Targeted drug delivery

Last updated

Targeted drug delivery, sometimes called smart drug delivery, [1] is a method of delivering medication to a patient in a manner that increases the concentration of the medication in some parts of the body relative to others. This means of delivery is largely founded on nanomedicine, which plans to employ nanoparticle-mediated drug delivery in order to combat the downfalls of conventional drug delivery. These nanoparticles would be loaded with drugs and targeted to specific parts of the body where there is solely diseased tissue, thereby avoiding interaction with healthy tissue. The goal of a targeted drug delivery system is to prolong, localize, target and have a protected drug interaction with the diseased tissue. The conventional drug delivery system is the absorption of the drug across a biological membrane, whereas the targeted release system releases the drug in a dosage form. The advantages to the targeted release system is the reduction in the frequency of the dosages taken by the patient, having a more uniform effect of the drug, reduction of drug side-effects, and reduced fluctuation in circulating drug levels. The disadvantage of the system is high cost, which makes productivity more difficult, and the reduced ability to adjust the dosages.

Contents

Targeted drug delivery systems have been developed to optimize regenerative techniques. The system is based on a method that delivers a certain amount of a therapeutic agent for a prolonged period of time to a targeted diseased area within the body. This helps maintain the required plasma and tissue drug levels in the body, thereby preventing any damage to the healthy tissue via the drug. The drug delivery system is highly integrated and requires various disciplines, such as chemists, biologists, and engineers, to join forces to optimize this system. [2]

Background

In traditional drug delivery systems such as oral ingestion or intravascular injection, the medication is distributed throughout the body through the systemic blood circulation. For most therapeutic agents, only a small portion of the medication reaches the organ to be affected, such as in chemotherapy where roughly 99% of the drugs administered do not reach the tumor site. [3] Targeted drug delivery seeks to concentrate the medication in the tissues of interest while reducing the relative concentration of the medication in the remaining tissues. For example, by avoiding the host's defense mechanisms and inhibiting non-specific distribution in the liver and spleen, [4] a system can reach the intended site of action in higher concentrations. Targeted delivery is believed to improve efficacy while reducing side-effects.

When implementing a targeted release system, the following design criteria for the system must be taken into account: the drug properties, side-effects of the drugs, the route taken for the delivery of the drug, the targeted site, and the disease.

Increasing developments to novel treatments requires a controlled microenvironment that is accomplished only through the implementation of therapeutic agents whose side-effects can be avoided with targeted drug delivery. Advances in the field of targeted drug delivery to cardiac tissue will be an integral component to regenerate cardiac tissue. [5]

There are two kinds of targeted drug delivery: active targeted drug delivery, such as some antibody medications, and passive targeted drug delivery, such as the enhanced permeability and retention effect (EPR-effect).

Targeting methods

This ability for nanoparticles to concentrate in areas of solely diseased tissue is accomplished through either one or both means of targeting: passive or active.

Passive targeting

Passive targeting is achieved by incorporating the therapeutic agent into a macromolecule or nanoparticle that passively reaches the target organ. In passive targeting, the drug's success is directly related to circulation time. [6] This is achieved by cloaking the nanoparticle with some sort of coating. Several substances can achieve this, with one of them being polyethylene glycol (PEG). By adding PEG to the surface of the nanoparticle, it is rendered hydrophilic, thus allowing water molecules to bind to the oxygen molecules on PEG via hydrogen bonding. The result of this bond is a film of hydration around the nanoparticle which makes the substance antiphagocytic. The particles obtain this property due to the hydrophobic interactions that are natural to the reticuloendothelial system (RES), thus the drug-loaded nanoparticle is able to stay in circulation for a longer period of time. [7] To work in conjunction with this mechanism of passive targeting, nanoparticles that are between 10 and 100 nanometers in size have been found to circulate systemically for longer periods of time. [8]

Active targeting

Active targeting of drug-loaded nanoparticles enhances the effects of passive targeting to make the nanoparticle more specific to a target site. There are several ways that active targeting can be accomplished. One way to actively target solely diseased tissue in the body is to know the nature of a receptor on the cell for which the drug will be targeted to. [9] Researchers can then utilize cell-specific ligands that will allow the nanoparticle to bind specifically to the cell that has the complementary receptor. This form of active targeting was found to be successful when utilizing transferrin as the cell-specific ligand. [9] The transferrin was conjugated to the nanoparticle to target tumor cells that possess transferrin-receptor mediated endocytosis mechanisms on their membrane. This means of targeting was found to increase uptake, as opposed to non-conjugated nanoparticles. Another cell-specific ligand is the RGD motif which binds to the integrin αvβ3. [10] This integrin is upregulated in tumor and activated endothelial cells. [11] Conjugation of RGD to chemotherapeutic-loaded nanoparticles has been shown to increase cancer cell uptake in vitro and therapeutic efficacy in vivo. [10]

Active targeting can also be achieved by utilizing magnetoliposomes, which usually serves as a contrast agent in magnetic resonance imaging. [9] Thus, by grafting these liposomes with a desired drug to deliver to a region of the body, magnetic positioning could aid with this process.

