therapeutic efficacy, reduces off-target effects, and enables the use of lower drug doses. Various nanocarriers, including liposomes, polymeric nanoparticles, and micelles, can be modified with ligands or antibodies to enhance targeting specificity and cellular uptake.

Principles of targeted drug delivery

  • Targeted drug delivery aims to selectively deliver therapeutic agents to specific sites in the body, minimizing systemic exposure and reducing side effects
  • Key principles include targeting specific cells or tissues, controlling , and improving and pharmacokinetics
  • Targeted delivery systems exploit differences between healthy and diseased tissues, such as overexpressed receptors or altered physiological conditions (pH, temperature)

Advantages vs traditional drug delivery

  • Targeted delivery improves therapeutic efficacy by increasing drug concentration at the desired site of action
  • Reduces off-target effects and systemic by minimizing drug exposure to healthy tissues
  • Enables the use of lower drug doses, potentially reducing the risk of adverse reactions and improving patient compliance
  • Allows for the delivery of poorly soluble or unstable drugs by encapsulating them in protective carriers

Passive vs active targeting strategies

Enhanced permeability and retention effect

Top images from around the web for Enhanced permeability and retention effect
Top images from around the web for Enhanced permeability and retention effect
  • relies on the enhanced permeability and retention (EPR) effect in tumor tissues
  • Tumor vasculature is leaky and poorly organized, allowing nanocarriers to accumulate in the tumor microenvironment
  • Lack of effective lymphatic drainage in tumors further promotes the retention of nanocarriers
  • EPR effect is influenced by factors such as tumor type, size, and location

Ligand-receptor interactions

  • involves the attachment of specific ligands to the surface of nanocarriers
  • Ligands bind to receptors overexpressed on the surface of target cells (transferrin receptor, folate receptor)
  • Ligand-receptor interactions facilitate cellular uptake and intracellular drug release
  • Examples of ligands include small molecules (folic acid), peptides (RGD), and aptamers

Antibody-antigen recognition

  • or antibody fragments can be conjugated to nanocarriers for active targeting
  • Antibodies recognize and bind to specific antigens expressed on the surface of target cells (HER2, EGFR)
  • Antibody-antigen interactions enable highly specific targeting and can trigger receptor-mediated endocytosis
  • Challenges include potential immunogenicity and high production costs

Nanocarriers for targeted delivery

Liposomes

  • Liposomes are spherical vesicles composed of a phospholipid bilayer enclosing an aqueous core
  • Can encapsulate both hydrophilic and hydrophobic drugs, protecting them from degradation
  • Surface modifications (, ) enhance circulation time and targeting specificity
  • Examples: Doxil (PEGylated liposomal doxorubicin), Onivyde (liposomal irinotecan)

Polymeric nanoparticles

  • Polymeric nanoparticles are solid colloidal particles made from biocompatible and biodegradable polymers (PLGA, PLA)
  • Drugs can be encapsulated, adsorbed, or conjugated to the polymer matrix
  • profiles can be achieved by adjusting polymer composition and particle size
  • Examples: Abraxane (albumin-bound paclitaxel), Genexol-PM (polymeric micelle formulation of paclitaxel)

Dendrimers

  • Dendrimers are highly branched, tree-like polymeric structures with a central core and multiple surface functional groups
  • Drugs can be encapsulated in the interior or conjugated to the surface groups
  • Precise control over size, shape, and surface functionality enables targeted delivery and enhanced solubility
  • Example: VivaGel (dendrimer-based vaginal microbicide for HIV prevention)

Micelles

  • Micelles are self-assembling nanostructures formed by amphiphilic block copolymers in aqueous environments
  • Hydrophobic drugs can be solubilized in the micelle core, while the hydrophilic shell provides and stealth properties
  • Responsive micelles can release drugs in response to specific stimuli (pH, temperature, enzymes)
  • Examples: Genexol-PM (polymeric micelle formulation of paclitaxel), NK105 (paclitaxel-loaded polymeric micelles)

Surface modifications of nanocarriers

PEGylation

  • PEGylation involves the attachment of polyethylene glycol (PEG) chains to the surface of nanocarriers
  • Creates a hydrophilic barrier that reduces protein adsorption and prevents opsonization and clearance by the mononuclear phagocyte system
  • Prolongs circulation time and improves the pharmacokinetic profile of nanocarriers
  • May hinder cellular uptake and drug release, requiring optimization of PEG density and chain length

Ligand conjugation

  • Ligands such as small molecules, peptides, or aptamers can be conjugated to the surface of nanocarriers
  • Enables active targeting by binding to specific receptors overexpressed on target cells
  • Ligand density, orientation, and spacing influence targeting efficiency and cellular uptake
  • Examples: folate-conjugated liposomes, RGD-conjugated polymeric nanoparticles

