Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics
Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics
Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics
Ebook1,237 pages13 hours

Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics

Rating: 0 out of 5 stars

()

Read preview

About this ebook

Multifunctional Systems for Combined Delivery, Biosensing, and Diagnostics explores how multifunctional nanocarriers are being used in combined delivery and diagnostics in contemporary medicine. Particular attention is given to efforts to i) reduce the side effects of therapeutic agents, ii) increase the pharmacological effect, and iii) improve aqueous solubility and chemical stability of different therapeutic agents. The chapters focus on applications of nanostructured materials and nanocarriers, highlighting how these can be used effectively in both diagnosis and delivery.

This applied focus makes the book an important reference source for those wanting to learn more about how specific nanomaterials and nanotechnology systems can help to solve drug delivery and diagnostics problems. This book is a valuable resource for materials scientists, bioengineers, and medical researchers who are looking for an applications-oriented guide on how nanotechnology and nanomaterials can be used effectively throughout the medical treatment process, from diagnosis to treatment.

  • Explores the benefits of using a variety of nanomaterials as drug delivery agents
  • Explains how nanocarriers can reduce the side effects of therapeutic agents
  • Provides an analysis of the pros and cons of using specific nanocarriers to solve particular diagnosis and delivery problems
LanguageEnglish
Release dateMay 3, 2017
ISBN9780323527262
Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics

Read more from Alexandru Mihai Grumezescu

Related to Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics

Related ebooks

Materials Science For You

View More

Related articles

Reviews for Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics - Alexandru Mihai Grumezescu

    Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics

    Edited by

    Alexandru Mihai Grumezescu

    University Politehnica of Bucharest,

    Bucharest, Romania

    Table of Contents

    Cover

    Title page

    Copyright

    Contributors

    Foreword

    Preface

    Chapter 1: Selective Effects of Azelaic Acid in Nanovesicles on Cell Lines

    Abstract

    1. Introduction and Historical Background

    2. Applications of Anticancer Drug-Loaded Nanovesicles

    3. Limitations of Available Therapies: Need of a Novel Therapeutic Strategy

    4. Conclusions

    Chapter 2: Antimicrobial Photodynamic Therapy (APDT) Action Based on Nanostructured Photosensitizers

    Abstract

    1. Antimicrobial Photodynamic Therapy (APDT)

    2. APDT for Bacterial Eradication

    3. Antimicrobial Action of APDT on Healing Processes

    4. Conclusions and Future Directions

    Chapter 3: Conspectus on Nanotechnology in Oral Cancer Diagnosis and Treatment

    Abstract

    1. Introduction

    2. Oral Cancer Diagnosis: Challenges

    3. Oral Cancer Treatment: Challenges

    4. Nanotechnology, Nanoparticles, and Nanomedicine

    5. Nanoparticles

    6. Conclusions

    Chapter 4: Lipid-Based Nanocarriers in Cancer Therapy

    Abstract

    1. Cancer Pathogenesis

    2. Cancer Treatments

    3. Limitations of Chemotherapy

    4. Nanocarriers in the Drug Delivery Process

    5. Lipid Nanocarriers

    6. Conclusions

    Chapter 5: Effect of Polymer-Based Nanoparticles on the Assay of Antimicrobial Drug Delivery Systems

    Abstract

    1. Introduction

    2. Classification of Antibiotics

    3. Mechanism of Action of Antibiotics

    4. Types of Antibacterial Agents

    5. Antibiotic Resistance

    6. Intracellular Delivery of Antimicrobial Agents via Polymer-Based Nanocarriers

    7. Classification of the Polymer-Based Nanocarriers for Antimicrobial Drug Delivery

    8. Analytical Strategies for the Characterization of Drug Delivery Systems

    9. Conclusions

    Chapter 6: Nanometric Biopolymer Devices for Oral Delivery of Macromolecules with Clinical Significance

    Abstract

    1. Introduction

    2. Mechanisms Behind Oral Transport of Macromolecules

    3. Hurdles to Oral Delivery of Macromolecules

    4. Enhancement of Oral Absorption of Macromolecules

    5. Biopolymer Nanodevices and Delivery of Macromolecules

    6. Conclusions

    Chapter 7: The Use of Nanotechnology in Modern Pharmacotherapy

    Abstract

    1. Introduction

    2. Polymer-Based Nanoparticles

    3. Lipid-Based Nanoparticles

    4. Carbon-Based Nanoparticles

    5. Nanodispersions

    6. Conclusions

    Chapter 8: Nanoparticle System for Anticancer Drug Delivery: Targeting to Overcome Multidrug Resistance

    Abstract

    1. Introduction

    2. Cancer and Cell Cycles

    3. Molecular Aspects of Multidrug Resistance

    4. Nanoparticle Drug Delivery Systems

    5. Conclusions

    Conflict of Interests

    Chapter 9: Application Potential of Engineered Liposomes in Tumor Targeting

    Abstract

    1. Introduction

    2. Engineered Liposomes

    3. Biological Targeting

    4. Combinatorial Drugs Approach

    5. Detachable PEG in Response to Tumor Stimulus

    6. Intracellular Delivery

    7. Remote-Controlled Payload Release

    8. Theranostic Liposomes

    9. Recent Advances

    10. Conclusions

    Chapter 10: Plant-Based Peroral Vaccines

    Abstract

    1. History and Principles of Plant-Made Vaccines

    2. Glycosylation of Antigenic Proteins

    3. PapillomatosIs, Vaccines (Gardasil, Cervarix), and our Approaches to the Proposition

    4. Future Perspectives

    Acknowledgments

    Chapter 11: Platelet-Rich Plasma Incorporated Nanostructures for Tissue Engineering Applications

    Abstract

    1. Introduction

    2. Growth Factor and PRP Delivery for Tissue Engineering Applications

    3. Chitosan-Based Nanomaterials for the Administration of PRP and Growth Factors

    4. Heparin-Containing Nanoparticles for the Entrapment of Growth Factors and PRP

    5. PRP and Growth Factor-Containing Electrospun Nanofibers for Regenerative Medicine

    6. Carbon Nanotube, Nanographene, and Graphene Oxide-Mediated Delivery of Therapeutics and Growth Factors

    7. Other Nanoparticle-Mediated Systems for Growth Factor Delivery

    8. Conclusions

    Chapter 12: Targeted Nanotherapeutics Based on Cancer Biomarkers

    Abstract

    1. Introduction to Nanotechnology

    2. Nanoparticles for Cancer Therapy

    3. Nanotransporters of Drug Delivery

    4. Targeted Therapeutics for Cancer Therapy

    5. Importance and Issues of Targeted Therapy

    6. Nanotherapeutics for Cancer Biomarkers

    7. HER2-Targeted Therapeutic Nanostructures

    8. EGFR-Based Targeted Therapeutics Nanostructures

    9. Folate-Targeted Therapeutics Nanostructures

    10. Conclusions and Future Directions

    Chapter 13: Therapeutic Nanoparticles for Targeted Delivery of Anticancer Drugs

    Abstract

    1. Introduction

    2. The Critical Impact of Passive and Active Targeting

    3. Strategies for Controlled Release

    4. Delivery Routes

    5. Conclusions and Future Perspectives

    Chapter 14: Pulmonary Administration of Biodegradable Drug Nanocarriers for More Efficacious Treatment of Fungal Infections in Lungs: Insights Based on Recent Findings

    Abstract

    1. Introduction

    2. Lung Morphology

    3. Lung Fungal Infection

    4. Treatment of Lung Fungal Infection

    5. Clinical Status to Treat Fungal Infection by Pulmonary Route

    6. Importance of Pulmonary Route for Drug Administration

    7. Principal Mechanisms of Respiratory Deposition

    8. Pulmonary Drug Delivery Devices

    9. Pulmonary Formulations to Treat Lung Fungal Infection

    10. Why Pulmonary Route?

    11. Limitations of Pulmonary Route (If Any)

    12. Conclusions

    Chapter 15: Alternative Technologies to Improve Solubility and Stability of Poorly Water-Soluble Drugs

    Abstract

    1. Introduction

    2. Biopharmaceutical Classification System

    3. Cyclodextrins

    4. Solid Dispersions

    5. Multicomponent Systems to Enhance Cyclodextrin Complexation

    6. Obtaining Methods of Inclusion Complexes, and Solid Dispersion

    7. Characterization of Systems

    8. Stability Study

    9. Conclusions

    Chapter 16: Oral Administration of Nanoparticles-Based TB Drugs

    Abstract

    1. Introduction

    2. Gastrointestinal Tract

    3. Nanoparticles as Drug Delivery Systems

    4. Future Directions and Challenges

    5. Conclusions

    Acknowledgments

    Chapter 17: Recombinant Lactic Acid Bacteria Secreting OxdC as a Novel Therapeutic Tool for the Prevention of Kidney Stone Disease

    Abstract

    1. Kidney Stones

    2. Hyperoxaluria

    3. Medical Management of Kidney Stones

    4. Probiotics

    5. Genetically Modified Lactic Acid Bacteria

    6. Risk Assessments of Genetically Modified Lactic Acid Bacteria

    7. Conclusions

    Index

    Copyright

    Elsevier

    Radarweg 29, PO Box 211, 1000 AE Amsterdam, Netherlands

    The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom

    50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States

    Copyright © 2017 Elsevier Inc. All rights reserved.

