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

Only $11.99/month after trial. Cancel anytime.

Lipid Nanocarriers for Drug Targeting
Lipid Nanocarriers for Drug Targeting
Lipid Nanocarriers for Drug Targeting
Ebook1,260 pages13 hours

Lipid Nanocarriers for Drug Targeting

Rating: 0 out of 5 stars

()

Read preview

About this ebook

Lipid Nanocarriers for Drug Targeting presents recent advances in the area of lipid nanocarriers. The book focuses on cationic lipid nanocarriers, solid lipid nanocarriers, liposomes, thermosensitive vesicles, and cubosomes, with applications in phototherapy, cosmetic and others. As the first book related to lipid nanocarriers and their direct implication in pharmaceutical nanotechnology, this important reference resource is ideal for biomaterials scientists and those working in the medical and pharmaceutical industries that want to learn more on how lipids can be used to create more effective drug delivery systems.

  • Highlights the most commonly used types of lipid nanocarriers and explains how they are applied in pharmacy
  • Shows how lipid nanocarriers are used in different types of treatment, including oral medicine, skin repair and cancer treatment
  • Assesses the pros and cons of using different lipid nanocarriers for different therapies
LanguageEnglish
Release dateApr 16, 2018
ISBN9780128136881
Lipid Nanocarriers for Drug Targeting

Read more from Alexandru Mihai Grumezescu

Related to Lipid Nanocarriers for Drug Targeting

Related ebooks

Science & Mathematics For You

View More

Related articles

Reviews for Lipid Nanocarriers for Drug Targeting

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

    Lipid Nanocarriers for Drug Targeting - Alexandru Mihai Grumezescu

    Lipid Nanocarriers for Drug Targeting

    Edited by

    Alexandru Mihai Grumezescu

    Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Romania

    Pharmaceutical Nanotechnology Series

    Table of Contents

    Cover image

    Title page

    Copyright

    List of Contributors

    Series Preface: Pharmaceutical Nanotechnology

    Preface

    Chapter 1. Lipid nanocarriers

    Abstract

    1.1 Introduction to Lipid Nanocarriers

    1.2 Liposome

    1.3 Ethosomes

    1.4 Transfersomes

    1.5 Solid Lipid Nanoparticles

    1.6 Nanostructured Lipid Carriers (NLC)

    1.7 Lipid Drug Conjugates (LDC)

    1.8 Marketed or Commercially Available Nanolipid Products

    1.9 Applications of Various Lipid Nanocarriers

    1.10 Conclusion

    1.11 Future Prospects

    References

    Further Reading

    Chapter 2. Lipid nanoparticulate systems: Modern versatile drug carriers

    Abstract

    2.1 Introduction

    2.2 Lipid Nanoparticulate Systems: Types and Definitions

    2.3 Methods of Preparation

    2.4 Structure and Morphology of Lipid-Based Nanoparticles

    2.5 Composition of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers

    2.6 Characterization of Lipid Nanoparticles

    2.7 Stability Aspects of Solid Lipid Nanoparticles/Nanostructured Lipid Carriers

    2.8 Safety and Toxicological Aspects

    2.9 Applications of Solid Lipid Nanoparticles/Nanostructured Lipid Carriers by Different Routes of Administration

    2.10 Conclusions

    Acknowledgments

    References

    Further Reading

    Chapter 3. Nanotechnology in phytotherapy: Current challenges of lipid-based nanocarriers for the delivery of natural products

    Abstract

    3.1 Introduction

    3.2 Lipid Nanocarriers Containing Natural Products

    3.3 Outlook

    References

    Further Reading

    Chapter 4. Innovative vesicles for dermal and transdermal drug delivery

    Abstract

    4.1 Introduction

    4.2 The Skin and Drug Penetration Routes

    4.3 Liposomes

    4.4 Ethosomes

    4.5 Transferosomes

    4.6 Niosomes

    4.7 Conclusion

    References

    Chapter 5. Engineering of bacterial outer membrane vesicles: Potential applications for the development of vaccines

    Abstract

    5.1 Introduction

    5.2 Engineering of Outer Membrane Vesicles

    5.3 Immunological Considerations of the Use of Outer Membrane Vesicles as Vaccines

    Acknowledgements

    References

    Chapter 6. Plasmid-DNA lipid nanovaccines: An innovative approach for a better world health

    Abstract

    6.1 Introduction

    6.2 Plasmid DNA Immunization

    6.3 Plasmid DNA Vaccines Design and Enhancement

    6.4 Plasmid DNA Bioprocessing

    6.5 Plasmid-DNA Lipid Nanovaccines

    6.6 Characterization of Lipid Nanovaccines

    6.7 Conclusions

    Acknowledgments

    References

    Further Reading

    Chapter 7. Ocular delivery of solid lipid nanoparticles

    Abstract

    7.1 Introduction

    7.2 Anatomy and Physiology of the Eye

    7.3 Routes and Barriers of Ocular Drug Delivery

    7.4 Most Common Eye Diseases Pharmacologically Treated

    7.5 Colloidal Carriers

    7.6 Lipid Nanoparticles

    7.7 Marketed Formulations and Ongoing Clinical Trials

    7.8 Experimental Preclinical Studies

    7.9 Safety and Toxicological Studies

    7.10 Conclusions

    Acknowledgements

    References

    Further Reading

    Chapter 8. Lipid-based nanoparticles for cancer treatment

    Abstract

    8.1 Introduction

    8.2 The Use of Nanoparticles in Cancer Drug Delivery

    8.3 Lipid-Based Nanoparticles in Cancer Treatment

    8.4 Techniques for Synthesizing Lipid-Based Nanoparticles

    8.5 Lipid-Based Nanoparticle Characterization

    8.6 The Challenges Facing Nanoparticle Formulations

    8.7 Targeting of Lipid-Based Nanoparticles Into Tumor Cells

    8.8 Nanoparticles in RNAi Therapeutics

    8.9 Conclusion and Future Outlook

    Acknowledgment

    References

    Chapter 9. Developing liposomal nanomedicines for treatment of patients with neuroblastoma

    Abstract

    9.1 Introduction

    9.2 Genetics and Biology of Neuroblastoma

    9.3 Current Treatments for Neuroblastoma

    9.4 Role for Nanomedicines in Better Treatment of Neuroblastoma

    9.5 Potential of Liposomal Nanomedicines for Use in Neuroblastoma Treatment

    9.6 Development of Liposomal Formulations

    9.7 Neuroblastoma Models for In Vivo Evaluation of Liposomal Formulations

    9.8 Liposomal Formulations of Therapeutic Agents Known to be Active in Neuroblastoma Patients

    9.9 Improving Therapeutic Activity by Combination Strategies

    9.10 Future Perspectives: Developing Combination Products for Use in Patients With Neuroblastoma

    Acknowledgments

    Abbreviations

    References

    Chapter 10. Lipid nanoparticles for topical and transdermal delivery of pharmaceuticals and cosmeceuticals: A glorious victory

    Abstract

    10.1 Introduction

    10.2 Skin

    10.3 Factors Influencing the Skin Permeability of Drugs

    10.4 Dermatopharmacokinetics

    10.5 Ideal Drugs for Dermal and Transdermal Delivery

    10.6 Advantages of Topical and Transdermal Drug Delivery

    10.7 Disadvantages of Dermal and Transdermal Delivery Systems

    10.8 Case to Case Review of Topical and Transdermal Lipid Nanoparticles: Preclinical status

    10.9 Case-by-Case Review of Topical and Transdermal Lipid Nanoparticles: Clinical Status

    10.10 Patented Lipid Nanoparticles Applied in Cosmeceuticals Delivery in Skin

    10.11 Conclusion

    References

    Further Reading

    Chapter 11. Cosmetic lipid nanocarriers

    Abstract

    11.1 Introduction

    11.2 Methods of Preparation

    11.3 Characterization

    11.4 Regulatory Aspects of Cosmetic Lipid Nanocarriers

    11.5 Conclusion

    References

    Chapter 12. Self-nanoemulsifying drug delivery systems (SNEDDS) and self-microemulsifying drug delivery systems (SMEDDS) as lipid nanocarriers for improving dissolution rate and bioavailability of poorly soluble drugs

