You are on page 1of 22

q 2006 by Taylor & Francis Group, LLC

q 2006 by Taylor & Francis Group, LLC

q 2006 by Taylor & Francis Group, LLC

Dedication
To the loving memory of my dad,
Mustafa Amiji, and to my mom, Shirin Amiji

q 2006 by Taylor & Francis Group, LLC

Foreword
We are at the threshold of a new era in cancer treatment and diagnosis, brought about by the
convergence of two disciplinesmaterials engineering and life sciencesthat 30 years ago might
have been difficult to envision. The product of this curious marriage, nanobiotechnology, is yielding
many surprises and fostering many hopes in the drug-development space. Nanoparticles,
engineered to exquisite precision using polymers, metals, lipids, and carbon, have been combined
with molecular targeting, molecular imaging, and therapeutic techniques to create a powerful set of
tools in the fight against cancer. The unique properties of nanomaterials enable selective drug
delivery to tumors, novel treatment methods, intraoperative imaging guides to surgery, highly
sensitive imaging agents for early tumor detection, and real-time monitoring of response to
treatment.
Using nanotechnology, it may be possible to leap over many of the hurdles of cancer drug
delivery that have confounded conventional drugs. These hurdles include hindered access to the
central nervous system through the bloodbrain barrier, sequestration by the reticulo-endothelial
system, inability to penetrate the interior of solid tumors, and overcoming multi-drug resistance
mechanisms. These obstacles can be mitigated by manipulating the size, surface charge,
hydrophilicity, and attached targeting ligands of a therapeutic nanoparticle.
The novelty of using nanotechnology for medical applications presents its own challenges,
similar in many ways to the challenges faced by the introduction of the first synthetic protein-based
drugs. In the early 1980s, protein-based drugs offered an entirely new approach to targeting and
treating disease. They could be engineered to be highly specific and even offered the capability of
separating the targeting and therapeutic functions of a drug, such as when monoclonal antibodies
are conjugated to cytotoxic agents. But monoclonal antibodies and recombinant protein-based
drugs necessitated a different approach to lead optimization, metabolism, and toxicity screening
from that of small-molecule drugs. Factors such as stability, immunogenicity, and species
specificity had to be considered and tested in ways that were not familiar to the developers of
traditional drugs.
Nanotechnology-based drugs will catalyze a reevaluation of optimization, metabolism, and
toxicity-screening protocols. Interactions between novel materials and biological pathways are
largely unknown but are widely suspected to depend heavily on physical and chemical
characteristics such as particle size, particle size distribution, surface area, surface chemistry
(including charge and hydrophobicity), shape, and aggregation statefeatures not usually
scrutinized for traditional drugs. Standard criteria for physicochemical characterization will have to
be established before safety testing can yield interpretable and reproducible results.
Would nanotechnology-based drugs be successful? All drugs have to run an obstacle course
through physiological and biochemical barriers. For monoclonal antibody drugs, only a minute
portion of an intravenously administered drug reaches its target. However, drugs based on
nanomaterials, including nanoparticles, have unique properties that enable them to specifically bind
to and penetrate solid tumors. Size and surface chemistries can be manipulated to facilitate
extravasation through tumor vasculature, or the therapeutic agent can be encapsulated in polymer
micelles or liposomes to prevent degradation and increase circulation half-life to improve the odds
of it reaching the target.
This level of functional engineering has not been available to either small-molecule or proteinbased drugs. For these drugs, modification of one characteristic, such as solubility or charge, can
have dramatic effects on other essential characteristics, such as potency or target specificity.
Nanoparticles, however, introduce a much higher degree of modularity and offer at least four
advantages over the antibody conjugates: (1) the delivery of a larger therapeutic payload per target
recognition event, enhancing potency; (2) the ability to carry multiple targeting agents, enhancing
q 2006 by Taylor & Francis Group, LLC