    No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

    This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein).

    Notices

    Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary.

    Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.

    To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein.

    Library of Congress Cataloging-in-Publication Data

    A catalog record for this book is available from the Library of Congress

    British Library Cataloguing-in-Publication Data

    A catalogue record for this book is available from the British Library

    ISBN: 978-0-323-52725-5

    For information on all Elsevier publications visit our website at https://www.elsevier.com/books-and-journals

    Publisher: Matthew Deans

    Acquisition Editor: Simon Holt

    Editorial Project Manager: Andrae Akeh

    Production Project Manager: Caroline Johnson

    Designer: Greg Harris

    Typeset by Thomson Digital

    Contributors

    Albert Abhishek,     Madurai Kamaraj University, Madurai, Tamil Nadu, India

    Mohammad Firoz Alam,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Neda Alasvand,     Materials and Energy Research Center, Tehran, Iran

    Maria H. Amaral,     University of Porto, Porto, Portugal

    Ravindran Ankathil,     Human Genome Centre, Health Campus University Sains Malaysia, Kubang Kerian, Kelantan, Malaysia

    Tarique Anwer,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Uddhav S. Bagul,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Samrat Chakraborty,     Jadavpur University, Kolkata, West Bengal, India

    Walter F. da Silva Júnior,     Federal University of Rio Grande do Norte, UFRN, Natal, Rio Grande do Norte, Brazil

    Priscila da Costa Carvalho de Jesus,     Center of Nanotechnology and Tissue Engineering, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil

    Ádley A.N. de Lima,     Federal University of Rio Grande do Norte, UFRN, Natal, Rio Grande do Norte, Brazil

    Jonas G. de Oliveira Pinheiro,     Federal University of Rio Grande do Norte, UFRN, Natal, Rio Grande do Norte, Brazil

    Fabia J.J. de Souza,     Federal University of Rio Grande do Norte, UFRN, Natal, Rio Grande do Norte, Brazil

    Moumita Dhara,     Jadavpur University, Kolkata, West Bengal, India

    Lopamudra Dutta,     Jadavpur University, Kolkata, West Bengal, India

    Mohamed E. Elmobark,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Mohamadreza Baghaban Eslaminejad,     Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran

    Marilene Estanqueiro,     University of Porto, Porto, Portugal

    Sivasamy Gomathi,     Madurai Kamaraj University, Madurai, Tamil Nadu, India

    Aysen Gumustas,     Ankara University, Ankara, Turkey

    Mehmet Gumustas

    Ankara University, Ankara

    Hitit University, Corum, Turkey

    P. Selcan Gungor-Ozkerim

    Biomaterials Innovation Research Center, Brigham and Women’s Hospital

    Massachusetts Institute of Technology, Cambridge, MA, United States

    Abouelhag Hussien,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Farah Islam,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Aamena Jabeen,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Ankit Jain,     Dr. Hari Singh Gour Central University, Sagar, Madhya Pradesh, India

    Sanjay K. Jain,     Dr. Hari Singh Gour Central University, Sagar, Madhya Pradesh, India

    Gyas Khan,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Joana Magalhães,     UCIBIO/REQUIMTE, University of Porto, Porto, Portugal

    Hamid Mahdavi

    Iran Polymer and Petrochemical Institute, Tehran

    Islamic Azad University, Sarvestan Branch, Iran

    Sabyasachi Maiti,     Gupta College of Technological Sciences, Asansol, West Bengal, India

    Hamid Mirzadeh,     Amirkabir University of Technology, Tehran, Iran

    Davod Mohebbi-Kalhori,     University of Sistan and Baluchestan, Zahedan, Iran

    Laboni Mondal,     Jadavpur University, Kolkata, West Bengal, India

    Carlos D.L.F.A. Moreira,     Federal University of Rio Grande do Norte, UFRN, Natal, Rio Grande do Norte, Brazil

    Masoud Mozafari,     Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran

    Biswajit Mukherjee,     Jadavpur University, Kolkata, West Bengal, India

    Vinod S. Nair,     Kerala University of Health Sciences, Thrissur, Kerala, India

    Aneta Ostróżka-Cieślik,     Medical University of Silesia, Katowice, Poland

    Sibel A. Ozkan,     Ankara University, Ankara, Turkey

    Tritsida Panyosak,     University of Phayao, Phayao, Thailand

    Eldho Paul,     Madurai Kamaraj University, Madurai, Tamil Nadu, India

    Paramita Paul,     Jadavpur University, Kolkata, West Bengal, India

    Marina Pinheiro,     UCIBIO/REQUIMTE, University of Porto, Porto, Portugal

    Fernando L. Primo,     São Paulo State University (UNESP), Araraquara, São Paulo, Brazil

    Ahmad Rabiee,     Iran Polymer and Petrochemical Institute, Tehran, Iran

    Maryam Rahmati,     Materials and Energy Research Center, Tehran, Iran

    Jayakumar Rajadas,     Stanford University, Palo Alto, CA, United States

    Anjana Ravindran,     Kerala University of Health Sciences, Thrissur, Kerala, India

    Ranjana Ravindran,     Kerala University of Health Sciences, Thrissur, Kerala, India

    Salette Reis,     UCIBIO/REQUIMTE, University of Porto, Porto, Portugal

    Chandrababu Rejeeth

    Bharathiar University, Coimbatore, Tamil Nadu, India

    Shanghai Jiao Tong University, Shanghai, China

    Natalya I. Rekoslavskaya

    Siberian Institute of Plant Physiology and Biochemistry of SB RAS

    Irkutsk Scientific Center (FASO), Irkutsk, Russia

    Maryam Saeidifar,     Materials and Energy Research Center, Tehran, Iran

    Mohammed M. Safhi,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Rurik K. Salyaev,     Siberian Institute of Plant Physiology and Biochemistry of SB RAS, Irkutsk, Russia

    Susana Santos,     INEB—Institute of Biomedical Engineering, i3S-Institute of Research and Innovation in Health, University of Porto, Porto, Portugal

    Beata Sarecka-Hujar,     Medical University of Silesia, Katowice, Poland

    Ponnusamy Sasikumar,     Madurai Kamaraj University, Madurai, Tamil Nadu, India

    Farshid Sefat,     University of Bradford, Bradford, United Kingdom

    Govindan Sadasivam Selvam,     Madurai Kamaraj University, Madurai, Tamil Nadu, India

    Ceyda T. Sengel-Turk,     Ankara University, Ankara, Turkey

    Soma Sengupta,     Jadavpur University, Kolkata, West Bengal, India

    Mohammad A. Shamekhi

    Iran Polymer and Petrochemical Institute, Tehran

    Islamic Azad University, Sarvestan Branch, Iran

    Rahimullah Siddiqui,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Sivagurunathan Moni Sivakumar,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    José M. Sousa Lobo,     University of Porto, Porto, Portugal

    Antonio C. Tedesco,     Center of Nanotechnology and Tissue Engineering, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil

    Ramar Thangam

    Bharathiar University, Coimbatore

    King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India

    Aleksandra M. Urbanska,     Irving Cancer Research Center, Columbia University, New York, NY, United States

    Bengi Uslu,     Ankara University, Ankara, Turkey

    Alexandre Vieira,     UCIBIO/REQUIMTE, University of Porto, Porto, Portugal

    Raju Vivek

    Bharathiar University, Coimbatore, Tamil Nadu, India

    Shanghai Jiao Tong University, Shanghai, China

    Foziyah Zakir,     College of Pharmacy, Jazan University, Jazan, Saudi Arabia

    Foreword

    Despite the huge progress in medical-related areas, numerous diseases are still far from being efficiently detected, treated, or prevented. The lack of success of current biomedical approaches relies on the complexity of some diseases, but also on their very diverse clinical symptoms, and wide variations among patients. A failed or incorrect detection of a health-threatening condition in its early stages usually leads to fatal effects on the patient. Moreover, wrong or late diagnosis involves increased costs, prolonged hospital stays, and numerous other patient and socioeconomical-related issues. In this context, a highly sensitive multifunctional system, able to detect, target and cure, or even prevent a health-damaging condition, is a widely investigated desiderate at the present time. Nanostructured sensors and drug-delivery systems bring a new light in the research and management of serious diseases, by ensuring a specific effect, a very targeted and controllable efficiency, and much lower side-effects, by being able to avoid damaging unaffected tissue.