Furthermore, a nanoparticle could possess the capability to be activated by a trigger that is specific to the target site, such as utilizing materials that are pH responsive. [9] Most of the body has a consistent, neutral pH. However, some areas of the body are naturally more acidic than others, and, thus, nanoparticles can take advantage of this ability by releasing the drug when it encounters a specific pH. [9] Another specific triggering mechanism is based on the redox potential. One of the side effects of tumors is hypoxia, which alters the redox potential in the vicinity of the tumor. By modifying the redox potential that triggers the payload release the vesicles can be selective to different types of tumors. [12]

By utilizing both passive and active targeting, a drug-loaded nanoparticle has a heightened advantage over a conventional drug. It is able to circulate throughout the body for an extended period of time until it is successfully attracted to its target through the use of cell-specific ligands, magnetic positioning, or pH responsive materials. Because of these advantages, side effects from conventional drugs will be largely reduced as a result of the drug-loaded nanoparticles affecting only diseased tissue. [13] However, an emerging field known as nanotoxicology has concerns that the nanoparticles themselves could pose a threat to both the environment and human health with side effects of their own. [14] Active targeting can also be achieved through peptide based drug targeting system. [15]

Delivery vehicles

There are different types of drug delivery vehicles, such as polymeric micelles, liposomes, lipoprotein-based drug carriers, nano-particle drug carriers, dendrimers, etc. An ideal drug delivery vehicle must be non-toxic, biocompatible, non-immunogenic, biodegradable, [5] and must avoid recognition by the host's defense mechanisms[3].

Peptides

Cell Surface Peptides provide one way to introduce drug delivery into a target cell. [16] This method is accomplished by the peptide binding to a target cells surface receptors, in a way that bypasses immune defenses that would otherwise compromise a slower delivery, without causing harm to the host. In particular, peptides, such as intercellular adhesion molecule-1, have shown a great deal of binding ability in a target cell. This method has shown a degree of efficacy in treating both autoimmune diseases as well as forms of cancer as a result of this binding affinity. [17] Peptide mediated delivery is also of promise due to the low cost of creating the peptides as well as the simplicity of their structure.

Liposomes

Liposomes are composite structures made of phospholipids and may contain small amounts of other molecules. Though liposomes can vary in size from low micrometer range to tens of micrometers, unilamellar liposomes, as pictured here, are typically in the lower size range, with various targeting ligands attached to their surface, allowing for their surface-attachment and accumulation in pathological areas for treatment of disease. Liposome.jpg
Liposomes are composite structures made of phospholipids and may contain small amounts of other molecules. Though liposomes can vary in size from low micrometer range to tens of micrometers, unilamellar liposomes, as pictured here, are typically in the lower size range, with various targeting ligands attached to their surface, allowing for their surface-attachment and accumulation in pathological areas for treatment of disease.

The most common vehicle currently used for targeted drug delivery is the liposome. [19] Liposomes are non-toxic, non-hemolytic, and non-immunogenic even upon repeated injections; they are biocompatible and biodegradable and can be designed to avoid clearance mechanisms (reticuloendothelial system (RES), renal clearance, chemical or enzymatic inactivation, etc.) [20] [21] Lipid-based, ligand-coated nanocarriers can store their payload in the hydrophobic shell or the hydrophilic interior depending on the nature of the drug/contrast agent being carried. [5]

The only problem to using liposomes in vivo is their immediate uptake and clearance by the RES system and their relatively low stability in vitro. To combat this, polyethylene glycol (PEG) can be added to the surface of the liposomes. Increasing the mole percent of PEG on the surface of the liposomes by 4-10% significantly increased circulation time in vivo from 200 to 1000 minutes. [5]

PEGylation of the liposomal nanocarrier elongates the half-life of the construct while maintaining the passive targeting mechanism that is commonly conferred to lipid-based nanocarriers. [22] When used as a delivery system, the ability to induce instability in the construct is commonly exploited allowing the selective release of the encapsulated therapeutic agent in close proximity to the target tissue/cell in vivo. This nanocarrier system is commonly used in anti-cancer treatments as the acidity of the tumour mass caused by an over-reliance on glycolysis triggers drug release. [22] [23] [24]

Additional endogenous trigger pathways have been explored through the exploitation of inner and outer tumor environments, such as reactive oxygen species, glutathione, enzymes, hypoxia, and adenosine-5’- triphosphate (ATP), all of which are generally highly present in and around tumors. [25] External triggers are also used, such as light, low frequency ultrasound (LFUS), electrical fields, and magnetic fields. [26] In specific, LFUS has demonstrated high efficacy in the controlled trigger of various drugs in mice, such as cisplatin and calcein. [27] [28]

Micelles and dendrimers

Another type of drug delivery vehicle used is polymeric micelles. They are prepared from certain amphiphilic co-polymers consisting of both hydrophilic and hydrophobic monomer units. [2] They can be used to carry drugs that have poor solubility. This method offers little in the terms of size control or function malleability. Techniques that utilize reactive polymers along with a hydrophobic additive to produce a larger micelle that create a range of sizes have been developed. [29]

Dendrimers are also polymer-based delivery vehicles. They have a core that branches out in regular intervals to form a small, spherical, and very dense nanocarrier. [30]

Biodegradable particles

Biodegradable particles have the ability to target diseased tissue as well as deliver their payload as a controlled-release therapy. [31] Biodegradable particles bearing ligands to P-selectin, endothelial selectin (E-selectin) and ICAM-1 have been found to adhere to inflamed endothelium. [32] Therefore, the use of biodegradable particles can also be used for cardiac tissue.