    Abstract

    12.1 Lipid Formulations/Carriers for Oral Administration of Drugs

    12.2 Transformation of Liquid SMEDDS/SNEDDS Into Solid Dosage Forms

    12.3 SNEDDS/SMEDDS Characterization

    12.4 Conslusion

    References

    Further Reading

    Chapter 13. Lipid-based nanomedicines: Current clinical status and future perspectives

    Abstract

    13.1 Lipid-Based Nanocarriers

    13.2 Lipid-Based Clinical Nanomedicines

    13.3 Ongoing Clinical Trials on Lipid-Based Formulations

    13.4 Conclusion and Future Perspective

    Acknowledgments

    References

    Further Reading

    Chapter 14. Structure and kinetics of synthetic, lipid-based nucleic acid carriers: Lipoplexes

    Abstract

    14.1 Introduction

    14.2 Equilibrium Lipoplex Structure (on the Nanoscale)

    14.3 The Large-Scale Structure of Lipoplexes (on the 100 nm–10 μm Scale)

    14.4 Lipoplex Synthesis Kinetics and Their Effect on the Macrostructure

    14.5 Lipoplex-Based Transfection

    14.6 Discussion

    14.7 Conclusion

    Conflict of Interests

    References

    Chapter 15. In vitro and in vivo characterization of pharmaceutical topical nanocarriers containing anticancer drugs for skin cancer treatment

    Abstract

    15.1 Introduction

    15.2 Skin

    15.3 Skin Cancer

    15.4 Treatment Options

    15.5 Topical Drug Delivery

    15.6 Topical Nanocarriers for Skin Cancer

    15.7 In Vitro Characterization

    15.8 Ex Vivo Characterization

    15.9 In Vivo Characterization

    15.10 Conclusion

    References

    Further Reading

    Index

    Copyright

    William Andrew is an imprint of Elsevier

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

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

    Copyright © 2018 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.

    British Library Cataloguing-in-Publication Data

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

    Library of Congress Cataloging-in-Publication Data

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

    ISBN: 978-0-12-813687-4

    For Information on all William Andrew publications visit our website at https://www.elsevier.com/books-and-journals

    Publisher: Matthew Deans

    Acquisition Editor: Laura Overend

    Editorial Project Manager: Thomas Van Der Ploeg

    Production Project Manager: Debasish Ghosh

    Cover Designer: Greg Harris

    Typeset by MPS Limited, Chennai, India

    List of Contributors

    Muneer Ahmad,     COMSATS Institute of Information Technology, Abbottabad, Pakistan

    Mursalin Ahmad,     COMSATS Institute of Information Technology, Abbottabad, Pakistan

    Atif Ali,     COMSATS Institute of Information Technology, Abbottabad, Pakistan

    Kessiane B. Almeida,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Maria Angeles Solinis Aspiazu,     University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain

    Emeli M. Araújo,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Marcel B. Bally

    British Columbia Cancer Research Centre, Vancouver, BC, Canada

    University of British Columbia, Vancouver, BC, Canada

    Centre for Drug Research and Development, Vancouver, BC, Canada

    Luigi Battaglia,     Università degli Studi di Torino, Turin, Italy

    Maria Carafa,     University Sapienza of Rome, Rome, Italy

    Anne C.A. Cardoso,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Ramesh Chandra,     University of Delhi, Delhi, Delhi, India

    Lina Chernov,     British Columbia Cancer Research Centre, Vancouver, BC, Canada

    Felisa Cilurzo,     University of Chieti-Pescara G. D’Annunzio, Chieti, Italy

    Maria C. Cristiano,     University of Catanzaro Magna Graecia, Germaneto–Catanzaro, Italy

    Nily Dan,     Drexel University, Philadelphia, PA, United States

    Ana del Pozo Rodriguez,     University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain

    Glauce C. Desmarais,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Rebecca J. Deyell

    British Columbia Children’s Hospital, Vancouver, BC, Canada

    British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada

    University of British Columbia, Vancouver, BC, Canada

    Jelena Đuriš,     University of Belgrade, Belgrade, Serbia

    Deborah Q. Falcão,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Valerie A. Ferro,     University of Strathclyde, Glasgow, United Kingdom

    Federica Foglietta,     Università degli Studi di Torino, Turin, Italy

    Marina Gallarate,     Università degli Studi di Torino, Turin, Italy

    Aurora García-Rendón,     University of Sonora, Hermosillo, Sonora, Mexico

    Adriana Garibay-Escobar,     University of Sonora, Hermosillo, Sonora, Mexico

    Roger Gilabert-Oriol,     British Columbia Cancer Research Centre, Vancouver, BC, Canada

    Dipanjana Gosh,     School of Pharmacy and Research, People’s University, Bhopal, Madhya Pradesh, India

    Vandana Gupta,     Department of Science and Technology, New Delhi, India

    Fredy R.S. Gutierrez,     Antonio Narino University, Bogotá, Colombia

    Roberto Guzmán,     University of Arizona, Tucson, AZ, United States

    Svetlana Ibrić,     University of Belgrade, Belgrade, Serbia

    Kiran Jyoti,     Sachdeva College of Pharmacy, Mohali, Punjab, India

    Amrita Kadari,     CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India

    Hira Khan,     Abbottabad University of Science and Technology, Abbottabad, Pakistan

    Marko Krstić,     University of Belgrade, Belgrade, Serbia

    Hitesh Kulhari

    CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India

    Central University of Gujarat, Gandhinagar, Gujarat, India

    Upendra Kumar Jain,     Chandigarh College of Pharmacy, Mohali, Punjab, India

    Barbara G. Lima,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Jitender Madan,     Chandigarh College of Pharmacy, Mohali, Punjab, India

    Đorđe Medarević,     University of Belgrade, Belgrade, Serbia

    Samanta C. Mourão,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Alexander B. Mullen,     University of Strathclyde, Glasgow, United Kingdom

    Elisabetta Muntoni,     Università degli Studi di Torino, Turin, Italy

    Leonor M. Nascimento,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Mohammad A. Obeid

    University of Strathclyde, Glasgow, United Kingdom

    Yarmouk University, Irbid, Jordan

    Adriana P. Oliveira,     Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil

    Donatella Paolino,     University of Catanzaro Magna Graecia, Germaneto–Catanzaro, Italy

    Mayur M. Patel,     Nirma University, Ahmedabad, Gujarat, India

    Deep Pooja,     CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India

    Om Prakash Katare,     Punjab University, Chandigarh, Punjab, India

    Sharad Prakash Pandey,     Truba Institute of Pharmacy and Research Centre, Bhopal, Madhya Pradesh, India

    Shruti U. Rawal,     Nirma University, Ahmedabad, Gujarat, India

    Paola S. Sanches,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Thamyres C. Santos,     Universidade Federal Fluminense, Niterói, RJ, Brazil

    Vaishali Sengar,     Chandigarh College of Pharmacy, Mohali, Punjab, India

    Loredana Serpe,     Università degli Studi di Torino, Turin, Italy

    Tripti Shukla,     School of Pharmacy and Research, People’s University, Bhopal, Madhya Pradesh, India

    Ramakrishna Sistla,     CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India

    Rothwelle J. Tate,     University of Strathclyde, Glasgow, United Kingdom

    Armando Tejeda-Mansir,     University of Sonora, Hermosillo, Sonora, Mexico

    Piyush Trivedi,     Rajiv Gandhi Proudyogiki Vishwavidyalaya, Bhopal, Madhya Pradesh, India

    Naveed Ullah Khan,     COMSATS Institute of Information Technology, Abbottabad, Pakistan

    Neeraj Upmanyu,     School of Pharmacy and Research, People’s University, Bhopal, Madhya Pradesh, India

    Juan D. Valderrama,     Antonio Narino University, Bogotá, Colombia

    Series Preface: Pharmaceutical Nanotechnology

    Alina M. Holban, University of Bucharest, Bucharest, Romania

    Due to its immense applicative potential, nanotechnology is considered the leading technology of the 21st century. The science and engineering of nanometer-sized materials is currently employed for the development of numerous scientific, industrial, ecological, and technological fields. Biology, medicine, chemistry, pharmacy, agriculture, food industry, and material science are the main fields which have benefited from the great technological progress developed in nanoscience.