    This book brings an updated and clear picture regarding the recent progress made in the field of disease detection, but also targeted nanodelivery of drugs for a wide variety of diseases, also aiming to offer a perspective for future approaches in diagnosis, treatment, and prophylaxis.

    A special attention is paid to the management of cancer, which is considered the disease of the century, and for whose research and therapy a huge amount of resources is invested annually. In this particular field, the design and implementation of multifunctional nanosystems represent the most feasible alternative for an efficient management, and it is considered a real strategy for reducing mortality and morbidity.

    The Editor selected the most recent, interesting, and innovative approaches, transposed in well-organized chapters, to offer a scientifically, updated but, at the same time, very accessible material for a wide audience.

    The book represents an attractive collection of up-to-date information in the field of multifunctional systems that can be simultaneously utilized for the detection and therapy of diseases, and is an important source of knowledge, offering at the same time novel perspectives in a very dynamic and challenging area.

    Alina Maria Holban

    University Politehnica of Bucharest, Bucharest, Romania

    Monica Cartelle Gestal

    University of Georgia, Athens, GA, United States

    Preface

    Numerous and varied diseases have emerged this century. In the context of constantly changing societies and population requirements, the medical field requires faster and more versatile tools for improved diagnosis and therapy.

    Multifunctional systems, designed at the micro- and nanoscale, were recently optimized not only to ensure a more efficient detection of diseases, but also a more personalized therapy.

    Biosensing coupled with the delivery of active formulations may represent a great solution to numerous difficult-to-treat and even incurable diseases. The use of very small devices to achieve a versatile diagnosis tool represents one of the most investigated aspects in the field of biomedicine. Progress made in the field of material engineering offered researchers and clinicians valuable alternatives in diagnosis, therapy, and prophylaxis, bringing at the same time a new light on the quality of life and survival expectance in critical patients.

    This book reviews and discusses the most novel and intriguing progress made in the field of multifunctional systems developed for diagnosis and therapy, with an emphasis on drug delivery and bioengineering progress in medical nanosystems.

    The book contains 17 chapters, prepared by outstanding international researchers from the United States, Iran, Russia, Saudi Arabia, Poland, India, Turkey, Portugal, Malaysia, Brazil, and Thailand.

    Chapter 1, prepared by Tritsida Panyosak and entitled Selective effects of azelaic acid in nanovesicles on cell lines, presents an up-to-date overview of an efficient therapeutic application for azelaic acid entrapped in bilayer vesicles for cancer treatment.

    Antonio Claudio Tedesco et al. in Chapter 2, entitled Antimicrobial photodynamic therapy (APDT) action based on nanostructured photosensitizers, present novel approaches used in APDT as a valuable method, extremely effective in cellular death mediated by the production of reactive species, which can overcome some of the most important problems of our days, namely the production of resistant or superresistant microorganisms selected over the years by the irrational use of antibiotics. Another approach presented here describes the basis of a new concept of photobiomodulation on the wound-healing process. This process increases the rates of tissue repair and, at the same time, acts simultaneously as an antibacterial and remodeling tissue protocol.

    Ravindran Ankathil et al. in Chapter 3, entitled Conspectus on nanotechnology in oral cancer diagnosis and treatment, present current advances in the field of nanotechnology, and especially the emergence of multimodal nanoparticles that offer the opportunity to develop future strategies significantly toward early diagnosis and more efficient treatment modalities for oral cancer.

    Chapter 4, prepared by Marilene Estanqueiro et al., entitled Lipid-based nanocarriers in cancer therapy, gives an up-to-date overview about: (1) cancer pathogenesis and treatments; (2) nanocarriers properties that make these systems advantageous for drug delivery; (3) liposomes and lipid nanoparticles, including their production methods; and (4) a critical revision of the research work developed under the application of liposomes and lipid nanoparticles in cytotoxic drug delivery.

    Chapter 5, entitled Effect of polymer-based nanoparticles on the assay of antimicrobial drug delivery systems, prepared by Mehmet Gumustas et al., underlines the effectiveness and in vitro/in vivo performance of various antibiotics encapsulated polymer-based nanoparticles in the treatment of infectious diseases. A collection of published papers is summarized from this perspective, and also comments on targeting microorganisms, advantage, and activity of the developed delivery system. Properties of analytical techniques for the determination of in vitro and in vivo samples with the validation parameters are presented.

    Sabyasachi Maiti, in Chapter 6, entitled Nanometric biopolymer devices for oral delivery of macromolecules with clinical significance, discusses various biological and physicochemical barriers, strategies adopted in augmenting bioabsorption of macromolecules, and the mechanisms behind their oral transport. A special emphasis is given to the recent developments in the area of nanobiopolymer devices for effective delivery of macromolecules.

    Chapter 7, prepared by Aneta Ostróżka-Cieślik and Beata Sarecka-Hujar, and entitled The use of nanotechnology in modern pharmacotherapy, reviews the structure, function, and utility of different nanoparticles, especially: nanospheres, nanocapsules, nanoliposomes, lipoplexes, lipospheres, dendrimers, fullerenes, carbon nanotubes, nanoemulsions, and nanosuspensions as potential drug carriers. Advantages and disadvantages of the particular nanostructures are also discussed.

    Chapter 8, entitled Nanoparticle system for anticancer drug delivery: targeting to overcome multidrug resistance, prepared by Mohammed M. Safhi et al., focuses on multiple drug resistance exhibited during cancer chemotherapy, and the influence of nanoparticles research in overcoming this issue.

    Ankit Jain and Sanjay K. Jain, in Chapter 9, entitled Application potential of engineered liposomes in tumor targeting, sums up the drug targeting potential of the engineered liposomes with contemporary trends in the field of liposomal research.

    Chapter 10, prepared by Rurik K. Salyaev and Natalya I. Rekoslavskaya and entitled Plant-based peroral vaccines, presents an up-to-date overview about plant derived peroral vaccines and their advantages, in comparison to traditional vaccines. Their impact in future therapeutics and prophylaxis is also discussed.

    Chapter 11, entitled Platelet-rich plasma incorporated nanostructures for tissue engineering applications, prepared by Mohammad Amin Shamekhi et al., reveals the progress of platelet-rich plasma– and growth factor–incorporated nanostructures. The authors expose the methodologies to load platelet-rich plasma and growth factors into nanostructures, such as exploiting heparin-conjugated nanospheres, electrospun nanofibers, graphene (oxide), and nanoliposomes.

    Raju Vivek et al., in Chapter 12, entitled Targeted nanotherapeutics based on cancer biomarkers, present a comprehensive overview of collected information of biomarkers-targeted therapeutic nanostructures for cancer nanotherapy.

    Chapter 13, entitled Therapeutic nanoparticles for targeted delivery of anticancer drugs, prepared by Neda Alasvand et al., reviews different types of anticancer drugs and engineering techniques for obtaining efficient delivery systems in cancer treatment. It also introduces the salient features of this approach, the hurdles that must be overcome, the associated perspectives, and practical constraints.

    Chapter 14 prepared by Biswajit Mukherjee et al., entitled Pulmonary administration of biodegradable drug nanocarriers for more efficacious treatment of fungal infections in lungs: insights based on recent findings, focuses on the progress in the oral delivery of drug nanocarriers, highlighting the pharmacokinetic and pharmacodynamics modulations, their advantages along the fundamental limitations, and the future perspective based on the recent findings in the field.

    Chapter 15, prepared by Walter Ferreira da Silva Júnior et al., entitled Alternative technologies to improve solubility and stability of poorly water-soluble drugs, summarizes the understanding of solid dispersions, and shows the applicability of these systems in unconventional systems.

    Joana Magalhães et al., in Chapter 16, entitled Oral administration of nanoparticles-based TB drugs, review the uptake and translocation of drugs in the gastrointestinal tract, the factors that affect drug stability, and other limitations of oral drug delivery. Additionally, the recent progress in nanodelivery systems for the oral administration of anti-TB (tuberculosis) drugs and their potential impact are discussed.

    Chapter 17, entitled Recombinant lactic acid bacteria secreting OxdC as a novel therapeutic tool for the prevention of kidney stone disease, prepared by Ponnusamy Sasikumar et al. discusses the potential impact of manipulating gut flora, with the potential probiotic bacteria that may have a positive impact on gut oxalate levels, and may decrease oxalate absorption.