Microalgae-based delivery

There are biocompatible microalgae hybrid microrobots for active drug-delivery in the lungs and the gastrointestinal tract. The microrobots proved effective in tests with mice. In the two studies, "Fluorescent dye or cell membrane–coated nanoparticle functionalized algae motors were further embedded inside a pH-sensitive capsule" and "antibiotic-loaded neutrophil membrane-coated polymeric nanoparticles [were attached] to natural microalgae". [33] [34] [35]

Artificial DNA nanostructures

The success of DNA nanotechnology in constructing artificially designed nanostructures out of nucleic acids such as DNA, combined with the demonstration of systems for DNA computing, has led to speculation that artificial nucleic acid nanodevices can be used to target drug delivery based upon directly sensing its environment. These methods make use of DNA solely as a structural material and a chemical, and do not make use of its biological role as the carrier of genetic information. Nucleic acid logic circuits that could potentially be used as the core of a system that releases a drug only in response to a stimulus such as a specific mRNA have been demonstrated. [36] In addition, a DNA "box" with a controllable lid has been synthesized using the DNA origami method. This structure could encapsulate a drug in its closed state, and open to release it only in response to a desired stimulus. [37]

Applications

Targeted drug delivery can be used to treat many diseases, such as the cardiovascular diseases and diabetes. However, the most important application of targeted drug delivery is to treat cancerous tumors. In doing so, the passive method of targeting tumors takes advantage of the enhanced permeability and retention (EPR) effect. This is a situation specific to tumors that results from rapidly forming blood vessels and poor lymphatic drainage. When the blood vessels form so rapidly, large fenestrae result that are 100 to 600 nanometers in size, which allows enhanced nanoparticle entry. Further, the poor lymphatic drainage means that the large influx of nanoparticles are rarely leaving, thus, the tumor retains more nanoparticles for successful treatment to take place. [8]

The American Heart Association rates cardiovascular disease as the number one cause of death in the United States. Each year 1.5 million myocardial infarctions (MI), also known as heart attacks, occur in the United States, with 500,000 leading to deaths. The costs related to heart attacks exceed $60 billion per year. Therefore, there is a need to come up with an optimum recovery system. The key to solving this problem lies in the effective use of pharmaceutical drugs that can be targeted directly to the diseased tissue. This technique can help develop many more regenerative techniques to cure various diseases. The development of a number of regenerative strategies in recent years for curing heart disease represents a paradigm shift away from conventional approaches that aim to manage heart disease. [5]

Stem cell therapy can be used to help regenerate myocardium tissue and return the contractile function of the heart by creating/supporting a microenvironment before the MI. Developments in targeted drug delivery to tumors have provided the groundwork for the burgeoning field of targeted drug delivery to cardiac tissue. [5] Recent developments have shown that there are different endothelial surfaces in tumors, which has led to the concept of endothelial cell adhesion molecule-mediated targeted drug delivery to tumors.

Liposomes can be used as drug delivery for the treatment of tuberculosis. The traditional treatment for TB is skin to chemotherapy which is not overly effective, which may be due to the failure of chemotherapy to make a high enough concentration at the infection site. The liposome delivery system allows for better microphage penetration and better builds a concentration at the infection site. [38] The delivery of the drugs works intravenously and by inhalation. Oral intake is not advised because the liposomes break down in the Gastrointestinal System.

3D printing is also used by doctors to investigate how to target cancerous tumors in a more efficient way. By printing a plastic 3D shape of the tumor and filling it with the drugs used in the treatment the flow of the liquid can be observed allowing the modification of the doses and targeting location of the drugs. [39]

See also

Related Research Articles

Nanomedicine is the medical application of nanotechnology. Nanomedicine ranges from the medical applications of nanomaterials and biological devices, to nanoelectronic biosensors, and even possible future applications of molecular nanotechnology such as biological machines. Current problems for nanomedicine involve understanding the issues related to toxicity and environmental impact of nanoscale materials.

<span class="mw-page-title-main">Drug delivery</span> Methods for delivering drugs to target sites

Drug delivery refers to approaches, formulations, manufacturing techniques, storage systems, and technologies involved in transporting a pharmaceutical compound to its target site to achieve a desired therapeutic effect. Principles related to drug preparation, route of administration, site-specific targeting, metabolism, and toxicity are used to optimize efficacy and safety, and to improve patient convenience and compliance. Drug delivery is aimed at altering a drug's pharmacokinetics and specificity by formulating it with different excipients, drug carriers, and medical devices. There is additional emphasis on increasing the bioavailability and duration of action of a drug to improve therapeutic outcomes. Some research has also been focused on improving safety for the person administering the medication. For example, several types of microneedle patches have been developed for administering vaccines and other medications to reduce the risk of needlestick injury.