    In the pharmaceutical field, nanotechnology has revolutionized traditional drug-design concept and the art of drug delivery. The idea of a highly specific nanoscale drug for the targeted therapy of diseases is now considered a feasible treatment for severe health conditions.

    Some scientists believe that the pharmaceutical domain has been reborn by the important contribution of nanotechnology. The field of pharmaceutical nanotechnology has the potential to offer innovative solutions for all diagnosis, therapy, and prophylaxis domains. Application of nanotechnology tools in pharmaceutical research and design is likely to result in moving the industry from a blockbuster drug model to personalized medicine. The current main focus of clinicians is to treat patients individually, not their general diagnosed diseases, which are usually difficult to diagnose or incorrectly diagnosed. There are compelling applications in the pharmaceutical industry where suitable nanotechnology tools can be successfully utilized. By designing and modifying drugs at nanoscale, pharmaceutical nanotechnology could be useful not only for the development of completely new therapeutic solutions, but also to add value to existing products. This possibility opens perspectives of success for pharmaceutical companies in existing markets, but also for new markets.

    Scientists have manifested an impressive interest on the field of pharmaceutical nanotechnology research in recent years. However, we face today a true dilemma of data unavailability, due to the multitude of existing information which can be highly inaccurate and contradictory. This is because of the lack of an efficient model for sorting the plethora of nanotechnology tools and information that exists, and strategically correlate those with potential opportunities into different segments of pharmaceutical research and design.

    This series is trying to cover the most relevant aspects regarding the great progress of nanotechnology in the pharmaceutical field and to highlight the currently emerging trend of pharmaceutical nanotechnology towards the personalized medicine concept.

    The 10 volumes of this series are structured to wisely offer relevant information regarding basic concepts and also to reveal the newest approaches and perspectives in pharmaceutical nanotechnology.

    Nanoscale Fabrication, Optimization, Scale-Up and Biological Aspects of Pharmaceutical Nanotechnology, introduces the readers into the amazing field of nanoscale design. Also, this volume facilitate understanding of the biological requirements of nanostructured pharmaceutical formulations for advanced drugs.

    In Design and Development of New Nanocarriers, the most recent progress made on the field of nano-delivery is discussed. Modern nanostructured drug carriers employ innovative solutions for the detection and treatment of various diseases in a personalized and efficient manner.

    Design of Nanostructures for Theranostics Applications, highlights the impressive impact of nanotechnology in the development of combined diagnosis and therapy concept: theranostics.

    Design of Nanostructures for Versatile Therapeutic Applications, offers a dynamic solution for immune modulation, treatment of diseases by natural-based products and infection control, while employing nanostructured solutions to achieve top results.

    Nanostructures for the Engineering of Cells, Tissues and Organs: From Design to Applications, is a highly investigated and debated field; tissue engineering, is dissected through this volume. Here is shown how nanotechnology has advanced research and applications in the manipulation and engineering of cells and tissues in vitro.

    Organic Materials as Smart Nanocarriers for Drug Delivery, deals with the specific world of organic nanomaterials, revealing their wide applications, types, and advantages in drug delivery.

    In the volume entitled: Inorganic Frameworks as Smart Nanomedicines, the main focus is to discuss the variety and properties of inorganic nanostructures for therapy and drug delivery in the context of improved personalized medicine.

    Lipid Nanocarriers for Drug Targeting, deals with recently developed lipid nanostructures and the advances made in drug targeting.

    Drug Targeting and Stimuli-Sensitive Drug Delivery Systems, dissects smart stimuli-responsive nanosystems employed to specifically detect various biochemical conditions and control the release of drugs.

    Fullerenes, Graphenes and Nanotubes, reveals major findings made on widely applied drug-design nanosystems, namely fullerens, graphenes and nanotubes. The impact of these nanostructures in pharmaceutical research is highlighted.

    All 10 volumes are nicely illustrated and chapters are organized into a logical manner to be accessible to a wide audience. The series is a valuable resource of new and comprehensive scientific proof on the intriguing and emerging field of pharmaceutical nanotechnology, which could be of a great use for scientists, engineers, pharmaceutical representatives, clinicians, and any non-specialist interested user.

    Preface

    Lipid Nanocarriers for Drug Targeting

    The aim of this book is to present the recent progress in the field of lipid nanocarriers, especially for drug delivery and targeting. Different types of lipid nanocarriers are overviewed with pro and cons, covering formulation, pharmacokinetic aspects, in vitro and in vivo activity, and also a comprehensive list of biomedical applications is provided. Special attention was paid to their synthesis methods, properties and delivery opportunities, highlighting recent trends on the field.

    The book Lipid Nanocarriers for Drug Targeting, contains 15 chapters, prepared by outstanding researchers from United States, Serbia, Pakistan, Canada, United Kingdom, Jordan, Spain, México, Colombia, Italy, Brazil, and India.

    Chapter 1, Lipid nanocarriers, prepared by Tripti Shukla et al., presents a special concern related to various types of lipid nanocarriers, their detailed description, composition, different methods of preparation, influence of various types of lipids on the different properties of such carriers. It also covers the various physicochemical, formulation, pharmacokinetic, and cytotoxic aspects of such carriers. Furthermore, it also includes the marketed formulations of lipid nanocarriers.

    Chapter 2, Lipid nanoparticulate systems: modern versatile drug carriers, prepared by Shruti U. Rawal and Mayur M. Patel, focuses on two major types of lipid nanoparticulate systems, viz. solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs). This chapter comprehensively describes the advantages offered by SLNs/NLCs, structure, type, and composition of SLNs/NLCs, methods employed for formulating SLNs/NLCs, characterization as well as safety and toxicological aspects of SLNs/NLCs. Further applications by different routes of administration are also discussed.

    Chapter 3, Nanotechnology in phytotherapy: current challenges of lipid-based nanocarriers for the delivery of natural products, prepared by Deborah Q. Falcão et al., reviews the application of lipid nanocarriers which load natural products, specifically from plants. This chapter also presents the latest applications and advantages of each lipid system as a carrier, as well as methods of obtention and characterization, regarding the particularities of natural products. It provides a systematic review of different approaches and assists the research and development of new products.

    Chapter 4, Innovative vesicles for dermal and transdermal drug delivery, prepared by Maria C. Cristiano et al., gives an up-to-date overview about the principal innovative elastic vesicles used for the improvement of dermal and transdermal administration of the different classes of drugs. The correlation between their composition and their ability to improve the permeation through the skin is also discussed. Classic liposomes, ethosomes, transferosomes, and niosomes represent the present and the future of drug delivery for the treatment of skin diseases.