Antibody attachment

  • Monoclonal antibodies or antibody fragments can be attached to the surface of nanocarriers
  • Provides highly specific targeting through antibody-antigen recognition
  • Can trigger receptor-mediated endocytosis and enhance intracellular drug delivery
  • Challenges include potential immunogenicity, high production costs, and stability issues

Challenges in targeted drug delivery

Biocompatibility and toxicity

  • Nanocarriers must be biocompatible and non-toxic to minimize adverse effects
  • Potential toxicity may arise from the nanocarrier material, surface modifications, or the drug itself
  • Long-term safety and biodegradability of nanocarriers need to be thoroughly evaluated
  • Interactions with the immune system and the potential for immunogenicity must be considered

Stability and drug release

  • Nanocarriers should maintain their integrity and protect the encapsulated drug during circulation
  • Premature drug release can lead to reduced efficacy and increased systemic toxicity
  • Controlled and triggered drug release mechanisms are essential for optimal therapeutic outcomes
  • Stability during storage and handling must be ensured for clinical translation

Manufacturing and scale-up

  • Reproducible and scalable manufacturing processes are crucial for the clinical translation of targeted delivery systems
  • Batch-to-batch variability and quality control issues can hinder large-scale production
  • Sterilization methods must be compatible with the nanocarrier properties and drug stability
  • Regulatory challenges and the need for standardized characterization methods can delay commercialization

Clinical applications of targeted therapies

Cancer treatment

  • Targeted delivery systems can improve the efficacy and safety of anticancer drugs
  • Examples: Doxil (PEGylated liposomal doxorubicin) for ovarian cancer, Kadcyla (antibody-drug conjugate) for HER2-positive breast cancer
  • Nanocarriers can overcome drug resistance mechanisms and enhance tumor penetration
  • Combination therapies with targeted delivery systems can exploit synergistic effects and improve treatment outcomes

Cardiovascular diseases

  • Targeted delivery can improve the treatment of atherosclerosis, thrombosis, and other cardiovascular conditions
  • Examples: liposomal alendronate for atherosclerotic plaque targeting, targeted delivery of thrombolytic agents for thrombosis
  • Nanocarriers can deliver drugs to specific regions of the cardiovascular system, such as the endothelium or injured vasculature
  • Targeted delivery can reduce systemic side effects associated with cardiovascular drugs

Neurodegenerative disorders

  • Targeted delivery systems can overcome the blood-brain barrier and deliver drugs to the central nervous system
  • Examples: targeted delivery of growth factors for Alzheimer's disease, nanocarriers for the delivery of siRNA in Parkinson's disease
  • Nanocarriers can target specific cell types (neurons, glial cells) or pathological features (amyloid plaques, Lewy bodies)
  • Targeted delivery can minimize systemic exposure and reduce the risk of neurotoxicity

Inflammatory conditions

  • Targeted delivery can improve the treatment of inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis
  • Examples: targeted delivery of anti-inflammatory drugs to the joints in rheumatoid arthritis, targeted delivery of siRNA for inflammatory bowel disease
  • Nanocarriers can target inflamed tissues or specific immune cells involved in the inflammatory process
  • Targeted delivery can reduce the systemic side effects associated with long-term use of anti-inflammatory drugs

Future perspectives in targeted delivery

Personalized medicine

  • Targeted delivery systems can be tailored to individual patient characteristics, such as genetic profile or disease subtype
  • Personalized approaches can improve therapeutic efficacy and minimize adverse effects
  • Companion diagnostics can help identify patients most likely to benefit from targeted therapies
  • Integration of omics data and advanced imaging techniques can guide the design and selection of personalized targeted delivery systems

Combination therapies

  • Targeted delivery systems can be used in combination with other therapeutic modalities, such as chemotherapy, radiotherapy, or immunotherapy
  • Combination therapies can exploit synergistic effects and overcome drug resistance mechanisms
  • Nanocarriers can co-deliver multiple drugs with different mechanisms of action for enhanced therapeutic outcomes
  • Rational design of combination therapies based on the understanding of disease biology and drug interactions is crucial

Theranostic approaches

  • Theranostic nanocarriers combine diagnostic and therapeutic functions in a single platform
  • Imaging modalities (MRI, PET, SPECT) can be used to monitor the biodistribution and target accumulation of nanocarriers
  • Real-time monitoring of treatment response can guide dosing and treatment decisions
  • Theranostic approaches can facilitate the development of personalized and adaptive treatment strategies
  • Integration of advanced imaging techniques and responsive drug release mechanisms can further enhance the potential of theranostic nanocarriers

Key Terms to Review (27)