selectivity; (3) the ability to carry multiple therapeutic agents, enabling targeted combination
therapies; and (4) the ability to bypass physiological and biological barriers.
We would all like to hasten the day when chemotherapythe administration of non-specific,
toxic anti-cancer agentsis relegated to medical history. Improvements in diagnostic screening
and the development of drugs that target specific biological pathways have begun to turn the tide
and contribute to a slow, yet consistent decline in death rates. While confirming that early diagnosis
and targeted therapies are pointing us in the right direction, the progress is still incremental.
Nanotechnology may be our best hope for overcoming many of the barriers faced by todays drugs
in the battle against cancer. The combined creative forces of engineering, chemistry, physics, and
biology will beget new and hopefully transformative options to old, intractable problems.
Piotr Grodzinski, Ph.D.
National Cancer Institute

q 2006 by Taylor & Francis Group, LLC

Preface
With parallel breakthroughs occurring in molecular biology and nanoscience/technology, the newly
recognized research thrust on nanomedicine is expected to have a revolutionary impact on the
future of healthcare. To advance nanotechnology research for cancer prevention, diagnosis, and
treatment, the United States National Cancer Institute (NCI) established the Alliance for
Nanotechnology in Cancer in September 2004 and has pledged $144.3 million in the next five years
(for details, visit http://nano.cancer.gov). Among the approaches for exploiting developments in
nanotechnology for cancer molecular medicine, nanoparticles offer some unique advantages as
sensing, delivery, and image enhancement agents. Several varieties of nanoparticles are available,
including polymeric conjugates and nanoparticles, micelles, dendrimers, liposomes, and
nanoassemblies.
This book focuses specifically on nanoscientific and nanotechnological strategies that are
effective and promising for imaging and treatment of cancer. Among the various approaches
considered, nanotechnology offers the best promise for targeted delivery of drugs and genes to the
tumor site and alleviation of the side effects of chemotherapeutic agents. Multifunctional
nanosystems offer tremendous opportunity for combining more than one drug or using drug and
imaging agents. The expertise of world-renowned academic and industrial researchers is brought
together here to provide a comprehensive treatise on this subject.
The book is composed of thirty-eight chapters divided into seven sections that address the
specific nanoplatforms used for imaging and delivery of therapeutic molecules. Section 1 focuses
on the rationale and fundamental understanding of targeting strategies, including pharmacokinetic
considerations for delivery to tumors in vivo, multifunctional nanotherapeutics, boron neutron
capture therapy, and the discussion on nanotechnology characterization for cancer therapy, as well
as guidance from the U.S. Food and Drug Administration on approval of nanotechnology products.
Section 2 focuses on polymeric conjugates used for tumor-targeted imaging and delivery, including
special consideration on the use of imaging to evaluate therapeutic efficacy. In Section 3, polymeric
nanoparticle systems are discussed with emphasis on biodegradable, long-circulating nanoparticles
for passive and active targeting. Section 4 focuses on polymeric micellar assemblies, where
sophisticated chemistry is applied for the development of novel nanosystems that can provide
efficient delivery to tumors. Many of the micellar delivery systems are undergoing clinical trials in
Japan and other countries across the globe. Dendritic nanostructures used for cancer imaging and
therapy are discussed in Section 5. Section 6 focuses on the oldest nanotechnology for cancer
therapyliposome-based delivery systemswith emphasis on surface modification to enhance
target efficiency and temperature-responsive liposomes. Lastly, Section 7 focuses on other lipid
nanosystems used for targeted delivery of cancer therapy, including nanoemulsions that can cross
biological barriers, solid-lipid nanoparticles, lipoprotein nanoparticles, and DQAsomes for
mitochondria-specific delivery.
Words cannot adequately express my admiration and gratitude to all of the contributing authors.
Each chapter is written by a world-renowned authority on the subject, and I am deeply grateful for
their willingness to participate in this project. I am also extremely grateful to Dr. Piotr Grodzinski
for providing the Foreword. Drs. Fredika Robertson and Mauro Ferrari have done a superb job in
laying the foundation by providing a chapter entitled Introduction and Rationale for
Nanotechnology in Cancer. I am grateful to Professor Kinam Park at Purdue University,
Professor Robert Langer at MIT, and Professor Vladimir Torchilin at Northeastern University, who
have been my mentors and collaborators, as well as many other researchers from academia and
industry. Special thanks are due to the postdoctoral associates and graduate students in my
laboratory at Northeastern University who have been the soldiers in the trenches in our quest to
use nanotechnology for the targeted delivery of drugs and genes to solid tumors. Lastly, I am deeply
q 2006 by Taylor & Francis Group, LLC

grateful to the wonderful people at Taylor & Francis-CRC Press, including Stephen Zollo, Patricia
Roberson, and many others, who have made the concept of this book into reality.
Any comments and constructive criticisms of the book can be sent to the editor at
m.amiji@neu.edu.