<span class="mw-page-title-main">Chemotactic drug-targeting</span>

Targeted drug delivery is one of many ways researchers seek to improve drug delivery systems' overall efficacy, safety, and delivery. Within this medical field is a special reversal form of drug delivery called chemotactic drug targeting. By using chemical agents to help guide a drug carrier to a specific location within the body, this innovative approach seeks to improve precision and control during the drug delivery process, decrease the risk of toxicity, and potentially lower the required medical dosage needed. The general components of the conjugates are designed as follows: (i) carrier – regularly possessing promoter effect also on internalization into the cell; (ii) chemotactically active ligands acting on the target cells; (iii) drug to be delivered in a selective way and (iv) spacer sequence which joins drug molecule to the carrier and due to it enzyme labile moiety makes possible the intracellular compartment specific release of the drug. Careful selection of chemotactic component of the ligand not only the chemoattractant character could be expended, however, chemorepellent ligands are also valuable as they are useful to keep away cell populations degrading the conjugate containing the drug. In a larger sense, chemotactic drug-targeting has the potential to improve cancer, inflammation, and arthritis treatment by taking advantage of the difference in environment between the target site and its surroundings. Therefore, this Wikipedia article aims to provide a brief overview of chemotactic drug targeting, the principles behind the approach, possible limitations and advantages, and its application to cancer and inflammation.

<span class="mw-page-title-main">Arginylglycylaspartic acid</span> Chemical compound

Arginylglycylaspartic acid (RGD) is the most common peptide motif responsible for cell adhesion to the extracellular matrix (ECM), found in species ranging from Drosophila to humans. Cell adhesion proteins called integrins recognize and bind to this sequence, which is found within many matrix proteins, including fibronectin, fibrinogen, vitronectin, osteopontin, and several other adhesive extracellular matrix proteins. The discovery of RGD and elucidation of how RGD binds to integrins has led to the development of a number of drugs and diagnostics, while the peptide itself is used ubiquitously in bioengineering. Depending on the application and the integrin targeted, RGD can be chemically modified or replaced by a similar peptide which promotes cell adhesion.

Microbubbles are bubbles smaller than one hundredth of a millimetre in diameter, but larger than one micrometre. They have widespread application in industry, medicine, life science, and food technology. The composition of the bubble shell and filling material determine important design features such as buoyancy, crush strength, thermal conductivity, and acoustic properties.

<span class="mw-page-title-main">Nanocarrier</span>

A nanocarrier is nanomaterial being used as a transport module for another substance, such as a drug. Commonly used nanocarriers include micelles, polymers, carbon-based materials, liposomes and other substances. Nanocarriers are currently being studied for their use in drug delivery and their unique characteristics demonstrate potential use in chemotherapy. This class of materials was first reported by a team of researchers of University of Évora, Alentejo in early 1960's, and grew exponentially in relevance since then.

Nanoparticles for drug delivery to the brain is a method for transporting drug molecules across the blood–brain barrier (BBB) using nanoparticles. These drugs cross the BBB and deliver pharmaceuticals to the brain for therapeutic treatment of neurological disorders. These disorders include Parkinson's disease, Alzheimer's disease, schizophrenia, depression, and brain tumors. Part of the difficulty in finding cures for these central nervous system (CNS) disorders is that there is yet no truly efficient delivery method for drugs to cross the BBB. Antibiotics, antineoplastic agents, and a variety of CNS-active drugs, especially neuropeptides, are a few examples of molecules that cannot pass the BBB alone. With the aid of nanoparticle delivery systems, however, studies have shown that some drugs can now cross the BBB, and even exhibit lower toxicity and decrease adverse effects throughout the body. Toxicity is an important concept for pharmacology because high toxicity levels in the body could be detrimental to the patient by affecting other organs and disrupting their function. Further, the BBB is not the only physiological barrier for drug delivery to the brain. Other biological factors influence how drugs are transported throughout the body and how they target specific locations for action. Some of these pathophysiological factors include blood flow alterations, edema and increased intracranial pressure, metabolic perturbations, and altered gene expression and protein synthesis. Though there exist many obstacles that make developing a robust delivery system difficult, nanoparticles provide a promising mechanism for drug transport to the CNS.

Nanoparticle drug delivery systems are engineered technologies that use nanoparticles for the targeted delivery and controlled release of therapeutic agents. The modern form of a drug delivery system should minimize side-effects and reduce both dosage and dosage frequency. Recently, nanoparticles have aroused attention due to their potential application for effective drug delivery.

<span class="mw-page-title-main">Debra Auguste</span> American chemical engineer

Debra Auguste is an American chemical engineer and professor at Northeastern University in the department of chemical engineering. Auguste is dedicated to developing treatments for triple negative breast cancer, one of the most aggressive and fatal cancers that disproportionately affects African American women. Her lab characterizes biomarkers of triple negative breast cancer and develops novel biocompatible therapeutic technologies to target and destroy metastatic cancer cells. Auguste received the 2012 Presidential Early Career Award for Scientists and Engineers and in 2010 was named in the 50 Most Influential African-Americans in Technology. In 2020, Auguste became an Elected Fellow of the American Institute for Medical and Biological Engineering.