    Chapter 5, Using bacterial outer membrane vesicles in vaccine design, prepared by Juan D. Valderrama and Fredy R.S. Gutierrez, reviews the current techniques for engineering Escherichia coli cells in order to produce proteins/antigens and encapsulate them into outer membrane vesicles for vaccine development and other applications in immunology.

    Chapter 6, Plasmid-DNA lipid nanovaccines: an innovative approach for a better world health, prepared by Aurora García-Rendón et al., provides a summary of the advances and major challenges associated with the fusion of plasmid DNA (pDNA) production technology and the nanoparticle delivery technologies to produce pDNA lipid nanovaccines.

    Chapter 7, Ocular delivery of solid lipid nanoparticles, prepared by Luigi Battaglia et al., presents the recent progress related to the therapeutic efficiency, compliance, and safety of ocular drugs, administered either to the anterior, or posterior segment of the eye, that can benefit from a nanotechnology approach. Muco-adhesion, with consequent increase of retention at the cornea, as well as permeation enhancement are the most important challenges for topical lipid nanoparticles delivery to the anterior segment of the eye.

    Chapter 8, Lipid-based nanoparticles for cancer treatment, prepared by Mohammad A. Obeid et al., highlights the main features of these nanoparticles and describes their types and classes. The contributors examine some of the approved formulations and some of those currently investigated in clinical trials. In addition, they discuss the use of lipid-based nanoparticles in the delivery of siRNA. These nanoparticles promise novel and better anticancer medications development for clinical cancer treatment.

    Chapter 9, Developing liposomal nanomedicines for treatment of patients with neuroblastoma, prepared by Roger Gilabert-Oriol et al., focuses on lipid-based formulations in part because liposomal formulations represent a clinically advanced technology. Several murine models of neuroblastoma are described, as these models are critically important to establish the potential therapeutic value of the candidate formulations. A literature search was completed to identify liposomal formulations that have been evaluated in the clinics and that are loaded with drugs with an established role in treating patients with neuroblastoma. Furthermore, the authors give examples of combination strategies involving liposomal formulations and other therapeutically active agents. The challenges of advancing such combinations towards clinical trials for patients with neuroblastoma are discussed.

    Chapter 10, Lipid nanoparticles for topical and transdermal delivery of pharmaceuticals and cosmeceuticals: a glorious victory, prepared by Vaishali Sengar et al., provides an overview of lipid nanoparticles intended for topical and transdermal delivery of pharmaceuticals and cosmeceuticals. Special prominence is given to classification and methods for preparation of lipid nanoparticles at industrial scale. Moreover, attention has also been paid on diseases of skin and pathways for transportation of lipid nanoparticles through skin. The chapter was concluded with a list of representative patented lipid nanoparticles in the field of cosmeceuticals.

    Chapter 11, Cosmetic lipid nanocarriers, prepared by Atif Ali et al., discusses cosmetic lipid nanocarriers and gives an overview of the stability of integrated substances, loading capacity, controlled/triggered release, occlusivity, film formation, and penetration through the skin. The production technology and related issues including large and small scale production, regulatory aspects, status of the excipients and physical and chemical stability are described.

    Chapter 12, Self-nanoemulsifying drug delivery systems (SNEDDS) and self-microemulsifying drug delivery systems (SMEDDS) as lipid nanocarriers for improving dissolution rate and bioavailability of poorly soluble drugs, prepared by M. Krstić et al., reviews the current state and recent progress in SMEEDS and SNEEDS formulation development, with particular emphasis on the approaches for overcoming their limitations and enabling wider commercial application of these lipid nanocarriers.

    Chapter 13, Lipid-based nanomedicines: current clinical status and future perspectives, prepared by Deep Pooja et al., describes the lipid-based clinical nanomedicines marketed for the treatment of cancer, age-related molecular degeneration, fungal infection, viral infection and pain management. Finally, the contributors enlighten the ongoing clinical trials on lipid-based nanomedicines.

    Chapter 14, Structure and kinetics of synthetic, lipid-based nucleic acid carriers: lipoplexes, prepared by Nily Dan, gives an up-to-date overview about lipoplexes which are characterized by two lengths. On the local scale, namely, 10–50 nm, the lipid and nucleic acid arrangement is set by thermodynamic equilibrium and is directly linked to the molecular properties of the lipids and the concentration of nucleic acid. On larger scales, of 100 nm–1 μm, lipoplex characteristics are set by the kinetics of the synthesis process, following a three-step process.

    Chapter 15, In vitro and in vivo characterization of pharmaceutical topical nanocarriers containing anticancer drugs for skin cancer treatment, prepared by Vandana Gupta and Piyush Trivedi, provides a basic understanding and description of the strategies that can be used to overcome skin cancer. These modalities are discussed in the context of their characterization and application for promoting and targeting the delivery of skin tumor drugs following topical, noninvasive administration.

    Chapter 1

    Lipid nanocarriers

    Tripti Shukla¹, Neeraj Upmanyu¹, Sharad Prakash Pandey² and Dipanjana Gosh¹,    ¹School of Pharmacy and Research, People’s University, Bhopal, Madhya Pradesh, India,    ²Truba Institute of Pharmacy and Research Centre, Bhopal, Madhya Pradesh, India

    Abstract

    Lipid nanocarriers have emerged as a very promising, emerging and rapidly developing tool for the delivery of various drugs lacking solubility, bioavailability and stability in the recent couple of decades. Recent studies show that about 40% of newer drugs have such problems. Initially, a lipid carrier was denoted by the liposome and similar vesicular systems, but currently they are categorized as colloidal nano lipid-based carriers (CNLBC). To avoid the limitation of these CNLBCs in pH- and enzyme-dependent degradation, especially when taken orally or in physical0 and chemical-related stability issues, newer lipid nanocarriers such as solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLCs), lipid drug conjugates (LDCs), and pharmacosomes have shown their importance at greater extent due to low toxicity, improved bioavailability, high biocompatibility, high drug-loading efficiency, protection from degradation in GIT, etc. Lipid nanocarriers can load both hydrophilic and lipophilic drug. Solubility is a rate-limiting step in the case of lipophilic drugs (BCS Class II and IV), which can be greatly modified by formulation of lipid nanocarriers. Similarly, lipidic nanocarriers can increase the permeability of most of the hydrophilic drugs (BCS I and III class) which is the rate limiting step this case. These carriers also shows good controlled and target specific drug delivery system which always attracts the attention of researchers.

    The current chapter aims to present a special concern related to various types of lipid nanocarriers, their detailed description on composition, different methods of preparation, influence of various types of lipids on the different properties of such carriers. It also covers the various physicochemical, formulation, pharmacokinetic, and cytotoxic aspects of such carriers. Furthermore, it includes the marketed formulations of lipid nanocarriers with their company name and trade name.