q 2006 by Taylor & Francis Group, LLC

Editor
Dr. Mansoor M. Amiji received his undergraduate degree in pharmacy from Northeastern
University in 1988 and his PhD in pharmaceutics from Purdue University in 1992. His areas of
specialization include polymeric biomaterials, advanced drug delivery systems, and nanomedical
technologies.
Dr. Amijis research interests include the synthesis of novel polymeric materials for medical and
pharmaceutical applications; surface modification of cationic polymers by the complexationinterpenetration method to develop biocompatible materials; the preparation and characterization
of polymeric membranes and microcapsules with controlled permeability properties for medical
and pharmaceutical applications; target-specific drug and vaccine delivery systems for
gastrointestinal tract infections; localized delivery of cytotoxic and anti-angiogenic drugs for
solid tumors in novel biodegradable polymeric nanoparticles; intracellular delivery systems for
drugs and genes using target-specific, long-circulating, biodegradable polymeric nanoparticles; and
gold and iron-gold core-shell nanoparticles for biosensing, imaging, and delivery applications. His
research has received sustained funding from the National Institutes of Health (NIH), the National
Science Foundation (NSF), foundations, and local industries.
Dr. Amiji is Professor and Associate Chair of the Pharmaceutical Sciences Department and CoDirector of the Northeastern University Nanomedicine Education and Research Consortium
(NERC). The NERC oversees a doctoral training grant in nanomedicine science and technology
that was co-funded by the NIH and NSF. He has two published books, Applied Physical Pharmacy
and Polymeric Gene Delivery: Principles and Applications, along with numerous manuscript
publications. He has also received a number of awards, including the 2003 Eurand Award for
Innovative Oral Drug Delivery Research, Third Prize.
Dr. Amiji has supervised the research efforts of over 50 postdoctoral associates, doctoral and
masters level graduate students, and undergraduate honors students over the last 13 years. His
teaching responsibilities include the Doctor of Pharmacy (PharmD) program and graduate
programs (MS and PhD) in pharmaceutical sciences, biotechnology, and nanomedicine.

q 2006 by Taylor & Francis Group, LLC

Contributors
Hamidreza Montazeri Aliabadi
Department of Pharmacy and Pharmaceutical
Sciences
University of Alberta
Edmonton, Alberta, Canada
Christine Allen
Department of Pharmaceutical Sciences
and
Department of Chemical Engineering and
Applied Chemistry
University of Toronto
Toronto, Ontario, Canada
Ashootosh V. Ambade
Department of Chemistry
University of Massachusetts at Amherst
Amherst, Massachusetts
Mansoor M. Amiji
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts
Joseph M. Backer
SibTech, Inc.
Newington, Connecticut
Marina V. Backer
SibTech, Inc.
Newington, Connecticut
You Han Bae
Department of Pharmaceutics and
Pharmaceutical Chemistry
University of Utah
Salt Lake City, Utah
Lajos P. Balogh
Department of Radiation Medicine
and
Department of Cellular Stress and
Cancer Biology
Roswell Park Cancer Institute
Buffalo, New York
q 2006 by Taylor & Francis Group, LLC

Rolf F. Barth
Department of Pathology
The Ohio State University
Columbus, Ohio
Reina Bendayan
Department of Pharmaceutical Sciences
University of Toronto
Toronto, Ontario, Canada
Tania Betancourt
The University of Texas at Austin
Austin, Texas
D. Bhadra
Department of Pharmaceutical Sciences
Dr. H.S. Gour University
Sagar, India
S. Bhadra
Department of Pharmaceutical Sciences
Dr. H.S. Gour University
Sagar, India
Sangeeta N. Bhatia
HarvardMIT Division of Health Sciences
and Technology
Massachusetts Institute of Technology
Cambridge, Massachusetts
Sarathi V. Boddapati
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts
Lisa Brannon-Peppas
The University of Texas at Austin
Austin, Texas
Mark Butler
Department of Chemical Engineering
and Applied Chemistry
University of Toronto
Toronto, Ontario, Canada
John C. Byrd
Division of HematologyOncology
NCI Comprehensive Cancer Center
The Ohio State University
Columbus, Ohio