    Alexandru M. Grumezescu

    University Politehnica of Bucharest, Bucharest, Romania, http://grumezescu.com/

    Chapter 1

    Selective Effects of Azelaic Acid in Nanovesicles on Cell Lines

    Tritsida Panyosak    University of Phayao, Phayao, Thailand

    Abstract

    New drug development is a complicated, costly, and time consuming process including preclinical and clinical trials, application for of new drug, and then approval by the US Food and Drug Administration (FDA). Liposomes and niosomes are nanovesicles that have been used widely as drug carriers. The encapsulation of drugs in these vesicular systems offers several advantages, including the modification of lipophilicity and hydrophilicity, decreasing toxicity, and increasing the stability, circulation time, and absorption of the drug. For example, safety assessment of azelaic acid (AA) in nanovesicles for pharmaceutical was determined. In the process, AA was encapsulated in niosomes and liposomes with the compositions of nonionic surfactant/cholesterol and phosphatidylcholine/cholesterol, respectively. AA and AA in nanovesicles, using MTT assay on three cancer cell lines comparing with vincristine, were investigated. AA incorporated in liposomes was found to be more potent than AA, and less potent than vincristine. AA incorporated in nanovesicles was more effective than AA in killing cancer cells; AA-nanovesicular formulations on normal cell lines picked up from mouse epidermis and AA encapsulated nanovesicles were moderated, when compared with cisplatin. Free drug of both nanovesicular formulations showed no growth inhibition. AA-incorporated nanovesicles have been proved to have antiproliferative effects in cancer cell lines. Furthermore, the safety of AA when incorporated in nanovesicles has been showed, with no toxicity to normal cell lines.

    Keywords

    azelaic acid

    nanovesicles

    SRB assay

    MTT assay

    Chapter Outline

    1 Introduction and Historical Background

    2 Applications of Anticancer Drug-Loaded Nanovesicles

    3 Limitations of Available Therapies: Need of a Novel Therapeutic Strategy

    4 Conclusions

    References

    1. Introduction and Historical Background

    New drug development is a complicated, costly, and time consuming process including preclinical and clinical trials (phase I, II, and III), applications made for the new drug, and approval from the US Food and Drug Administration (FDA). Enhancing the safety and efficacy of existing drugs has been attempted by different methods, most importantly by drug delivery at targeted sites. Drug delivery systems could equip extended circulating half-lives so that smaller quantities of a drug are required for a longer period of time, relieving the patient of side-effects caused by nonspecific tissue uptake, and providing protection against enzymatic degradation. Often drug delivery systems can select to permeate the cancerous cell and to decrease the toxicity for normal cell thus the encapsulated cancer drug can decrease the course of therapy. Lipid and nonionic surfactant-based drug delivery systems have attracted much attention from researchers as potential carriers of various bioactive molecules that could be used for therapeutic applications. Liposomes and niosomes are nanovesicles that have been widely used as drug carriers. Liposomes are composed of phospholipids, whereas niosomes contains nonionic surfactants. Niosomes alleviate the disadvantages associated with liposomes, such as chemical instability, variable purity of phospholipids, and high cost (Panyosak et al., 2009). The encapsulation of drugs in these vesicular systems offers several advantages, including the modification of lipophilicity and hydrophilicity, decreasing toxicity, and increasing stability, circulation time, and absorption of the drug. Many commercial drug delivery systems in the shape of liposomes/niosomes have already been stored with great success (Panyosak et al., 2007). For example, azelaic acid (AA), a naturally appearing substance, has been shown to have antimicrobial and anticancer activities (Breathnach, 1999), and it is an attractive drug candidate since it has nontoxic, nonteratogenic, and nonmutagenic properties. In cell cultures, AA has been shown to exhibit a dose- and time-dependent antiproliferative effect on several tumor cell lines, such as human cutaneous malignant melanoma, and human choroidal melanoma. It can penetrate selectively through tumor cells, as compared to normal cells (Breathnach, 1999).

    AA can be obtained by oleic acid and nitric acid oxidation, as well as by chemical, physical, or biological oxidation of free and esterified fatty acids. AA remains in the urine of normal individuals, and in excess in the urine of ketosis patients and in patients with a congenital component or acquired inability to β-oxidize monocarboxylic acids—dicarboxylic aciduria. It lacks acute or chronic toxicity, and it is nonteratogenic and has nonmutagenic effect. As an effect of topical application, Breathnach (1999) reported that after application of AA on lesions of primary cutaneous malignant melanoma, this latter exhibited the clinical regression of lesions, attended by destruction of malignant melanocytes, and return to normal organization of epidermis and dermis. In vitro, AA is a competitive inhibitor of tyrosinase, the key enzyme that generates melanocytes. The areas of antitumoral, antibacterial, and antiinflammatory activities have already been successfully applied to the treatment of acne (Breathnach, 1999). Currently, the FDA has approved finacea (AA) foam, 15%, for the topical treatment of the inflammatory papules and pustules of mild to moderate rosacea. The approval is based on results from two pivotal clinical trials examining the efficacy and safety of finacea foam, compared to its foam vehicle (without the drug AA) in the topical treatment of papulopustular rosacea (FDA, 2015), and its antipigmentary activity in treating melasma. The AA effects of antitumor approach will be examined from the viewpoint of its potential as a general antitumoral agent. This possibility will be investigated, as it could have a role as fundamental therapy, as synergistic therapies, as well as palliative therapy, by local, oral, or systemic administration in a number of cancer conditions (Breathnach, 1999). In vivo, AA is probably generated by lipoperoxidation of free and esterified cis-polyunsaturated fatty acids, such as those normally present in cell membrane phospholipids, and it may act as a natural antioxidant in vivo. In vitro, AA is a competitive inhibitor of oxidoreductive enzymes, including tyrosinase, enzymes involved in the synthesis of DNA, such as thioredoxin reductase and DNA polymerase, and oxidoreductases of the mitochondrial respiratory cycle. It is a potent inhibitor of microsomal 5-alpha-reductase, and inhibits anaerobic glycolysis. In vitro, it is a scavenger of toxic oxygen species, especially the toxic hydroxyl free radical. It also generated inhibitors, inhibiting the generation of reactive oxygen species by neutrophils. Some of these activities are involved in its antitumoral activities, and it can be administered to humans topically, orally, and in the form of an AA disodium salt by intrainjection or infusion, intravenously, intraarterially, and intralymphatically. Moreover, Breathnach (1999) reviewed that prolonging the infusion with successive doses of similar concentration produces sustained higher serum concentrations over a certain period. It has been evaluated that 90% of maximal uptake should be reached in the plateau phase of constant infusion of 2.2 g/h, with a maximum cellular (normal) uptake of 0.657 g/h (Breathnach, 1999). The serum levels achieved are equivalent to 5 × 10–3 mol/L and above, which is the level at which AA has an antiproliferative and cytotoxic effect on tumoral cells in culture (Breathnach, 1999). However, 60% of administered AA is eliminated in urine, leaving an unchanged structure within 12 h, and, in the mitochondrial part, is metabolized by β-oxidation via pimelic and glutaric acids to acetyl coenzyme A. This enters the Krebs cycle, and gives rise to CO2, and to malonyl coenzyme A that may be involved in fatty acid biosynthesis. AA generated no toxic metabolites, and crosses the blood-brain barrier. In culture cells, AA has been exhibited to have a time- and dose-dependent reversible antiproliferative and cytotoxic action as follows: tumoral cell lines are human cutaneous malignant melanoma, human choroidal melanoma, Harding–Passey and Cloudman murine melanoma, human squamous cell carcinoma, Raji lymphoma, and leukemia-derived cell lines, and fibroblastic lines (Breathnach, 1999). AA has been shown to penetrate selectively tumoral cells (HeLa, KB, and B16F10), as compared with normal cells (Panyosak et al., 2009). The antitumoral effect is connected to DNA synthesis inhibitor, and may damage mitochondria. AA also affects the karyotype of melanoma cells exposed to subtoxic doses in long-term culture, selectively affecting undifferentiated cells with a high growth rate and chromosomal abnormalities. It has an effect in in vitro on plasminogen activator activity, and reduces the fibrinolytic activity of cultured melanoma cells. The fibrinolytic potential of tumor cells correlates with their respective malignancy, and may play an important role in tumor invasion, progression, and metastasis (Breathnach, 1999). An antiviral effect has been reported. AA has an antibacterial activity against a wide range of organisms and, as secondary infection, can be a complication in some tumoral situations. The tumoral situations in which AA has been used and shown to be effective clinically are melanoma in situ and primary cutaneous malignant melanoma, and Grade IIIA melanoma with satellitosis. There are reports of a beneficial effect on penile lentiginosis and reticulate acropigmentation of Kitamura (Breathnach, 1999).