    Keyworlds

    Lipid nanocarriers; liposome; ethosome; transfersome; SLN; NLC; LDC

    Chapter Outline

    1.1 Introduction to Lipid Nanocarriers 2

    1.1.1 Classification: (Mehanna et al., 2012) 3

    1.2 Liposome 3

    1.2.1 Introduction 3

    1.2.2 Classification Of Liposomes x 4

    1.2.3 Composition 4

    1.2.4 Method of Preparation of Liposomes 5

    1.2.5 Characterization 8

    1.3 Ethosomes 8

    1.3.1 Introduction 8

    1.3.2 Composition  8

    1.3.3 Method of Preparation of Ethosomes 8

    1.3.4 Characterization 11

    1.4 Transfersomes 11

    1.4.1 Introduction 11

    1.4.2 Composition 13

    1.4.3 Method of Preparation of Transfersomes 13

    1.4.4 Characterization 14

    1.5 Solid Lipid Nanoparticles 15

    1.5.1 Introduction 15

    1.5.2 Classification 15

    1.5.3 Composition 17

    1.5.4 Method of Preparation of Solid Lipid Nanoparticle 18

    1.5.5 Characterization 20

    1.6 Nanostructured Lipid Carriers (NLC) 21

    1.6.1 Introduction 21

    1.6.2 Classification 21

    1.6.3 Composition 22

    1.6.4 Methods for Preparation Nanostructured Lipid Carriers 23

    1.6.5 Chacterization 24

    1.7 Lipid Drug Conjugates (LDC) 24

    1.7.1 Introduction 24

    1.7.2 Composition 24

    1.7.3 Method of Preparation of Lipid Drugs Conjugates 24

    1.7.4 Characterization 27

    1.8 Marketed or Commercially Available Nanolipid Products 27

    1.9 Applications of Various Lipid Nanocarriers 28

    1.9.1 Applications of Liposomes 28

    1.9.2 Applications of Solid Lipid Nanoparticles 32

    1.9.3 Application of Nanostructured Lipid Carriers 35

    1.9.4 Application of Lipid Drug Conjugates 37

    1.10 Conclusion 38

    1.11 Future Prospects 38

    References 39

    Further Reading 47

    1.1 Introduction to Lipid Nanocarriers

    The prefix nano comes from the Greek word nanos, meaning dwarf. Prof. Norio Taniguchi at Tokyo Science University coined the term Nanotechnology in 1974. This term is further used by Drexler in 1986 in his book Engines of creation: The Coming Era of Nanotechnology (De Villiers et al., 2008).

    Various novel drug delivery systems prepared by polymers and solvents present in the market are still very limited in use because of their high production cost, high polymer cost, toxicity, and allergy of polymer and solvent in the body (Muller and Keck, 2004).

    To overcome these problems related with novel drug delivery systems prepared by polymer and solvent, many researchers have diverted their interest to lipid-based nanocarrier systems, such as liposomes, transfersomes, ethosomes, solid lipid nanoparticles (SLN), nanostructured lipid carriers, lipid drug conjugates, etc. (Chen et al., 2010).

    In the last decade, lipids have gained much interest as carriers for the delivery of drugs with poor water solubility. If any therapeutic agent is added into it, therapeutic effectiveness of drugs will be maximized (Pouton, 2006).

    Lipid-based nanocarriers are an acceptable approach and have gained importance in the current era because of their various notable properties, such as low toxicity, improved bioavailability, high biocompatibility, high drug-loading efficiency, high protection from degradation in the gastro intestinal tract (GIT), controlled-release behavior, easy to scale-up and sterilize, easy to validate, and also because of their suitability for drug delivery by various routes and sites of administration (Liu et al., 2010b; Chime et al., 2013).

    Lipid nanocarriers can load both hydrophilic and lipophilic drugs, and also be administered by various routes such as oral, topical, ocular, parenteral, and pulmonary routes. Solubility is a rate-limiting step in the case of lipophilic drugs (BCS Class II and IV), which can be greatly modified by formulation of lipid nanocarriers. Similarly, lipidic nanocarriers can also increase the permeability of most of the hydrophilic drugs (BCS Class I and III), which is the rate-limiting step this case. Novel lipid excipients used for the development of lipid nanocarriers play a very important and vital role for enhancing the therapeutic effect of various drugs (Jannin et al., 2008).

    Various lipids used for the preparation of lipid nanocarriers are those which are biodegradable and showing biocompatibility in physiological media or biological fluid. These lipids will also be discussed in this chapter (Müller et al., 1996).

    1.1.1 Classification: (Mehanna et al., 2012)

    Very commonly studied lipid nanocarriers are classified as follows:

    Lipid Nanocarriers

    Vesicular approaches:

    Liposome

    Ethosomes

    Transfersomes

    Nonvesicular approaches:

    Solid lipid nanoparticles (SLNs)

    Nanostructured lipid carriers (NLC)

    Lipid drug conjugates (LDC)

    1.2 Liposome

    1.2.1 Introduction

    The name liposome is derived from two Greek words: lipos meaning fat and soma meaning body. Liposomes were first produced in England at the Babraham Institute in Cambridge in 1961, by British hematologist Dr. Alec D. Bangham who was studying phospholipids and blood clotting (Bangham et al., 1962). Liposomes are concentric bilayered vesicles in which an aqueous volume is entirely enclosed by a membranous lipid bilayer mainly composed of natural or synthetic phospholipids. A liposome can be formed at a variety of sizes, such as unilamellar or multilamellar. Liposomes are also known as artificially prepared vesicles which are made up of a lipid bilayer along with filled drug (Mansoori et al., 2012). The diameter of a liposome varies from 0.02 to 10 μm. These are tremendously studied by researchers due to their versatile nature (Bangham et al., 1962; Mansoori et al., 2012) (Fig. 1.1).

    Figure 1.1 Liposome.

    1.2.2 Classification Of Liposomes x

    Liposomes can be classified by various methods, it will be represented by pictorial representation in Fig. 1.2 (Kulkarni et al., 2011; Vyas and Khar, 2004; Jain and Jain, 1997).

    Figure 1.2 Classification of liposomes.

    1.2.3 Composition

    Main excipients used for the preparation of liposomes are Phospholipids (Natural or Synthetic), steroids, polymeric materials, solvents etc. These excipients with their uses in liposome preparation are summarized in given Table 1.1 (Himanshu et al., 2011).

    Table 1.1

    Various lipids used for the preparation of liposomes (Mansoori et al., 2012).

    Liposomes can be prepared from a variety of lipids or mixture of lipids. Phospholipids are very commonly used. Few examples are as follows:

    Phospholipids: These are derived from phosphatidic acid. Phospholipids containing glycerol are used for the preparation of liposomes. Glycerol is the backbone of phospholipid moiety. Saturated fatty acids are used for the preparation of stable liposomes. Mainly phasphotidyl choline (lecithin) phospholipids are used for the preparation of liposome. These are extracted from egg yolk, soya bean, and, in lesser amount, from bovine heart and spinal cord. They are used as principal phospholipid in the preparation of liposome because of their relatively low cost, neutral charge, and less reactive nature.

    Sphingolipids: These lipids are derived from sphingosine. Sphingosine is a very important component of plant and animal cell. It is incorporated in the liposomes to provide a surface charge layer on liposomes.

    Sterols: These are added to the liposome for reducing the permeation of lipid membrane to water soluble molecule, to stabilize the membrane, and to reduce the fluidity of lipid bilayer.

    Polymeric materials: These materials are formed by polymerization of synthetic phospholipids with diactylenic group in the hydrocarbon chain when exposed to UV light. Liposomes formed by these materials show a significantly higher permeability barrier for entrapped water soluble drugs.

    1.2.4 Method of Preparation of Liposomes

    Basic steps involved for the preparation of liposomes are:

    First step—Drying down lipids from organic solvent.

    Second step—Dispersion of lipid in aqueous media.

    Third step—Purification of the resultant liposome.

    Fourth step—Analysis or characterization of the final product.

    Common methods used for the preparation of liposomes are discussed in the following sections.

    1.2.4.1 Hydration method (Bangham Method)

    This method is mainly used for the preparation of multilamellar large vesicles (MLVs) type of liposomes. This is an widely used method for the preparation of liposomes. Chloroform and methanol mixture is used as solvent in to a 250 mL round-bottomed flask for the dissolution of lipid mixture and charged components. This flask is attached to a rotary evaporator connected with vacuum pump and rotated at 60 rpm speed for 30 minutes at about 30°C. At this temperature, organic solvents are evaporated by leaving a dry lipid residue at the bottom of the flask. Nitrogen is then introduced in the flask after detachment of vacuum pump. Solvent residue is removed from the flask by lyophilization technique. The flask is then flushed with nitrogen and 5 mL of phosphate buffer. The flask is again rotated until the removal of all the lipids from the flask. The formed white milky suspension is kept for 2 hours for the completion of swelling process to give liposomes (Akbarzadeh et al., 2013).