<span class="mw-page-title-main">Dextran drug delivery systems</span> Polymeric drug carrier

Dextran drug delivery systems involve the use of the natural glucose polymer dextran in applications as a prodrug, nanoparticle, microsphere, micelle, and hydrogel drug carrier in the field of targeted and controlled drug delivery. According to several in vitro and animal research studies, dextran carriers reduce off-site toxicity and improve local drug concentration at the target tissue site. This technology has significant implications as a potential strategy for delivering therapeutics to treat cancer, cardiovascular diseases, pulmonary diseases, bone diseases, liver diseases, colonic diseases, infections, and HIV.

Conventional drug delivery is limited by the inability to control dosing, target specific sites, and achieve targeted permeability. Traditional methods of delivering therapeutics to the body experience challenges in achieving and maintaining maximum therapeutic effect while avoiding the effects of drug toxicity. Many drugs that are delivered orally or parenterally do not include mechanisms for sustained release, and as a result they require higher and more frequent dosing to achieve any therapeutic effect for the patient. As a result, the field of drug delivery systems developed into a large focus area for pharmaceutical research to address these limitations and improve quality of care for patients. Within the broad field of drug delivery, the development of stimuli-responsive drug delivery systems has created the ability to tune drug delivery systems to achieve more controlled dosing and targeted specificity based on material response to exogenous and endogenous stimuli.

<span class="mw-page-title-main">Gated drug delivery systems</span> Method of controlled drug release

Gated drug delivery systems are a method of controlled drug release that center around the use of physical molecules that cover the pores of drug carriers until triggered for removal by an external stimulus. Gated drug delivery systems are a recent innovation in the field of drug delivery and pose as a promising candidate for future drug delivery systems that are effective at targeting certain sites without having leakages or off target effects in normal tissues. This new technology has the potential to be used in a variety of tissues over a wide range of disease states and has the added benefit of protecting healthy tissues and reducing systemic side effects.

<span class="mw-page-title-main">Reduction-sensitive nanoparticles</span> Drug delivery method

Reduction-sensitive nanoparticles (RSNP) consist of nanocarriers that are chemically responsive to reduction. Drug delivery systems using RSNP can be loaded with different drugs that are designed to be released within a concentrated reducing environment, such as the tumor-targeted microenvironment. Reduction-Sensitive Nanoparticles provide an efficient method of targeted drug delivery for the improved controlled release of medication within localized areas of the body.

Protein nanotechnology is a burgeoning field of research that integrates the diverse physicochemical properties of proteins with nanoscale technology. This field assimilated into pharmaceutical research to give rise to a new classification of nanoparticles termed protein nanoparticles (PNPs). PNPs garnered significant interest due to their favorable pharmacokinetic properties such as high biocompatibility, biodegradability, and low toxicity Together, these characteristics have the potential to overcome the challenges encountered with synthetic NPs drug delivery strategies. These existing challenges including low bioavailability, a slow excretion rate, high toxicity, and a costly manufacturing process, will open the door to considerable therapeutic advancements within oncology, theranostics, and clinical translational research.

pH-responsive tumor-targeted drug delivery is a specialized form of targeted drug delivery that utilizes nanoparticles to deliver therapeutic drugs directly to cancerous tumor tissue while minimizing its interaction with healthy tissue. Scientists have used drug delivery as a way to modify the pharmacokinetics and targeted action of a drug by combining it with various excipients, drug carriers, and medical devices. These drug delivery systems have been created to react to the pH environment of diseased or cancerous tissues, triggering structural and chemical changes within the drug delivery system. This form of targeted drug delivery is to localize drug delivery, prolongs the drug's effect, and protect the drug from being broken down or eliminated by the body before it reaches the tumor.

Intranasal drug delivery occurs when particles are inhaled into the nasal cavity and transported directly into the nervous system. Though pharmaceuticals can be injected into the nose, some concerns include injuries, infection, and safe disposal. Studies demonstrate improved patient compliance with inhalation. Treating brain diseases has been a challenge due to the blood brain barrier. Previous studies evaluated the efficacy of delivery therapeutics through intranasal route for brain diseases and mental health conditions. Intranasal administration is a potential route associated with high drug transfer from nose to brain and drug bioavailability.

<span class="mw-page-title-main">Ligand-targeted liposome</span> Ligand-targeted liposomes for use in medical applications

A ligand-targeted liposome (LTL) is a nanocarrier with specific ligands attached to its surface to enhance localization for targeted drug delivery. The targeting ability of LTLs enhances cellular localization and uptake of these liposomes for therapeutic or diagnostic purposes. LTLs have the potential to enhance drug delivery by decreasing peripheral systemic toxicity, increasing in vivo drug stability, enhancing cellular uptake, and increasing efficiency for chemotherapeutics and other applications. Liposomes are beneficial in therapeutic manufacturing because of low batch-to-batch variability, easy synthesis, favorable scalability, and strong biocompatibility. Ligand-targeting technology enhances liposomes by adding targeting properties for directed drug delivery.