    2. Applications of Anticancer Drug-Loaded Nanovesicles

    Medical liposome applications are therapeutic and diagnostic applications. Drugs, makers encapsulated in liposomes, act as a model, tool, or reagent for basic studies of cell interactions, for the process of recognition and mode of action, but many drugs have a narrow therapeutic index. However, in various cases, the appropriate drug carriers can reduce the toxicity, or enhance the efficacy, of drugs. These were explained by pharmacokinetics and biodistribution. The drug–liposome delivery systems have benefits and limitations that depend on the synergy of liposomes with cells, and their destination in in vivo after being given drug-loaded nanovesicles in liposomes as route of administration. In addition, both in vitro and in vivo studies of the interaction with cells have shown that interactions are either simple adsorption or subsequent endocytosis. Moreover, Lasic (1992) reported that the interaction is an exchange between bilayer and cell membrane, and that liposomes are rapidly cleared from to the circulation by the macrophages located in liver, spleen, and bone marrow (Lasic, 1992). The modes of liposome action as drug delivery systems can give several advantages over conventional dosage forms, especially for parenteral, topical, and pulmonary routes of administration. Liposomes show different biodistribution and pharmacokinetics than free drug molecules. Liposomes can be used to improve therapeutic efficacy. The limitations are the retardation of bioavailability of the drug. Lasic (1995) reported that drug-loaded liposomes have advantages that can be applied as various dosages, summarized to seven categories, as follows (Lasic, 1995):

    1. Improved solubility of lipophilic and amphiphilic drugs. Examples include porphyrins, amphotericin B, minoxidil, peptides, and anthracyclines. Furthermore, hydrophilic drugs, such as doxorubicin or acyclovir, and drug-loaded liposomes at high concentrations gave solubility several folds over their aqueous solubility (Lasic, 1992).

    2. Passive targeting to the cells of the immune system, especially cells of the mononuclear phagocytic system (reticuloendothelial system): examples are antimonials, amphotericin B, porphyrins, and also vaccines, immunomodulators, or (immuno) supressors (Lasic, 1995).

    3. Formulations of liposomal actions, such as sustained release system, or locally administered routes: examples are doxorubicin, cytosine arabinose, cortisones, biological proteins, or peptides, such as vasopressin (Lasic, 1992).

    4. Site-avoidance mechanism: liposomes do not set in certain organs, such as heart, kidneys, brain, and nervous system. This will reduce cardiotoxicity (doxorubicin), nephrotoxicity (amphotericin B), and neurotoxicity (Lasic, 1995).

    5. Specific targeting site of liposomes with surface attached ligands can bind to target cells, or can be delivered to the target tissue by local structural conditions, such as leaky and badly formed blood vessels, and their basal capillaries (Lasic, 1995).

    6. Improve the transfer of hydrophilic, charged molecules, such as chelators, antibiotics, plasmids, and genes into cells (Lasic, 1995).

    7. Improve penetration into tissues, especially in the case of dermally applied liposomal dosage forms. Examples include anesthetics, corticosteroids, and insulin. Liposome encapsulation is selected to be used as carrier when drugs are very potent, toxic, and have very short lives in the blood circulation or at the sites of local administration (Lasic, 1995).