    1.2.4.2 Ethanol injection method

    In this method, an ethanol solution of the lipids is rapidly injected directly into an excess of saline or other aqueous medium through a fine needle. The ethanol is diluted in water and phospholipids molecules are dispersed evenly through the medium. This procedure yields a high proportion of small unilamellar vesicles (SUVs). The ethanol injection method is very simple method for the preparation of liposomes (Riaz, 1996).

    1.2.4.3 Ether injection

    This method is similar to the ethanol injection method. It involves injecting the immiscible organic solution very slowly into an aqueous phase through a narrow needle at the temperature of vaporizing of organic solvent. In this method, the lipids are carefully treated and there is a risk of oxidative degradation (Riaz, 1996).

    1.2.4.4 Sonication

    This method reduces the size of the vesicles and imparts energy to a lipid suspension. This can be achieved by exposing the MLV to ultrasonic irradiation.

    There are two methods of sonication:

    1. Using bath sonicator.

    2. Using probe sonicator.

    Probe sonication is used for suspensions which require high energy in small volume. The bath sonicator is used for a large volume of dilute lipids. The disadvantage of the probe sonicator is contamination of the preparation with metal from the tip of the probe. By this method, small unilamellar vesicles are formed and they are purified by ultracentrifugation (Troy and Beringer, 2006).

    1.2.4.5 Microemulsification method

    In the microemulsification method, microfluidizer equipment is used to prepare small vesicles from a concentrated lipid suspension. The lipids can be introduced into the fluidizer as a suspension of large MLVs. The equipment pumps the fluid at very high pressure through a 5 μm screen. It is then forced into long micro channels, in which two streams of dried fluids collide together at right angles with high velocity. The fluid collected can be recycled through the pump and interaction chamber until vesicles of spherical dimensions are obtained (Troy and Beringer, 2006).

    1.2.5 Characterization

    Liposomes can be prepared by any one of the above described method. After preparation of liposomes, they are evaluated by various parameters. These parameters are summarized in Table 1.2 (Kaur et al., 2013; Lasic, 1997).

    Table 1.2

    1.3 Ethosomes

    1.3.1 Introduction

    Ethosomes are also called ethanolic liposomes. Ethosomes are high alcohol–containing lipid-based elastic vesicular systems that enhance topical drug delivery (Pandey, 2011). Dissolution of the lipids and drug in ethanol (20%–45%) takes place for the preparation of ethosomes (Dayan and Touitou, 2000). Ethosomes are able to improve in vitro and in vivo skin delivery of various drugs, both under occlusive and nonocclusive conditions, and is proven by many researchers (Prasanthi and Lakshmi, 2012). Hydrophilic, lipophilic, and amphiphilic drugs can be incorporated in all type of ethosomes (Bhalaria et al., 2009). The size range for ethosome may vary from tens of nanometers to microns (μ) (Patel, 2007) (Fig. 1.3).

    Figure 1.3 Ethosome.

    1.3.2 Composition

    Ethosmes are prepared by using phospholipid, polyglycol, alcohols, and cholesterol. A few examples of excipients with their uses are summarized in given Table 1.3 (Touitou et al., 1996).

    Table 1.3

    1.3.3 Method of Preparation of Ethosomes

    Ethosomal formulation can be prepared by methods as described further in the section. All the methods are convenient, do not require any sophisticated equipment, and are also easy to scale up to industrial level.

    1.3.3.1 Cold method

    In this method, ethanol is used for the dissolution of phospholipid, drug and other lipid materials at room temperature in a covered vessel, by vigorous stirring with the use of stirrer. Polyols or propylene glycol is added during stirring. This mixture is heated up to 30°C in a water bath. Hot water of 30°C is added to the mixture and stirred for 5 min in a covered vessel. Sonication or extrusion method is used for decreasing the size of ethosomal vesicle up to the desired extend. A refrigerator is used for the storage of ethosomes (Touitou et al., 1996).

    1.3.3.2 Hot method

    In this method, water is heated up to 40°C in a water bath and phospholipid is dispersed in the water by heating until a colloidal solution is obtained. Ethanol and propylene glycol are mixed and heated up to 40°C in a separate vessel. The organic phase is then added to the aqueous one. Ethosomes are formed after the stirring of mixture (Touitou, 1998).

    1.3.3.3 Classic mechanical dispersion method

    In this method, drug and phospholipids are dissolved in 3:1 ratio of chloroform:ethanol. The organic solvent is then removed by rotary vacuum evaporator. Finally, hydration is done to achieve ethosomes (Jaiswal et al., 2016).

    1.3.3.4 Classic method

    In this method, ethanol is used for dissolution of phospholipid and drug at 30°C. To this, double-distilled water is added and stirred at 700 rpm. The resulting vesicles are homogenized by passing through a polycarbonate membrane.

    1.3.4 Characterization

    As the ethosomes are prepared these are evaluated by various parameters shown and summarized in Table 1.4 (Zhaowu et al., 2009).

    Table 1.4

    1.4 Transfersomes

    1.4.1 Introduction

    The term transfersome (elastic vesicles) and their underlying concept were introduced in 1991 by Gregor Cevc (Prajapati et al., 2011). Transfersome is a term registered as a trademark by the German company IDEA AG, and used to refer to its proprietary drug delivery technology. The word transfersome means carrying body, and is derived from the Latin word transferre, meaning to carry across, and the Greek word soma, for body (Cevc, 2004). In the broadest sense, a transfersome is an highly adaptable, stress-responsive, and complex aggregate. Transfersomes are self-regulating and self-optimizing vesicles possessing an aqueous core surrounded by a complex lipid bilayer. Transfersomes are also considered ultradeformable liposomes. This enables the transfersome to cross various transport barriers efficiently and then act as a drug carrier for noninvasive targeted drug delivery and sustained release of therapeutic agents (Prajapati et al., 2011). Transfersomes penetrate the stratum corneum, i.e., skin, by either intracellular route or the transcellular route. Hydrostatic gradient is responsible for movement of intact vesicles from skin (Cevc, 2004).

    1.4.2 Composition

    Transfersomes are prepared by using various excipients, such as phospholipids, surfactants, alcohols, buffering agents, etc. These are summarized in Table 1.5 with examples and their uses.

    Table 1.5

    1.4.3 Method of Preparation of Transfersomes

    Following techniques have been generally used to prepare transfersomes (Singh, 2013; Venkatesh et al., 2014; Miatmoko et al., 2015; Cevc and Chopra, 2016a,b).

    1.4.3.1 Vortexing-sonication method

    In the vortexing-sonication method, phospho-tidylcholine, ethyl acetate, and therapeutic agents are blended in a phosphate buffer and vortexed to attain a milky suspension. The suspension is sonicated, followed by extrusion through poly-carbonate membranes. Cationic transfersomes have also been prepared by this method, which involves mixing cationic lipids, such as N-[1-(2, 3-dioleyloxy) propyl]-N,N,N-trimethyl ammonium chloride, with phosphate buffered saline (PBS) to obtain a concentration of 10 mg/mL, followed by the addition of sodium deoxycholate. The blend is vortexed and sonicated, followed by extrusion through a polycarbonate (100-nm) filter.

    1.4.3.2 Rotary evaporation-sonication method

    The rotary evaporation-sonication method involves dissolution of phosphatidylcholine and Ethyl acetate (EA) in a blend of chloroform and methanol (2:1, v/v), followed by the removal of organic solvent using rotary evaporation under reduced pressure at 50°C. The film deposited is hydrated with a solution of the therapeutic agent in a suitable aqueous phase while rotating the flask for one hour at room temperature. The vesicles produced are left to swell for two hours at room temperature, followed by 30 min of sonication in a bath sonicator so as to decrease their volume. Extrusion of vesicles then occurs through a sandwich of 100- and 200-nm polycarbonate membranes.