Active targeting: Active targeting refers to the deliberate and strategic approach of directing therapeutic agents specifically to desired cells or tissues, enhancing the effectiveness of drug delivery while minimizing side effects. This method often involves the use of targeting ligands, such as antibodies or small molecules, that recognize specific biomarkers present on the surface of target cells, facilitating selective binding and uptake.
Antibody attachment: Antibody attachment refers to the process where antibodies, which are specialized proteins produced by the immune system, bind to specific antigens on pathogens or infected cells. This binding is crucial for targeted drug delivery, as it allows therapeutic agents to be delivered precisely to diseased cells while sparing healthy tissues, enhancing the efficacy and safety of treatments.
Antibody-drug conjugates: Antibody-drug conjugates (ADCs) are targeted cancer therapies that combine an antibody with a cytotoxic drug, allowing for the selective delivery of the drug to cancer cells while sparing healthy tissues. By attaching the drug to an antibody that specifically binds to tumor-associated antigens, ADCs enhance the efficacy of treatment and reduce systemic toxicity, representing a significant advancement in targeted drug delivery.
Bioavailability: Bioavailability refers to the proportion of a drug or substance that enters the systemic circulation when it is introduced into the body, making it available for therapeutic effect. This concept is crucial because it influences how effectively a drug performs in its intended role, impacting factors like dose-response relationships and absorption rates.
Biocompatibility: Biocompatibility refers to the ability of a material or substance to perform with an appropriate host response in a specific application, particularly in medical settings. It is crucial for ensuring that medical devices, drug delivery systems, and implants interact safely and effectively with biological tissues. This includes not causing an adverse immune reaction, promoting healing, and functioning properly within the body.
Clinical trials: Clinical trials are research studies conducted to evaluate the effectiveness and safety of new drugs or treatment protocols in humans. They are essential for determining whether a new drug works as intended and for identifying any potential side effects. These trials often involve different phases, each designed to answer specific research questions, helping to ensure that only safe and effective therapies reach the market.
Controlled release: Controlled release refers to a drug delivery system designed to release a medication at a predetermined rate, typically over an extended period. This method enhances therapeutic effectiveness by maintaining consistent drug levels in the bloodstream, reducing side effects, and improving patient compliance. It plays a significant role in optimizing the delivery of drugs, particularly in targeted therapies where precise dosage is crucial for maximizing treatment outcomes.
Distribution: Distribution refers to the process by which a drug is dispersed throughout the body's fluids and tissues after administration. It involves understanding how factors like blood flow, tissue permeability, and the binding of drugs to proteins influence the extent and rate at which a drug reaches its target sites, impacting efficacy and safety.
Drug release: Drug release refers to the process by which a pharmaceutical compound is liberated from its formulation and becomes available for absorption into the body. This process is crucial in determining the onset, intensity, and duration of a drug's therapeutic effect, as it directly influences how quickly and effectively a drug can exert its action at the target site. Effective drug release mechanisms are essential for optimizing the efficacy of targeted drug delivery systems, ensuring that the therapeutic agents reach their intended location in the body in a controlled manner.
Enhanced permeability and retention effect: The enhanced permeability and retention (EPR) effect refers to the tendency of certain sizes of nanoparticles and macromolecules to preferentially accumulate in tumor tissue due to its leaky vasculature and impaired lymphatic drainage. This phenomenon is crucial in the context of drug delivery, as it allows for targeted therapy, increasing the effectiveness of treatments while reducing side effects.
FDA Approval: FDA approval is the process by which the U.S. Food and Drug Administration evaluates and authorizes new drugs and medical devices for public use. This rigorous process ensures that products meet safety, efficacy, and quality standards before they can be marketed. Approval is essential for antibiotics, central nervous system drugs, and targeted drug delivery systems as it assures healthcare providers and patients of their reliability and effectiveness in treating various conditions.
Intranasal Delivery: Intranasal delivery refers to the administration of drugs through the nasal cavity for systemic or local effect. This route bypasses first-pass metabolism and allows for rapid absorption into the bloodstream, making it particularly useful for drugs that require quick onset of action. The nasal mucosa has a rich blood supply, which facilitates efficient drug absorption and delivery directly to the central nervous system in some cases.
Ligand conjugation: Ligand conjugation refers to the process of chemically linking a ligand, which is a molecule that binds to another molecule, typically a receptor or protein, to a drug or delivery system. This technique enhances the specificity and effectiveness of targeted drug delivery by ensuring that the therapeutic agent reaches its intended site of action, minimizing off-target effects. By modifying the properties of the ligand, such as increasing its binding affinity or altering its pharmacokinetics, ligand conjugation can significantly improve the therapeutic index of a drug.
Ligand-receptor binding: Ligand-receptor binding refers to the specific interaction between a ligand, which can be a small molecule or a biomolecule, and a receptor, typically a protein, on the surface of a cell. This binding is crucial for triggering various biological responses, such as signal transduction and cellular communication, which play a key role in the efficacy of targeted drug delivery systems that aim to direct therapeutic agents to specific sites in the body.