    Furthermore, Storm et al. (1987) reported that the efficacy was in many cases accommodated because of the decrease in bioavailability of the drug, especially if the tumor was not a phagocytic system, or located in the organs of mononuclear phagocytic system. In some cases, such as systemic lymphoma, the effect of liposome encapsulation showed improved efficacy due to the sustained release effect (Storm et al., 1987) while, in several other cases, the isolation of the drug into tissues of mononuclear phagocytic system actually reduced its efficacy. Gabizon (1989) presented liposomes in anticancer therapy, and that many different liposomal formulations of various anticancer agents were shown to be less toxic than the plain drug (Gabizon, 1989). Doxorubicin has been extensively studied with liposomes. In most cases, the toxicity of drugs entrapped in liposomes was decreased by about 50%. This includes both short-term and chronic toxicities, because liposomal encapsulation decreases the distribution of the drug molecules toward many tissues (Gabizon, 1989). Moreover, Lau et al. (2005) reported that bleomycin that was entrapped in liposomes (bleosome) has been aimed at the treatment of skin cancer. Free bleomycin in in vitro was shown LD50 approximately three-fold lower than bleosome on human squamous cell carcinoma (SCC) cells. Furthermore, free drug was shown nearly 30 times lower on human cutaneous keratinocytes (NEB-1) cells (Lau et al., 2005). In addition, Maswadeh et al. (2002) reported that the cytotoxic and cytostatic activities of the liposome-encapsulated vinblastine were examined against six leukemic human cell lines. The major improvement in vinblastine retention in buffer, as well as in culture medium, was achieved by employing dipalmitoylphosphatidylglycerol (DPPG). The 50% growth-inhibiting (GI50) and cytostatic activity of liposomal vinblastine did not seem to be affected by the type of the liposome, while the 50% cytotoxic activity (LC50) was affected in four out of the six cell lines tested (Maswadeh et al., 2002). Furthermore, liposomes prepared from lipids isolated from wheat germ were used to examine the percentage of vinblastine encapsulation, and its capacity into liposomes. The cytotoxic/cytostatic activity of these liposomal formulations has been examined against nine human leukemic cell lines. The results showed that a remaining percentage of 90% vinblastine in liposomes. The holding of the drug into liposomes was investigated and found to be time-dependent at 37°C. Liposomes loaded with vinblastine showed a mean of LC50 at 124.6 nM, and mean of GI50 at 30.8 nM (Maswadeh et al., 2000). On the other hand, Sharma et al. (1995) showed that taxanes, such as paclitaxel and docetaxel, act against ovarian cancer and other malignancies, while SB-T-1011, a taxane semisynthetic form, shows similar or greater in vitro cytostatic activity than paclitaxel, depending on the tumor cell line. The administration of taxanes is problematic because of their low solubility in most pharmaceutically agreeable solvents. This formulation used clinically contains cremophor/ethanol or polysorbate 80/ethanol, excipients that may cause serious adverse effects; meanwhile, in order to eliminate these vehicles, Sharma et al. (1995) have prepared paclitaxel liposome formulations. Antitumor activity was evaluated against A121a, a taxane-sensitive human ovarian (subcutaneous xenografts) tumor, growing as in athymic nude mice. Free taxane and taxane entrapped in liposomal formulations showed similar antitumor effects. The antitumor activity of paclitaxel and SB-T-1011 was similar, and docetaxel was more potent than either paclitaxel or SB-T-1011. On the whole, taxane liposomes were better and more easily tolerated via intravenous administration than taxane formulated in cremophor/ethanol (Sharma et al., 1995). Although the clinical formulation of paclitaxel is an important anticancer agent, it has significant side-effects, few sides of which are related to its formulation in cremophor/ethanol. Paclitaxel is difficult to formulate for intravenous administration since its structure is partly hydrophobic. However, Sharma et al. (1997) reported the therapeutic effects of two liposome formulations of paclitaxel against human tumor (ovarian A121) growing subcutaneously (xenograft in athymic nude mice). The liposome formulations used were ETL and TTL; the ETL formulation was composed of a single lipid, egg phosphatidylcholine (PC), and the TTL formulation was composed of a mixture of 3 lipids: di-elaidoyl PC, di-myristoyl, PC and 1-stearoyl,2-caproyl PC. TTL was used as a stable aqueous suspensions or as a reconstituted form of lyophilisation, while ETL was used as a reconstituted only lyophilate. Both paclitaxel–liposome formulations were much better tolerated than taxol, after intravenous or intraperitoneal administration. There were acute reactions seen after taxol administration, but were not exhibited when paclitaxel–liposome formulations were administered. All ETL and TTL preparations significantly delayed A121 tumor growth, similarly to taxol at equivalent doses. Furthermore, Sharma et al. (1997) showed, with pharmacokinetic data, that it is possible that paclitaxel rapidly dissociates from ETL or TTL after intravenous administration, and distributes in a manner similarly to taxol. ETL and TTL formulations may be useful clinically not only for eliminating the toxic effects of the cremophor/ethanol vehicle, but also for allowing alterations in the route of drug administration (Sharma et al., 1997). Cevc (1996) reported that transfersomes (TFs) are vesicles composed of phospholipids with 10%–25% surfactant (such as sodium cholate) and 3%–10% ethanol. The surfactant molecules confer ultradeformability on the TFs, and this reportedly allows them to squeeze through channels in the stratum corneum that are less than one-tenth the diameter of the TF (Cevc, 1996). Moreover, Alvi et al. (2011) showed that cytotoxicity study was carried out on HaCaT cells. The IC50 value of TFs (1.02 μmol/L), liposomes (6.83 μmol/L), and niosomes (9.91 μmol/L) was found to be less than 5-FU (15.89 μmol/L) at 72 h. 5-FU-loaded TFs were found to be most cytotoxic on the HaCaT cell line, compared to liposomes and niosomes. They concluded that vesiculization of 5-FU enhances the cytotoxic effect of 5-FU (Alvi et al., 2011). Hofer et al. (2000) reported that TFs have claimed to transport low- and high-molecular weight molecules in noninvasive forms into the body, such as several biologic characteristics of interleukin-2 (IL-2) and interferon (IFN) that were encapsulated in TFs. Blank TFs compose of natural phosphatidylcholine and sodium cholate. Moreover, TFs are able to entrap recombinant human IL-2 and human hybrid IFN. The biologic activity of immunotransfersomes was measured by a cytotoxic lymphoid line assay for IL-2, and an A549-encephalomyocarditis virus assay for IFN, respectively; concentrations of proteins were determined by ELISA. A large amount of IL-2 and IFN are able to incorporate into TFs: approximately 75%–80%. Furthermore, the increased lipid/protein ratio (90.9/1.0, respectively) led to a growing probability of association. They were able to show that IL-2 and IFN are trapped by TFs in a biologically active form, and in sufficient concentrations for immunotherapy. Moreover, Maswadeh et al. (2001) reported that the two liposome and TF formulations with or without encapsulated vinblastine were examined against nine human cell lines and the parameters GI50, TGI, LC50 (NCI protocol). Free DPPC/sodium cholate liposomes were found to exhibit strong antiproliferative activity, in contrast to the other three free liposomal formulations (DPPC/cholesterol, DMPC/cholesterol, DMPC/sodium cholate). Vinblastine encapsulated into the liposomes was found to exhibit 20-fold less activity on average, in the three parameters calculated compared to the free vinblastine (Maswadeh et al., 2001). Ethosomes (Touitou et al., 2000) are liposomes with high alcohol contents (up to 45%) that are capable of enhancing penetration to deep tissues and the systemic circulation (Heather, 2005). It is proposed that the alcohol fluidizes the ethosomal lipids and stratum corneum bilayer lipids, thus allowing the soft, malleable ethosomes to penetrate. In cell line studies, the cytotoxicity of vesicular formulation was determined by MTT assay, and the results showed that no significant toxicity was observed up to the concentration of 0.58 μmol/mL of lamivudine in ethosomal formulation. In contrast, in case of marked oral solution formulation and drug solution in PBS, cytotoxic concentration was observed at 0.24 μmol/mL concentration (Jain et al., 2007). Fang et al. (2009) showed that the skin route of administration of 5-aminolevulinic acid–photodynamic therapy (ALA–PDT) is an optional treatment for psoriasis that provides long-term therapeutic effects, is nontoxic, and enjoys better compliance with patients. However, the precursor of ALA is hydrophilic, thus its ability to penetrate the skin is limited. They employed a highly potent ethosomal system (phosphatidylethanolamine; PE) to investigate the penetration behavior of ALA and the recovery of skin in a hyperproliferative murine model. They found that the application of ethosomes produced a significant increase in cumulative amounts of 5–26-fold in normal and hyperproliferative murine skin samples, when compared to a hydrophilic ALA aqueous solution. The hydrophilic ALA solution appeared less precise in terms of the penetration mode in hyperproliferative murine skin. After the ethosomes had been applied, the protoporphyrin IX (PpIX) intensity increased about 3.64-fold, compared with that of the hydrophilic ALA solution. Furthermore, paclitaxel-loaded ethosomes are proposed as skin drug delivery systems for the treatment of this pathology due to their suitable physicochemical characteristics, and to their enhanced skin penetration ability for deep dermal delivery. In vitro data show that the skin application of paclitaxel-loaded ethosomes improved the permeation of paclitaxel in a stratum corneum–epidermis membrane model, and increased its antiproliferative activity in a squamous cell carcinoma model, as compared to the free drug (Paolino et al., 2012). From previous studies that exhibited the drug delivery by using nanovesicles, there is a promise to improve the therapeutic effect for cancer treatment, and to decrease toxicity to normal cells. The aim of the researcher is to improve the quality of life for cancer patients. However, the cancer drugs still present side effects. Thus, the researcher stills needs to find out the drug that is safe for patients. Enhancing safety and efficacy of the remaining drugs has been attempted using different methods and, most importantly, drug delivery at targeted sites. Drug delivery systems could equip extended circulating half-lives so that a smaller amount of drug is required for a longer period of time, relieving the patient of side-effects caused by nonspecific tissue uptake, and providing protection against enzymatic degradation. The encapsulated drug delivery systems can select to permeate the cancerous cell and relieve the patient of side-effects caused by nonselective uptake to normal cell. Although AA has cytotoxic effect; the poor solubility of AA limits its therapeutic applications. Thus, AA loaded nanovesicles were proper formulations as drug delivery. AA was entrapped in liposomes [l-α-dipalmitoyl phosphatidylcholine (DPPC)/cholesterol (CHL)] and niosomes with the compositions of Tween 61/CHL. AA shows efficient encapsulation in liposomes and niosomes. These encapsulated systems showed nanosize characteristics with good physical stability and size uniformity. AA entrapped and nonentrapped in nanovesicles, using MTT assay in three cancer cell lines (HeLa, KB and B16F10) compared with vincristine, were examined. The formulations showed selective activity on cell lines. In the liposomal form of AA, the antiproliferative activity was 0.67 times that of vincristine, or 92.67 times that of AA in HeLa cell lines, whereas in niosomal form it showed activity 0.34 times that of vincristine, or 57.92 times that of AA in B16F10 cell lines. AA entrapped in nanovesicles was more effective than AA in killing cancer cells (Panyosak et al., 2007). Moreover, the safety assessment of AA in nanovesicles has not been demonstrated on normal cell lines (mouse epidermal cell lines, JB6). Therefore, the SRB assay used for cell density determination, based on the measurement of cellular protein contents, has been optimized for the toxicity screening of compounds to adherent cells in a 96-well plate. The IC50 of JB6 cells with blank liposomes (DPPC/CHL, 7:3) and blank niosomes (Tween 61/CHL, 1:1) did not show the cytotoxic effect, whereas Tween 61 showed a few growth inhibitory effect on JB6 cells, most likely due to the surfactant activity of Tween 61 that may solubilize the cell membrane (Panyosak et al., 2009). Moreover, liposomes composed of phospholipids were of a more lipid nature than niosomes composed of the surfactant Tween 61. The proper hydrophobic and hydrophilic balance of AA may be adjusted suitably by different bilayer vesicular systems to obtain the maximum cytotoxicity activity. The cytotoxic effect of AA entrapped in nanovesicles on normal epidermal cell lines (mouse cell lines, JB6) was moderate, when compared with plain drugs and cisplatin. These may be due to the selective penetration of AA to tumor cells, as compared with normal cells and nanovesicles that gave sustained release effect of the drug to expose the cell lines (Panyosak et al., 2009). Although the niosomal system represents an attractive alternation to the liposomal system for the improvement of drug delivery and action, due to its lower cost, high stability and alleviation of storage issues, its toxicity due to its surfactant composition should be a concern.

    3. Limitations of Available Therapies: Need of a Novel Therapeutic Strategy

    In spite of everything said and done, cancer still remains a challenge for the 21st century. Although anticancer drug therapy has contributed significantly to improved patient/disease management, its current use is associated with several shortcomings and inconveniences to the patient. In the available therapies, the anticancer drug-loaded nanovesicles are reported to be the most preferred option over other therapies. However, anticancer drug-loaded nanovesicles are associated with several limitations that still remain the thrust of a novel therapy in this regard. The most important problem with cancer therapy today is the development of multidrug resistance (MDR) against anticancer agents developed during the course of therapy that leads to poor clinical outcomes. The possible reasons for MDR may be the principal mechanism, at the present time cancers show resistance to chemotherapy drugs, and this is a major factor in the failure to kill cancers of many dosage forms of MDR-chemotherapy. Chemotherapy kills drug-sensitive cells, but leaves behind a higher proportion of drug-resistant cells when the tumor begins to grow again, and chemotherapy may fail due to the remaining tumor cells that are now resistant. Commonly found chemoresistance in cancer is observed in P-glycoprotein and the so-called multidrug resistance associated protein (MRP) (Nature Biotechnology, 2000). Development of a novel drug delivery system (NDDS) or/and a new chemical entity appears to be the option. No doubt there has been tremendous interest from researchers in the development of NDDS for the treatment of cancer, but the outcome is far from the expectations. NDDSs encompass both controlled and targeted drug delivery carriers, and there already exist a number of carriers, such as dendrimers, nanovesicles, microspheres, nanoparticles, etc. The several applications as well as the key features of the NDDS in general would be beyond the scope of this article. One of the emerging areas of NDDS research is nanotechnology—which is not a new word to the scientific society of today.