    1.4.3.3 Modified hand-shaking method

    In this method phospholipid, edge activator, and the drug are taken in a clean, dry, round bottom flask, and then lipid mixture dissolved in 1:1 v/v chloroform: methanol. The organic solvent was evaporated by using hand-shaking method until completely dry. The deposited lipid film was hydrated with PBS (pH 7.4) by rotation in a reverse direction for 1 hour at room temperature and the resulting vesicles were swollen for 2 hours at room temperature to achieve large multilamellar vesicles.

    1.4.4 Characterization

    After the development of transfersome, vesicles are evaluated by various parameters. These parameters are summarized in Table 1.6 (Kong et al., 2015; Cevc and Chopra, 2016a,b; Ferreira et al., 2016).

    Table 1.6

    1.5 Solid Lipid Nanoparticles

    1.5.1 Introduction

    SLNs are colloidal carriers ranging from 50 to 1000 nm prepared by dispersion of physiological lipid into water or in aqueous solution of surfactant solution. These nanolipid carriers were discovered in 1991 as an alternative carrier system to older carrier systems, such as emulsions, liposomes, and polymeric nano- and microparticles. SLN are gaining a great importance because of their unique properties, such as large surface area, high drug-loading capacity, controlled release action (fast or sustained), site-specific targeting and increased bioavailability of any therapeutic agent. Major components used for the preparation of SLN are lipids and surfactants. Lipids used for SLN preparation must be biocompatible, biodegradable, and solid at room temperature. Nonionic surfactants are used for the stability of SLNs (Kesharwani et al., 2016; Patil et al., 2016; De La Torre and De Pinho, 2015; Jafar et al., 2015) (Fig. 1.4).

    Figure 1.4 Solid lipid nanoparticle.

    1.5.2 Classification

    SLNs are broadly classified into three types, depending on nature and type of lipids, chemical nature and structure of drug, solubility of active ingredient in the lipid, type and concentration of surfactant, temperature during production, and production method used (Rawat et al., 2010) (Fig. 1.5).

    Figure 1.5 Classification of solid lipid nanoparticles.

    1.5.2.1 Solid lipid nanoparticle Type I or homogenous matrix model

    Type I SLNs are prepared by cold homogenization method. In this method, solid drug is dispersed in melted solid lipid. After cooling, solidification of the lipid drug combination takes place. This solidified complex is grounded in its solid state to provide uniform drug distribution in SLN.

    1.5.2.2 Solid lipid nanoparticles, Type II or drug-enriched shell model

    Type II SLNs are produced by hot homogenization method. The lipid is melted by increasing the temperature of the system and drug is dispersed in the melted lipid. During the cooling of hot o/w nanoemulsion system, lipid is solidified first and deposited on the outer layer of shell. This concentrated outer layer of shell shows a burst effect on drug release. Concentration of drug on outer shell can be modified as per requirements.

    1.5.2.3 Solid lipid nanoparticles, Type III or drug-enriched CORE model

    Type III SLNs are those in which active pharmaceutical ingredient is concentrated in the center of SLNs. Drug concentration is very high and near to its saturation in the lipid melt. When this highly concentrated lipid cooled down, solubility of drug in lipid melt will be reduced and deposited on the center of the SLN leads to formation of drug enriched core.

    1.5.3 Composition

    SLN are prepared by a combination of lipids, fatty alcohol, wax, triglycerides and surfactants. These are compiled in Table 1.7 (Reddy et al., 2015; Chen et al., 2015; Srivastava and Khan, 2016).

    Table 1.7

    Major excipients used for the preparation of SLNs are:

    Compritol ATO 888: These lipids are widely used for the preparation of SLN because of various advantages such as stability, sustained release effect, and better lymphatic targeting. Combination of compritol with cocao butter, resulting high entrapment efficiency of saquinavir drug, is increased when SLN is prepared (Reddy et al., 2015; Chen et al., 2015).

    Glyceryl monostearate (GMS) & glyceryl monooleate (GMO): Glyceryl monostearate is generally used for the preparation of cosmeceuticals or cosmetics. The melting point for GMS is 50–55°C. Glyceryl monooleate is obtained by the esterification of glycerides of oleic acid and its melting point is 35–45°C. The drug entrapment efficiency of GMS is higher than GMO (Ekambaram and Sathali, 2011). Both agents GMS and GMO are prepared by the process of esterification which develops hydrophobicity to the glycerides and increases the lymphatic uptake of glycerides (Kumar et al., 2007).

    Fatty acids: Tetradecanoic acid, stearic acid, and palmitic acid are used for the preparation of SLN. Stearic acid shows the highest entrapment efficiency because of the highest length of its fatty acid chain (Rawat et al., 2010).

    Fatty alcohol: Fatty alcohols used for the preparation of SLNs are cetyl alcohol and stearyl alcohol. These elements are metabolized by fatty alcohol dehydrogenase (FADH) present in the liver (Dong and Mumper, 2006).

    Wax: Cacao butter, carnuba wax, cetyl palmitate, and beeswax are mainly used for the preparation of SLN. When cacoa butter is compared with beeswax, it shows more lipophilic character and higher drug release rate. Cacoa butter is obtained from theobroma cacao and shows lower animal toxicity and higher biocompatibility than the semisynthetic lipids. Cetyl palmitate shows better stability, lower in vitro toxicity, and better in vitro degradation rate, compared to compritol and glycerides (Kim et al., 2005).

    1.5.4 Method of Preparation of Solid Lipid Nanoparticle

    SLNs are prepared from various methods, as discussed further (Mukherjee et al., 2009; Bhattacharjee, 2013; Ekambaram et al., 2012).

    1.5.4.1 High pressure homogenization (HPH)

    As the name indicates, high pressure homogenization is a technique in which high pressure (100–2000 bar) is applied for the production of SLNs. In this technique, a fluid accelerates at very high speed, i.e., more than 1000 Km/h. Due to high pressure and speed, particles break into the submicron size range. Two approaches, cold and hot homogenization, are applied on the same principle, i.e., mixing of drug with melted lipid.

    1.5.4.1.1 Hot homogenization

    This technique is applied at higher temperature, i.e., more than the melting point of lipids. A high shear mixing device is used for the preparation of preemulsion of aqueous emulsifier phase and of the drug-loaded lipid melts. The principle behind this technique is that, as the temperature of the system increases, particle sizes of the lipid globules decrease because of the low viscosity of the inner phase. SLN of heat-sensitive drugs cannot be prepared by this method because they can be destroyed at high temperature.

    1.5.4.1.2 Cold homogenization

    SLN of heat-sensitive drugs and lipids can be prepared by this cold homogenization technique. The purpose behind the development of cold homogenization is to overcome the problems following hot homogenization. Firstly in this technique the lipid melt containing drug is cooled and ground to lipid microparticles. These lipid microparticles are dispersed in cold surfactant solution to yield presuspension. This presuspension is homogenized at or below the room temperature and gravitational force breaks the lipid microparticles into SLN.

    1.5.4.2 Ultrasonication/high speed homogenization

    In this method, a probe sonicator or bath sonicator is used for the preparation of SLNs. Energy waves are required for the breakdown of lipid drug mixture in to nanosized range.

    1.5.4.3 Solvent evaporation

    In this method, lipidic materials are dissolved in an organic solvent that is immiscible with water, such as cyclohexane. This organic solvent is then emulsified with aqueous phase. Evaporation of the solvent leads to precipitation of lipids in the aqueous media and subsequent formation of nanoparticles of 25 nm mean size were prepared. The solvent is evaporated under reduced pressure (40–60 mbar).