Robert B. Campbell
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts
Shing-Ming Cheng
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts
Gerard G. M. DSouza
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts
Marina A. Dobrovolskaia
Nanotechnology Characterization Laboratory
NCI Frederick
Frederick, Maryland
Amber Doiron
The University of Texas at Austin
Austin, Texas
P. K. Dubey
Department of Pharmaceutical Sciences
Dr. H. S. Gold University
Sagar, India
Omid C. Farokhzad
Department of Anesthesiology, Perioperative
and Pain Medicine
Brigham and Womens Hospital and Harvard
Medical School
Boston, Massachusetts
Mauro Ferrari
The Brown Foundation Institute of
Molecular Medicine
M. D. Anderson Cancer Center
and
Alliance for Nanohealth
The University of Texas
Houston, Texas
Alberto A. Gabizon
Oncology Institute
Shaare Zedek Medical Center and
Hebrew University
Jerusalem, Israel
q 2006 by Taylor & Francis Group, LLC

Jinming Gao
Simmons Comprehensive Cancer Center
University of Texas Southwestern
Medical Center
Dallas, Texas
Hamidreza Ghandehari
Department of Pharmaceutical Sciences
University of Maryland
Baltimore, Maryland
Todd J. Harris
HarvardMIT Division of Health Sciences
and Technology
Massachusetts Institute of Technology
Cambridge, Massachusetts
Mitsuru Hashida
Department of Drug Delivery Research
Graduate School of Pharmaceutical Sciences
Kyoto University
Sakyo-ku, Kyoto, Japan
Yuriko Higuchi
Department of Drug Delivery Research
Graduate School of Pharmaceutical Sciences
Kyoto University
Sakyo-ku, Kyoto, Japan
Kang Moo Huh
Department of Polymer Science
and Engineering
Chungnam National University
Yuseong-Gu, Daejeon, South Korea
Edward F. Jackson
Division of Diagnostic Imaging
The University of Texas M. D. Anderson
Cancer Center
Houston, Texas
N. K. Jain
Department of Pharmaceutical Sciences
Dr. H.S. Gour University
Sagar, India
Eun-Kee Jeong
Department of Radiology
University of Utah
Salt Lake City, Utah

Ji Hoon Jeong
Department of Pharmaceutics and
Pharmaceutical Chemistry
University of Utah
Salt Lake City, Utah
Sangyong Jon
Department of Life Science
Gwangju Institute of Science and Technology
Gwangju, South Korea
Shigeru Kawakami
Department of Drug Delivery Research
Graduate School of Pharmaceutical Sciences
Kyoto University
Sakyo-ku, Kyoto, Japan
Mohamed K. Khan
Department of Radiation Medicine
and
Department of Cellular Stress and
Cancer Biology
Roswell Park Cancer Institute
Buffalo, New York
Chalermchai Khemtong
Simmons Comprehensive Cancer Center
University of Texas Southwestern
Medical Center
Dallas, Texas
Sun Hwa Kim
Department of Biological Sciences
Korea Advanced Institute of Science
and Technology
Daejeon, South Korea

Andras G. Lacko
Departments of Molecular Biology and
Immunology
University of North Texas Health Science
Center
Fort Worth, Texas
Robert Langer
Department of Chemical Engineering
Massachusetts Institute of Technology
Cambridge, Massachusetts
Afsaneh Lavasanifar
Department of Pharmacy and Pharmaceutical
Sciences
University of Alberta
Edmonton, Alberta, Canada
Eun Seong Lee
Amorepacific Corporation/R&D Center
Giheung-gu Yongin-si Gyeonggi-do
South Korea
Helen Lee
Department of Pharmaceutical Sciences
University of Toronto
Toronto, Ontario, Canada
Robert J. Lee
NCI Comprehensive Cancer Center
and
NSF Nanoscale Science and
Engineering Center
The Ohio State University
Columbus, Ohio