    4. Conclusions

    Development of novel drug delivery systems (NDDS) appears to be the option to go forward. No doubt there has been a tremendous interest from researchers in the development of NDDS for the treatment of cancer, but the outcome is far from the expectations. However, AA is attractive nontoxic, nonteratogenic, and nonmutagenic anticancer candidate, because of its antiproliferative and cytotoxic effect on a variety of tumoral cells. AA can be efficiently encapsulated in liposomes and niosomes. These encapsulated systems exhibited nanosize characteristics with good physical stability and size uniformity. In the liposomal form of AA, the antiproliferative activity is 0.67 times that of vincristine, or 92.67 times that of AA in HeLa cell lines whereas, in niosomal form, it showed activity 0.34 times that of vincristine, or 57.92 times that of AA in B16F10 cell lines. For the cytotoxic effect of AA in nanovesicular formulations on cell growth determined by the SRB assay, AA—including blank liposomes and niosomes—showed moderate cytotoxic effect, compared with cisplatin on normal epidermal cell lines (JB6, mouse cell lines). Blank liposomes and niosomes exhibited no growth inhibitory effect on normal cells. This study has demonstrated the safety of AA when entrapped in nanovesicles due to its nontoxicity to normal cell lines. Although the niosomal system represents an attractive alternative to the liposomal system for the improvement of drug delivery and action, due to its lower cost, high stability, and alleviation of storage issues, its toxicity due to its surfactant composition should be a concern. The results of this study can be applied to further development of an efficient therapeutic application for AA entrapped in bilayer vesicles for cancer treatment.

    References

    Alvi IA, Madan J, Kaushik D, Sardana S, Pandey RS, Ali A. Comparative study of transfersomes, liposomes, and niosomes for topical delivery of 5-fluorouracil to skin cancer cells: preparation, characterization, in-vitro release, and cytotoxicity analysis. Anticancer Drugs. 2011;22(8):774–782.

    Breathnach AS. Azelaic acid: potential as a general antitumoural agent. Med. Hypotheses. 1999;52:221–226.

    Cevc G. Transfersomes, liposomes and other lipid suspensions on the skin: permeation enhancement, vesicle penetration, and transdermal drug delivery. Crit. Rev. Ther. Drug Carrier Syst. 1996;13:257–388.

    Fang YP, Huang YB, Wu PC, Tsai YH. Topical delivery of 5-aminolevulinic acid-encapsulated ethosomes in a hyperproliferative skin animal model using the CLSM technique to evaluate the penetration behavior. Eur. J. Pharm. Biopharm. 2009;73:391–398.

    Food and Drug Administration, 2015. Finacea® Foam. Available from: http://www.fda.gov/downloads/drugs/informationondrugs/ucm458596.pdf

    Gabizon A. Liposomes as a drug delivery system in cancer therapy. In: Roerdink FHD, Kron AM, eds. Drug Carrier Systems. Chichester, UK: Wiley; 1989:185–211.

    Heather AEB. Transdermal drug delivery: penetration enhancement techniques. Curr. Drug Deliv. 2005;2:23–33.

    Hofer C, Hartung R, Göbel R, Deering P, Lehmer A, Breul J. New ultradeformable drug carriers for potential transdermal application of Interleukin-2 and Interferon-α: theoretic and practical aspects. World J. Surg. 2000;24:1187–1189.

    Jain S, Tiwar AK, Sapra B, Jai NK. Formulation and evaluation of ethosomes for transdermal delivery of lamivudine. AAPS PharmSciTech. 2007;8(4):E1–E9: Article 111.

    Lau KG, Hattori Y, Chopra S, O’Toolec EA, Storey A, Nagai T, Maitani Y. Ultra-deformable liposomes containing bleomycin: in vitro stability and toxicity on human cutaneous keratinocyte cell lines. Int. J. Pharm. 2005;300:4–12.

    Lasic DD. Liposomes. Am. Sci. 1992;80:20–31.

    Lasic DD. Applications of liposomes. In: Lipowsky R, Sackmann E, eds. Handbook of Biological Physics. Menlo Park, CA: Elsevier Science B.V; 1995:491–519.

    Maswadeh H, Hatziantoniou S, Demetzos C, Dimas K, Georgopoulos A, Rallis M. Encapsulation of vinblastine into new liposome formulations prepared from triticum (wheat germ) lipids and its activity against human leukemic cell lines. Anticancer Res. 2000;20(6B):4385–4390.

    Maswadeh H, Demetzos C, Dimas K, Hatziantoniou S, Georgopoulos A, Rallis M, Dallas P, Papaioannou G. Accumulation of vinblastine into transfersomes and liposomes in response to a transmembrane ammonium sulfate gradient and their cytotoxic/cytostatic activity in vitro. Anticancer Res. 2001;21(4A):2577–2583.

    Maswadeh H, Demetzos C, Dimas K, Loukas YL, Georgopoulos A, Mavromoustakos T, Papaioannou GT. In-vitro cytotoxic/cytostatic activity of anionic liposomes containing vinblastine against leukaemic human cell lines. J. Pharm. Pharmacol. 2002;54(2):189–196.

    Nature Biotechnology, 2000. Cancer multidrug resistance, Nat. Biotechnol. Available from: http://www.nature.com/nbt/journal/v18/n10s/pdf/nbt1000_IT18.pdf

    Panyosak, A., Manosroi, A., Rojanasakul, Y., Manosroi, J., 2007. Characteristics and anti-proliferative activity of azelaic acid and its derivatives entrapped in bilayer vesicles in cancer cell lines. Controlled Release Society German Chapter Annual Meeting, Freiburg, Germany, Poster presentation, p. 15.

    Panyosak A, Manosroi J, Rojanasakul Y, Manosroi A. Safety assessment of azelaic acid and its derivatives entrapped in nanovesicles. Hum. Exp. Toxicol. 2009;28:387–392.

    Paolino D, Celia C, Trapasso E, Cilurzo F, Fresta M. Paclitaxel-loaded ethosomes®: potential treatment of squamous cell carcinoma, a malignant transformation of actinic keratosis. Eur. J. Pharm. Biopharm. 2012;81:102–112.

    Sharma A, Straubinger RM, Ojim aI, Bernacki RJ. Antitumor efficacy of taxane liposomes on a human ovarian tumor xenograft in nude athymic mice. J. Pharm. Sci. 1995;84(12):1400–1404.

    Sharma A, Mayhew E, Bolcsak L, Cavanaugh C, Harmon P, Janoff A, Bernacki RJ. Activity of paclitaxel liposome formulations against human ovarian tumor xenografts. Int. J. Cancer. 1997;71(1):103–107: 28.

    Storm GFH, Roerdink PA, Steerenberg WH, Crommelin DJA. Influence of lipid composition on the antitumor activity exerted by doxorubicin containing liposomes in a rat solid tumor model. Cancer Res. 1987;47:3366–3372.

    Touitou E, Dayan N, Bergelson L, Godin B, Eliaz M. Ethosomes - novel vesicular carriers for enhanced delivery: characterization and skin penetration properties. J. Control Release. 2000;65:403–418.

    Further Reading

    Baca-Estrada ME, Foldvari M, Babiuk SL, Babiuk LA. Vaccine delivery: lipid—based delivery systems. J. Biotechnol. 2000;83:1–2.

    Bangham AD, Horne RW. Negative staining of phospholipids and their structured modofication by surface active agents as observed in the electron microscope. J. Mol. Biol. 1964;8:660–668.

    Bouwstra JA, Gooris GS, Spek JA, Bras W. Structural investigations of human stratum corneum by small angle X-ray scattering. J. Invest. Dermatol. 1991;97:1005–1012.

    Carmichael AS, Marks R, Graupe KA. Topical azelaic acid in the treatment of rosacea. J. Dermatol. Treat. 1993;4(1):19–24.

    Cunliffe WJ. The clinical efficacy of azelaic acid in the treatment of acne. Rev. Contemp. Pharmacother. 1993;4:433–439.

    Elewski BE, Fleischer AB, Pariser DM. A comparison of 15% azelaic acid gel and 0.75% metronidazole gel in the topical treatment of papulopustular rosacea: results of a randomized trial. Arch. Dermatol. 2003;139:1444–1450.

    Gatti R, Cavrini V, Roveri P. Chromatographia. 1992;33:13–18.

    Gollnick H, Schramm M. Topical therapy in acne. J. Eur. Acad. Dermatol. 1998;11:S8–S12.

    Gude RP, Jadhav MG, Rao SGA, Jagtap AG. Effects of niosomal cisplatin and combination of the same with theophylline and with activated macrophages in murine B16F10 melanoma model. Cancer Biother. Radiopharm. 2002;17:183–192.

    Holland KT, Bojar R. The effect of azelaic acid on cutaneous bacteria. J. Dermatol. Treat. 1989;1:17–19.

    Jansen AB, Russell TJ. Some novel penicillin derivatives. J. Chem. Soc. 1965;65:2127–2132.

    Kent GL, Yoshiyuki H, Sunil C, Edel AO, Alan S, Tsuneji N, Yoshie M. Ultra-deformable liposomes containing bleomycin: In vitro stability and toxicity on human cutaneous keratinocyte cell lines. Int. J. Pharm. 2005;300:4–12.

    Lipowsky R. The conformation of membranes. Nature. 1992;349:475–481.

    Mayer-da-Silva A, Gollnick H, Detmar M, Gassmuller J, Parry A, Muller R, Orfanos CE. Effects of azelaic acid on sebaceous gland, sebum excretion rate and keratinization pattern in human skin. An in vivo and in vitro study. Acta Derm. Venereol. Suppl. 1989;143:20–30.