<span class="mw-page-title-main">Artificial white blood cells</span> Alternative method of immunotherapy

Artificial white blood cells are typically membrane bound vesicles designed to mimic the immunomodulatory behavior of naturally produced leukocytes. While extensive research has been done with regards to artificial red blood cells and platelets for use in emergency blood transfusions, research into artificial white blood cells has been focused on increasing the immunogenic response within a host to treat cancer or deliver drugs in a more favorable fashion. While certain limitations have prevented leukocyte mimicking particles from becoming widely used and FDA approved, more research is being allocated to this area of synthetic blood which has the potential for producing a new form of treatment for cancer and other diseases.

Theranostics, also known as theragnostics, is an emerging field in precision medicine that combines diagnostic and therapeutic approaches to provide the potential for personalized treatment and real-time monitoring of the effectiveness of treatments. Improvements in imaging techniques and targeted therapies are the basis of the field of theranostics. When medical imaging is coupled with the development of novel radiotracers and contrast agents, theranostics may provide opportunities for precise diagnosis and targeted therapy.

Moein Moghimi is a British professor and researcher in the fields of nanomedicine, drug delivery and biomaterials. He is currently the professor of Pharmaceutics and Nanomedicine at the School of Pharmacy and the Translational and Clinical Research Institute at Newcastle University. He is also an adjoint professor at the Skaggs School of Pharmacy, University of Colorado Denver.