    1.5.4.4 Solvent emulsification-diffusion method

    The major advantage of this method is the preparation of SLNs of thermo-sensitive drugs, because heat is not required. SLN of 30–100 nm diameter can be obtained by this technique.

    1.5.4.5 Supercritical fluid method

    This technique is also known as solventless technique of SLN production. Liquid carbon dioxide is a good choice of solvent for SLN preparation by this technique. Rapid expansion of supercritical carbon dioxide solutions (RESS), particle from gas saturated solution (PGSS), aerosol solvent extraction solvent (ASES), or supercritical fluid extraction of emulsions (SFEE) is required for the preparation of SLNs by this method.

    1.5.4.6 Microemulsion-based method

    In this method of SLN preparation, dilution of microemulsion is done. A microemulsion is a transparent mixture composed of a low-melting fatty acid, an emulsifier, coemulsifier and water. Hot microemulsion (i.e., 65–70°C) is dispersed in cold water under stirring for the preparation of SLNs. To facilitate rapid lipid crystallization and to prevent aggregation of lipid molecules, high temperature gradient is needed.

    1.5.4.7 Spray drying method

    If the melting point of a lipid is high (i.e., more than 70°C) this spray drying technique is used for SLN preparation. For good results, 20% solution of trehalose is used in an ethanol-water mixture and 1% solution of trehalose in water for spray drying.

    1.5.4.8 Double emulsion method

    In this technique, w/o/w double emulsion is prepared; drug dispersed in emulsion must be encapsulated with stabilizer to prevent partitioning of it into the external water phase of emulsion during solvent evaporation.

    1.5.4.9 Precipitation method

    In this method, an organic solvent (e.g., chloroform or ethanol) is used to dissolve lipids and the prepared solution is emulsified with an aqueous phase. Organic solvent from this emulsion is evaporated and the dissolved lipid is precipitated to form SLN.

    1.5.4.10 Film-ultrasound dispersion

    In this method, lipid and drug is dissolved in organic solvent. After rotation and evaporation of organic solvent, a lipid film is deposited inside the round bottom flask. This lipid layer is hydrated by aqueous solution. By using ultrasound waves, SLNs of uniform size are then formed.

    1.5.5 Characterization

    After preparation of SLNs by any suitable method these SLN are evaluated by following parameters as shown in Table 1.8 (Waghmare et al., 2012).

    Table 1.8

    1.6 Nanostructured Lipid Carriers (NLC)

    1.6.1 Introduction

    Nanostructured lipid carriers are developed to solve the problems related to SLN. Nanostructured lipid carriers are the combination of solid and liquid lipids. Because of combination of both liquid and solid lipids these have a less organized internal structure, show high drug-loading capacity and less drug expulsion property. Therefore, NLCs are considered superior than SLNs (Soni et al., 2015). Nanostructured lipid carriers show controlled release behavior and provide better chemical stability to the drug. These lipid carriers can easily go for scale up process (Selvamuthukumar and Velmurugan, 2012) (Fig. 1.6).

    Figure 1.6 Nanostructured lipid carrier.

    1.6.2 Classification

    Nanostructured lipid carriers can be visualized by using three models as discussed further (Muller et al., 2011).

    1.6.2.1 Imperfect type nanostructured lipid carriers

    In this model, different categories of lipids (different fatty acids) are mixed. This imperfect arrangement of lipids provides more space for the drug molecule to be incorporated.

    1.6.2.2 Multiple types nanostructured lipid carriers

    In this model, the highest drug loading is provided by mixing of solid and a little amount of liquid lipid. In this model, the drug is dissolved in the oil and protected by solid lipid from degradation. These types of NLCs are similar to multiple emulsions, such as w/o/w because of oil-in-solid lipid-in-water dispersion.

    1.6.2.3 Amorphous type nanostructured lipid carriers

    Drug expulsion occurring because of crystallization is prevented by amorphous type NLCs. In this model, some special types of lipids, such as hydroxyl octacosanyl, hydroxyl stearate, and isopropyl myristate particles are used to prevent crystallization upon cooling.

    1.6.3 Composition

    The Main components of NLCs are solid lipid, liquid lipid, emulsifier and water. These are summarized in Table 1.9 (Fang et al., 2013).

    Table 1.9

    Major components used for the preparation of NLCs are discussed in this chapter.

    Liquid lipids: Nanostructured lipid carriers are mainly prepared by medium chain triglycerides (MCT) and oleic acid. An MCT is prepared by a combination of various unique fatty acids, such as caprylic, capric, caproic, lauric, and myristic fatty acids. MCT are saturated lipids, solidify at 0°C, and also have low viscosity.

    Natural edible oils, such as corn oil, soybean oil, and sunflower oil can also be used as liquid lipid for NLC production. Some of these oils have natural antioxidants, leading to protection against oxidative degradation. Nanostructured lipid carriers are also prepared by squalene, a liquid lipid. It is a triterpene and also found in plants, animals, and humans.

    Solid lipids: Glyceryl behenate, glyceryl palmitostearate, glyceryl monostearate/monostearin, cetyl palmitate, and stearic acid are the most common solid lipids used for the preparation of nanostructured lipid carriers. Unsaturated lipids undergo oxidation faster than saturated lipids. Glyceryl behenate is made by diacylglycerols of behenic acid and also with different quantities of mono- and tri-acylglycerols. Glyceryl behenate has many imperfections in its crystalline lattice, which leads to high entrapment efficiency. Glyceryl palmitostearate is made up of a mixture of mono-, di-, and tri-acylglycerols of palmitic and stearic fatty acids. This lipid shows sustained-release action.

    Glyceryl monostearate is a mixture of mono and di acylglycerols. It is mainly used in cosmetics and pharmaceutical formulations. It works as stabilizer, nonionic emulsifier, emollient and as plastisizer in pharmaceutical formulations. Stearic acid is also widely as a solid lipid for the preparation of NLCs. This lipid is a primary component of lipid in animals and plant sources.

    Emulsifiers: All the NLCs are stabilized by various emulsifiers. Generally, a combination of surfactants are used for the preparation of stable formulation. Amphiphilic, lipophilic, and hydrophilic surfactants are equally important for the preparation of NLCs.

    1.6.4 Methods for Preparation Nanostructured Lipid Carriers

    1.6.4.1 Hot homogenization

    This technique is used at a temperature higher than melting point of lipids. Firstly, aqueous, i.e., double-distilled water, hydrophilic emulsifiers; then lipid phase, i.e., solid and liquid lipids, lipophilic emulsifiers are prepared separately. Both the phases are then maintained at the same temperature for a predetermined time. Aqueous phase is added to the lipid phase and homogenized by a high-shear homogenizer. This mixture is again treated with probe-type or water-bath type sonicator for achieving the small and uniform-sized NLCs (Hung et al., 2011).

    1.6.4.2 Cold homogenization

    This technique is mainly used for heat-sensitive drugs. In this technique, the solid lipid is ground to lipid microparticles and these microparticles are dispersed in a cold emulsifier solution to yield presuspension. Next, the prepared suspension is homogenized at room temperature. Cavitation force produced during homogenization breaks the microparticles directly to NLCs (Muèller et al., 2000).

    1.6.4.3 Microemulsion

    This technique is used for large scale production of lipid nanoparticles. In this technique, a transparent and thermodynamically stable emulsion, i.e., microemulsion, is formed by mixing of melted lipids, emulsifiers, and water at correct ratio. Nanostructured lipid carriers are formed by addition of this microemulsion to water, which leads to precipitation of the lipid

    Enjoying the preview?
    Page 1 of 1