    Nazzaro-Porro M, Passi S. Identification of tyrosinase inhibition in cultures of Pityrosporum. J. Invest. Dermatol. 1978;71:205–208.

    Nazzaro-Porro M, Passi S, Balus L, Breathnach AS. Effect of dicarboxylic acids on lentigo maligna. J. Invest. Dermatol. 1979;72:296–305.

    Papahadjopoulos D. Liposomes and their use in biology and medicine. Ann. NY Acad. Sci. 1978;308:1–412.

    Papazisis KT, Geromichalos GD, Dimitriadis KA, Korsaris AH. Optimization of the sulforhodamine B colorimetric assay. J. Immunol. Methods. 1997;208:151–158.

    Skehan P, Storeng R, Scudiero D, Monks A, McMahon J, Varren JT, Bokesch H, Kenney S, Boyd M. New colorimetric cytotoxicity assay for anticancer drug screening. J. Natl. Cancer Inst. 1990;82:1107–1112.

    Stamatiadis D, Bulteau-Portois MC, Mowszowicz I. Inhibition of 5 α-reductase activity in human skin by zinc and azelaic acid. Brit. J. Dermatol. 1988;119:627–632.

    Thiboutot D, Thieroff-Ekerdt R, Graupe K. Efficacy and safety of azelaic acid (15%) gel as a new treatment for papulopustular rosacea: results from two vehicle-controlled, randomized phase III studies. J. Am. Acad. Dermatol. 2003;48:836–845.

    Chapter 2

    Antimicrobial Photodynamic Therapy (APDT) Action Based on Nanostructured Photosensitizers

    Antonio C. Tedesco*

    Fernando L. Primo**

    Priscila da Costa Carvalho de Jesus*

    *    Center of Nanotechnology and Tissue Engineering, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil

    **    São Paulo State University (UNESP), Araraquara, São Paulo, Brazil

    Abstract

    Important features are linked to the concept of antimicrobial photodynamic therapy (APDT). One approach is the need of effective strategies to overcome bacterial resistance to antibiotics. In this context, APDT has emerged as a valuable method, once cellular death is mediated by the production of reactive oxygen or nitrogen species (ROS or RNS), so it is very unlikely that resistant microorganisms may be selected. Another approach regards to photobiomodulation on wound healing for, simultaneously, antibacterial and remodeling tissue effects, as severe wounds are normally compromised by infection. APDT with appropriate photoactive nanodrugs specially designed for this purpose may contribute to both the wound regenerative process of the skin and at the same time protects and eradicates bacterial infections, accelerating the healing process with less or no side effects. Several issues are involved on APDT, among the design and choice of the nanostructured photosensitizer and how to certificate that it will penetrate the cellular cytoplasm or specific cellular organelles in the target tissue. For instance, Gram-positive bacteria are sensitive to APDT with a wide range of porphyrins and phthalocyanine compounds used as nanoencapsulated photosensitizers. On the other hand, Gram-negative have considerable resistance to the APDT process, as their external membrane may act as a barrier for permeability of the drug, besides being negatively charged. New efforts to overcome this barrier are under study with good results in the eradication of microorganisms, such as bacteria, fungi, viruses, and protozoa, by photoinactivation. Therefore, the selection of an ideal nanomaterial as drug delivery system is crucial to understand and develop more efficient APDT protocols based on the mechanisms of the antimicrobial inactivation.

    Keywords

    antimicrobial photodynamic therapy

    nanobiotechnology

    photosensitizers

    laser inactivation

    phthalocyanine

    polymeric nanoparticles

    photoprocesses

    Chapter Outline

    1 Antimicrobial Photodynamic Therapy (APDT)

    1.1 Background

    1.2 Mechanisms of Photosensitization

    1.3 Inactivation of Fungi and Bacteria

    2 APDT for Bacterial Eradication

    2.1 Bacterial Resistance to Antibiotics

    2.2 Gram-Positive and Gram-Negative Bacterial Behavior

    2.3 Nanostructured Photosensitizers

    2.4 Liposomal Formulation

    3 Antimicrobial Action of APDT on Healing Processes

    3.1 Skin Regeneration Processes

    3.2 Nanotechnology and Photosensitizers in APDT

    4 Conclusions and Future Directions

    References

    1. Antimicrobial Photodynamic Therapy (APDT)

    1.1. Background

    Historically, the use of light and dyes to treat diseases dates from the ancient Egypt, approximately 4000 years ago, when vitiligo was treated by a combination of plants orally administered, and exposure to sunlight. The success of the treatment was a result of photodynamic reactions mediated by psolaren, a natural product present on the extract of Ammimajus, a furanocoumarin-containing plant (Baltazar et al., 2015). However, the systematic study of photosensitizing reactions was only initiated at the beginning of the 20th century, when the director of the Institute of Pharmacology at the University of Munich, Hermann von Tappeiner, and his doctoral student, Oscar Raab, conducted experiments with the organic photosensitizer acridine orange, in the presence of daylight, on the protozoon Paramecium caudatum. By that time they had already concluded that the concentration of the dye and light intensity were determinant to eliminate protozoa. Together with the dermatologist Jesionek, in 1903, von Tappeiner treated patients with lupus vulgaris, stage II syphilis, and superficial skin cancer with topical eosin red solution (1%–5%), and obtained successful results (Babilas et al., 2010). They observed indications that the phototoxic effect only occurred in presence of oxygen, so these dyes were further designated photosensitizers, and the observed phenomenon was called photodynamic action (Baltazar et al., 2015).

    The first photosensitizer widely tested was hematoporphyrin (Hp), obtained from dried blood treated with sulfuric acid. Hausman, in 1911, promoted the first in vitro tests using Hp against paramecia and erythrocytes. He also verified the effect of Hp on murine skin after exposure to light (Baltazar et al., 2015). Meyer-Betz, in 1912, studied the photodynamic effect of porphyrins in humans, injecting himself 200 mg of Hp. Subsequent exposure of small fractions of his arm skin to visible light resulted in a strong reaction of sunburn, indicating that Hp has a phototoxic effect. Years later, in 1950, Schwartz discovered that the side-effect observed by Meyer-Betz was not because of Hp that is rapidly released in the organism, but it was caused by a mixture of dimeric and oligomeric derivatives of haematoporphyrin that were easily formed during the purification of this compound (Sternberg and Dolphin, 1998).

    The tendency of porphyrins in accumulating preferentially in tumors was discovered in 1924, when Policard observed that tumor tissues were more fluorescent than normal ones. This was based on experiments using wood lamp to detect fluorescence after Hp application in rat sarcoma (Baltazar et al., 2015). In 1942, Auler and Banzer demonstrated the preferential accumulation of hematoporphyrin derivatives (HpD) in tumor tissues, and their destruction by visible light action (Nyman and Hynninen, 2004). In 1955, Schwartz and coworkers verified that the phototoxicity of Hp could be diminished by treatment with acetic and sulfuric acids, obtaining HpD. However, the development of photodynamic therapy as a protocol to be used in vivo for diagnosis and treatment of diseases only started in 1960 by the studies of Lipson and Baldes (Lipson et al., 1961). They reported the properties of chemically modified HpD through partial polymerization reactions. This compound presents the ability of selectively localizing in tumors, being the first dye to be used in photodynamic therapy (PDT). In the period 1960–1967, Lipson and coworkers pioneered the use of combinations of HpD and visible light to treat breast cancer.

    The clinical use of PDT in patients with cancer dates from the end of 1970 decade, when the first studies of the effects of HpD with light in five patients with bladder cancer were published. In 1975, Dougherty et al. (1975) reported the first series of studies in patients successfully treated with HpD using the principles of PDT. From the variety of tumors evaluated, all responded to the treatment. Since this work was published, more than 200 clinical trials using PDT were reported, including for tumors in the skin, head and neck, digestive system, urinary system (prostate and bladder), and brain (Agostinis et al., 2011).

    Regarding the use of PDT in microorganisms, referred as antimicrobial photodynamic therapy (APDT), Shawar and Cooper (1990) reported the importance of the dye concentration, its absorption, and its relation with the photosensitization of Bacillus subtilis and Streptococcus faecalis, using HpD as photosensitizer and monochromatic visible light at a wavelength of 630 nm. Kinetic studies conducted by the same authors indicated that the drug amount connected to the cells increases with the increase in drug concentration on the incubation sample, until the saturation of the nonidentified binding sites (Shawar and Cooper, 1990). In 1993, the first photosensitive drug, photofrin, was approved for clinical use in Canada, being approved 2 years later by the US Food and Drug Administration (Baltazar et al., 2015).

    1.2. Mechanisms of Photosensitization

    The interaction of visible laser light with the photosensitizer drugs (PS)

    Enjoying the preview?
    Page 1 of 1