References

  1. Muller, R; Keck, C (2004). "Challenges and solutions for the delivery of biotech drugs – a review of drug nanocrystal technology and lipid nanoparticles". Journal of Biotechnology. 113 (1–3): 151–170. doi:10.1016/j.jbiotec.2004.06.007. PMID   15380654.
  2. 1 2 Saltzman, W. Mark; Torchilin, Vladimir P. (2008). "Drug delivery systems". AccessScience. McGraw-Hill Companies. doi:10.1036/1097-8542.757275.
  3. Trafton, A. Tumors Targeted Using Tiny Gold Particles. MIT Tech Talk. 2009, 53, 4–4.
  4. Bertrand N, Leroux JC.; Leroux (2011). "The journey of a drug carrier in the body: an anatomo-physiological perspective". Journal of Controlled Release. 161 (2): 152–63. doi:10.1016/j.jconrel.2011.09.098. PMID   22001607.
  5. 1 2 3 4 5 6 Scott, Robert C; Crabbe, Deborah; Krynska, Barbara; Ansari, Ramin; Kiani, Mohammad F (2008). "Aiming for the heart: targeted delivery of drugs to diseased cardiac tissue". Expert Opinion on Drug Delivery. 5 (4): 459–70. doi:10.1517/17425247.5.4.459. PMID   18426386. S2CID   71338475.
  6. Sagnella, S.; Drummond, C. Drug Delivery: A Nanomedicine Approach. Australian Biochemist. [Online] 2012, 43, 5–8, 20. The Australian Society for Biochemistry and Molecular Biology.
  7. Vlerken, L. E. V.; Vyas, T. K.; Amiji, M. M. Poly(Ethylene Glycol)-Modified Nanocarriers for Tumor-Targeted and Intracellular Delivery. Pharm. Res.2007, 24, 1405–1414.
  8. 1 2 Gullotti, E.; Yeo, Y. Extracellularly Activated Nanocarriers: A New Paradigm of Tumor Targeted Drug Delivery. Mol. Pharm., [Online] 2009, 6, 1041-1051. ACS Publications.
  9. 1 2 3 4 5 Galvin, P.; Thompson, D.; Ryan, K. B.; Mccarthy, A.; Moore, A. C.; Burke, C. S.; Dyson, M.; Maccraith, B. D.; Gun’Ko, Y. K.; Byrne, M. T.; Volkov, Y.; Keely, C.; Keehan, E.; Howe, M.; Duffy, C.; Macloughlin, R. Nanoparticle-Based Drug Delivery: Case Studies for Cancer and Cardiovascular Applications. Cell. Mol. Life Sci. [Online] 2011, 69, 389–404.
  10. 1 2 Alipour, Mohsen; Baneshi, Marzieh; Hosseinkhani, Saman; Mahmoudi, Reza; Jabari Arabzadeh, Ali; Akrami, Mohammad; Mehrzad, Jalil; Bardania, Hassan (April 2020). "Recent progress in biomedical applications of RGD-based ligand: From precise cancer theranostics to biomaterial engineering: A systematic review". Journal of Biomedical Materials Research. Part A. 108 (4): 839–850. doi:10.1002/jbm.a.36862. ISSN   1552-4965. PMID   31854488. S2CID   209417891.
  11. Liu, Jie; Yuan, Shuanghu; Wang, Linlin; Sun, Xindong; Hu, Xudong; Meng, Xue; Yu, Jinming (2019-01-10). "Diagnostic and Predictive Value of Using RGD PET/CT in Patients with Cancer: A Systematic Review and Meta-Analysis". BioMed Research International. 2019: e8534761. doi: 10.1155/2019/8534761 . ISSN   2314-6133. PMC   6348803 . PMID   30733968.
  12. Noyhouzer, Tomer; L’Homme, Chloé; Beaulieu, Isabelle; Mazurkiewicz, Stephanie; Kuss, Sabine; Kraatz, Heinz-Bernhard; Canesi, Sylvain; Mauzeroll, Janine (2016-05-03). "Ferrocene-Modified Phospholipid: An Innovative Precursor for Redox-Triggered Drug Delivery Vesicles Selective to Cancer Cells". Langmuir. 32 (17): 4169–4178. doi:10.1021/acs.langmuir.6b00511. ISSN   0743-7463. PMID   26987014.
  13. Mitra, A. K.; Kwatra, D.; Vadlapudi, A. D. Drug Delivery; Jones & Bartlett Learning: Burlington, Massachusetts, 2015.
  14. Jong, W. H. D.; Borm, P. J. A. Drug Delivery and Nanoparticles: Applications and Hazards. Int. J. Nanomedicine. [Online] 2008, 3, 133–149. The National Center for Biotechnology Information.
  15. He, X; Bonaparte, N; Kim, S; Acharya, B; Lee, JY; Chi, L; Lee, HJ; Paik, YK; Moon, PG; Baek, MC; Lee, EK; KIM, JH; KIM, IS; Lee, BH (2012). "Enhanced delivery of T cells to tumor after chemotherapy using membrane-anchored, apoptosis-targeted peptide". Journal of Controlled Release. 162 (6): 521–8. doi:10.1016/j.jconrel.2012.07.023. PMID   22824781.
  16. Majumdar, Sumit; Siahaan, Teruna J. (May 2012). "Peptide-mediated targeted drug delivery: TARGETED DRUG DELIVERY". Medicinal Research Reviews. 32 (3): 637–658. doi:10.1002/med.20225. PMID   20814957. S2CID   206251084.
  17. Alley, Stephen C; Okeley, Nicole M; Senter, Peter D (August 2010). "Antibody–drug conjugates: targeted drug delivery for cancer". Current Opinion in Chemical Biology. 14 (4): 529–537. doi:10.1016/j.cbpa.2010.06.170. PMID   20643572.
  18. Torchilin, VP “Multifunctional Nanocarriers.” Adv Drug Deliv Rev 2006 Dec; 58 (14): 1532-55 doi: 10.1016/j.addr.2006.09.009
  19. Cobleigh, M; Langmuir, VK; Sledge, GW; Miller, KD; Haney, L; Novotny, WF; Reimann, JD; Vassel, A (2003). "A phase I/II dose-escalation trial of bevacizumab in previously treated metastatic breast cancer". Seminars in Oncology. 30 (5 Suppl 16): 117–24. doi:10.1053/j.seminoncol.2003.08.013. PMID   14613032.
  20. Seidman, A.; Hudis, C; Pierri, MK; Shak, S; Paton, V; Ashby, M; Murphy, M; Stewart, SJ; Keefe, D (2002). "Cardiac Dysfunction in the Trastuzumab Clinical Trials Experience". Journal of Clinical Oncology. 20 (5): 1215–21. doi:10.1200/JCO.20.5.1215. PMID   11870163.
  21. Brufsky, Adam (2009). "Trastuzumab-Based Therapy for Patients With HER2-Positive Breast Cancer". American Journal of Clinical Oncology. 33 (2): 186–95. doi:10.1097/COC.0b013e318191bfb0. PMID   19675448. S2CID   41559516.