Liposomal delivery: Liposomal delivery is a drug delivery system that uses liposomes, which are spherical vesicles composed of lipid bilayers, to encapsulate and transport therapeutic agents. This method enhances the bioavailability and stability of drugs while also providing a means for targeted delivery to specific tissues or cells, minimizing side effects and improving therapeutic efficacy.
Metabolism: Metabolism refers to the complex set of biochemical reactions that occur within living organisms to maintain life, including the conversion of food into energy and the synthesis of necessary compounds. It plays a crucial role in drug development and pharmacology, influencing how drugs are processed in the body, their efficacy, and potential side effects, particularly in the context of specific therapeutic areas and drug design.
Microencapsulation: Microencapsulation is a process that involves enclosing tiny particles or droplets within a coating material to create small capsules. This technique is commonly used in drug delivery systems to enhance the stability and controlled release of active pharmaceutical ingredients, allowing for targeted delivery to specific areas within the body. By improving solubility, bioavailability, and protecting sensitive compounds, microencapsulation plays a crucial role in optimizing therapeutic efficacy.
Monoclonal antibodies: Monoclonal antibodies are lab-made molecules engineered to bind to specific targets, often proteins, on cells or pathogens. These antibodies are produced by identical immune cells that are clones of a unique parent cell, allowing for precise targeting in various therapeutic applications, especially in targeted drug delivery systems where they can deliver drugs directly to diseased cells while minimizing effects on healthy tissue.
Nanoparticle delivery: Nanoparticle delivery refers to the use of nanoscale materials to transport drugs or therapeutic agents directly to targeted sites in the body. This approach enhances the efficacy of treatments by improving drug solubility, stability, and bioavailability while minimizing side effects, making it a critical component of modern medicine and targeted therapies.
Passive targeting: Passive targeting refers to a drug delivery strategy where therapeutic agents accumulate in specific tissues or tumors primarily due to the physiological and biological characteristics of those tissues, rather than through active mechanisms like specific binding. This approach relies on the enhanced permeability and retention (EPR) effect, which allows larger molecules or nanoparticles to enter tumor tissues more easily than normal tissues.
Pegylation: Pegylation is the process of attaching polyethylene glycol (PEG) chains to molecules, such as drugs or proteins, to enhance their pharmacological properties. This modification can improve solubility, increase stability, and prolong circulation time in the bloodstream, which is crucial for targeted drug delivery systems that aim to enhance therapeutic efficacy while minimizing side effects.
Personalized medicine: Personalized medicine is a medical approach that tailors treatment and healthcare strategies to individual characteristics, including genetic makeup, lifestyle, and environment. This method aims to optimize the effectiveness of therapies by considering the unique biological differences among patients, particularly in the treatment of diseases like cancer. By leveraging advancements in genomics and molecular biology, personalized medicine enhances targeted drug delivery and improves outcomes for patients through more precise interventions.
Small molecule inhibitors: Small molecule inhibitors are low molecular weight compounds that can selectively inhibit the function of specific proteins, often involved in disease processes. These compounds play a crucial role in targeted drug delivery by binding to specific targets, such as enzymes or receptors, thereby blocking their activity and potentially altering disease progression. Their ability to penetrate cells easily allows for a focused therapeutic effect, enhancing the effectiveness of treatment while minimizing side effects.
Stability: Stability refers to the ability of a molecule or system to maintain its structure and function over time, resisting degradation or change under various conditions. In drug design and delivery, stability is crucial as it influences the effectiveness, safety, and shelf life of a drug. Factors such as chemical composition, environmental conditions, and interactions with other molecules all play a role in determining the stability of a drug formulation or delivery system.
Targeted drug delivery: Targeted drug delivery is a method of delivering medication directly to a specific site in the body to maximize therapeutic effects while minimizing side effects. This approach enhances the effectiveness of the drug by ensuring that it reaches the intended target, such as a tumor or inflamed tissue, thereby improving treatment outcomes and reducing damage to healthy tissues. The precision of targeted drug delivery can also facilitate controlled release, allowing for sustained drug action over time.
Theranostics: Theranostics refers to a personalized approach in medicine that combines therapeutic and diagnostic capabilities, allowing for the simultaneous diagnosis and treatment of diseases, particularly cancer. This innovative concept integrates targeted drug delivery with imaging techniques to monitor treatment response, enabling tailored therapies based on individual patient profiles.
Toxicity: Toxicity refers to the degree to which a substance can harm living organisms. It encompasses various factors such as dose, exposure route, and the specific biological mechanisms that lead to harmful effects. Understanding toxicity is crucial in many fields, particularly in drug development, as it helps predict how compounds will behave in biological systems and informs safety assessments.
© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.