  22. 1 2 Lee, Jinhyun Hannah; Yeo, Yoon (2015-03-24). "Controlled drug release from pharmaceutical nanocarriers". Chemical Engineering Science. Pharmaceutical Particles and Processing. 125: 75–84. Bibcode:2015ChEnS.125...75L. doi:10.1016/j.ces.2014.08.046. PMC   4322773 . PMID   25684779.
  23. Cho, Kwangjae; Wang, Xu; Nie, Shuming; Chen, Zhuo Georgia; Shin, Dong M. (2008-03-01). "Therapeutic nanoparticles for drug delivery in cancer". Clinical Cancer Research. 14 (5): 1310–1316. doi:10.1158/1078-0432.CCR-07-1441. ISSN   1078-0432. PMID   18316549.
  24. Taléns-Visconti R, Díez-Sales O, de Julián-Ortiz JV, Nácher A (Apr 2022). "Nanoliposomes in Cancer Therapy: Marketed Products and Current Clinical Trials". International Journal of Molecular Sciences. 23 (8): 4249. doi: 10.3390/ijms23084249 . PMC   9030431 . PMID   35457065.
  25. Mo, Ran; Gu, Zhen (2016-06-01). "Tumor microenvironment and intracellular signal-activated nanomaterials for anticancer drug delivery". Materials Today. 19 (5): 274–283. doi: 10.1016/j.mattod.2015.11.025 . ISSN   1369-7021.
  26. Wang, Yanfei; Kohane, Daniel S. (2017-05-09). "External triggering and triggered targeting strategies for drug delivery". Nature Reviews Materials. 2 (6): 17020. Bibcode:2017NatRM...217020W. doi:10.1038/natrevmats.2017.20. ISSN   2058-8437.
  27. AlSawaftah, Nour M.; Awad, Nahid S.; Paul, Vinod; Kawak, Paul S.; Al-Sayah, Mohammad H.; Husseini, Ghaleb A. (2021-06-02). "Transferrin-modified liposomes triggered with ultrasound to treat HeLa cells". Scientific Reports. 11 (1): 11589. Bibcode:2021NatSR..1111589A. doi:10.1038/s41598-021-90349-6. ISSN   2045-2322. PMC   8172941 . PMID   34078930.
  28. Schroeder, Avi; Honen, Reuma; Turjeman, Keren; Gabizon, Alberto; Kost, Joseph; Barenholz, Yechezkel (2009-07-01). "Ultrasound triggered release of cisplatin from liposomes in murine tumors". Journal of Controlled Release. 137 (1): 63–68. doi:10.1016/j.jconrel.2009.03.007. ISSN   0168-3659. PMID   19303426.
  29. Macosko, Cristopher W. "Polymer Nanoparticles Improve Delivery of Compounds” University of Minnesota Office for Technology Commercialization. "Nanodelivery". Archived from the original on 2012-03-24.
  30. Pili, R.; Rosenthal, M. A.; Mainwaring, P. N.; Van Hazel, G.; Srinivas, S.; Dreicer, R.; Goel, S.; Leach, J.; et al. (2010). "Phase II Study on the Addition of ASA404 (Vadimezan; 5,6-Dimethylxanthenone-4-Acetic Acid) to Docetaxel in CRMPC". Clinical Cancer Research. 16 (10): 2906–14. doi:10.1158/1078-0432.CCR-09-3026. PMID   20460477. S2CID   477849.
  31. Homsi, J.; Simon, G. R.; Garrett, C. R.; Springett, G.; De Conti, R.; Chiappori, A. A.; Munster, P. N.; Burton, M. K.; et al. (2007). "Phase I Trial of Poly-L-Glutamate Camptothecin (CT-2106) Administered Weekly in Patients with Advanced Solid Malignancies". Clinical Cancer Research. 13 (19): 5855–61. doi: 10.1158/1078-0432.CCR-06-2821 . PMID   17908979.
  32. Vogel, V. G.; Costantino, JP; Wickerham, DL; Cronin, WM; Cecchini, RS; Atkins, JN; Bevers, TB; Fehrenbacher, L; et al. (2006). "Effects of Tamoxifen vs Raloxifene on the Risk of Developing Invasive Breast Cancer and Other Disease Outcomes: The NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 Trial". JAMA. 295 (23): 2727–41. doi:10.1001/jama.295.23.joc60074. PMID   16754727.
  33. "Algae micromotors join the ranks for targeted drug delivery". Chemical & Engineering News. Retrieved 19 October 2022.
  34. Zhang, Fangyu; Zhuang, Jia; Li, Zhengxing; Gong, Hua; de Ávila, Berta Esteban-Fernández; Duan, Yaou; Zhang, Qiangzhe; Zhou, Jiarong; Yin, Lu; Karshalev, Emil; Gao, Weiwei; Nizet, Victor; Fang, Ronnie H.; Zhang, Liangfang; Wang, Joseph (22 September 2022). "Nanoparticle-modified microrobots for in vivo antibiotic delivery to treat acute bacterial pneumonia". Nature Materials. 21 (11): 1324–1332. Bibcode:2022NatMa..21.1324Z. doi:10.1038/s41563-022-01360-9. ISSN   1476-4660. PMC   9633541 . PMID   36138145.
  35. Zhang, Fangyu; Li, Zhengxing; Duan, Yaou; Abbas, Amal; Mundaca-Uribe, Rodolfo; Yin, Lu; Luan, Hao; Gao, Weiwei; Fang, Ronnie H.; Zhang, Liangfang; Wang, Joseph (28 September 2022). "Gastrointestinal tract drug delivery using algae motors embedded in a degradable capsule". Science Robotics. 7 (70): eabo4160. doi:10.1126/scirobotics.abo4160. ISSN   2470-9476. PMC   9884493 . PMID   36170380. S2CID   252598190.
  36. Kahan, M; Gil, B; Adar, R; Shapiro, E (2008). "Towards Molecular Computers that Operate in a Biological Environment". Physica D: Nonlinear Phenomena . 237 (9): 1165–1172. Bibcode:2008PhyD..237.1165K. doi:10.1016/j.physd.2008.01.027.
  37. Andersen, Ebbe S.; Dong, Mingdong; Nielsen, Morten M.; Jahn, Kasper; Subramani, Ramesh; Mamdouh, Wael; Golas, Monika M.; Sander, Bjoern; et al. (2009). "Self-assembly of a nanoscale DNA box with a controllable lid". Nature. 459 (7243): 73–6. Bibcode:2009Natur.459...73A. doi:10.1038/nature07971. hdl: 11858/00-001M-0000-0010-9363-9 . PMID   19424153. S2CID   4430815.
  38. Medscape from WebMD [Internet]. New York: WebMD LLC; 1994-2015. Liposomes as Drug Delivery Systems for the Treatment of TB; 2011 [cited 2015 May 8] Available from: http://www.medscape.com/viewarticle/752329_3
  39. Hirschler, Ben (December 16, 2014). "3D Printing Points Way to Smarter Cancer Treatment". Reuters. London.

Further reading