Tae-il Kim
School of Chemistry and Molecular
Engineering
Seoul National University
Seoul, South Korea

Sang Cheon Lee


Nanomaterials Application Division
Korea Institute of Ceramic Engineering and
Technology
Guemcheon-gu, Seoul, South Korea

Sushma Kommareddy
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts

Chun Li
M. D. Anderson
Cancer Center
The University of Texas
Houston, Texas

Vinod Labhasetwar
Departments of Pharmaceutical Sciences and
Biochemistry and Molecular Biology
University of Nebraska Medical Center
Omaha, Nebraska
q 2006 by Taylor & Francis Group, LLC

Yongqiang Li
Department of Pharmaceutical Sciences
University of Toronto
Toronto, Ontario, Canada

Bruce R. Line
Department of Radiology, and Greenebaum
Cancer Center
University of Maryland
Baltimore, Maryland
Jubo Liu
Department of Pharmaceutical Sciences
University of Toronto
Toronto, Ontario, Canada
Zheng-Rong Lu
Departments of Pharmaceutics and
Pharmaceutical Chemistry
University of Utah
Salt Lake City, Utah
S. Mahor
Department of Pharmaceutical Sciences
Dr. H. S. Gour University
Sagar, India
Walter J. McConathy
Department of Internal Medicine
University of North Texas Health Science
Center
Fort Worth, Texas
Scott E. McNeil
Nanotechnology Characterization Laboratory
NCI Frederick
Frederick, Maryland
T. J. Miller
Center for Drug Evaluation and Research
Food and Drug Administration
Silver Spring, Maryland
Amitava Mitra
Department of Pharmaceutical Sciences and
Center for Nanomedicine and
Cellular Delivery
University of Maryland
Baltimore, Maryland
S. M. Moghimi
Molecular Targeting and Polymer
Toxicology Group
School of Pharmacy
University of Brighton
Brighton, United Kingdom
q 2006 by Taylor & Francis Group, LLC

Randall J. Mrsny
Center for Drug Delivery/Biology
Welsh School of Pharmacy
Cardiff University
Cardiff, United Kingdom
R. S. R. Murthy
Pharmacy Department
The M.S. University of Baroda
Vadodara, India
Natarajan Muthusamy
NCI Comprehensive Cancer Center
The Ohio State University
Columbus, Ohio
Anjan Nan
Department of Pharmaceutical Sciences and
Center for Nanomedicine and
Cellular Delivery
University of Maryland
Baltimore, Maryland
Maya Nair
Departments of Molecular Biology and
Immunology
University of North Texas Health
Science Center
Fort Worth, Texas
Norased Nasongkla
Simmons Comprehensive Cancer Center
University of Texas Southwestern
Medical Center
Dallas, Texas
David Needham
Department of Mechanical Engineering
and Materials Science
Duke University
Durham, North Carolina
Tooru Ooya
Department of Materials Science
Japan Advanced Institute of Science
and Technology
Tatsunokuchi, Ishikawa, Japan

Jong-Sang Park
Department of Chemistry and Molecular
Engineering
Seoul National University
Seoul, South Korea

N. Sadrieh
Center for Drug Evaluation and Research
Food and Drug Administration
Silver Spring, Maryland

Kinam Park
Departments of Pharmaceutics and
Biomedical Engineering
Purdue University
West Lafayette, Indiana

Sanjeeb K. Sahoo
Department of Pharmaceutical Sciences
Nebraska Medical Center
Omaha, Nebraska
and
Institute of Life Sciences
Bhubaneswar, India

Tae Gwan Park


Department of Biological Sciences
Korea Advanced Institute of Science and
Technology
Daejeon, South Korea

Elamprakash N. Savariar
Department of Chemistry
University of Massachusetts at Amherst
Amherst, Massachusetts

Anil K. Patri
Nanotechnology Characterization Laboratory
NCI Frederick
Frederick, Maryland

Dinesh B. Shenoy
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts and Novavax, Inc.
Malvern, Pennsylvania

Ana Ponce
Department of Biomedical Engineering
Duke University
Durham, North Carolina

Patrick Lim Soo


Department of Pharmaceutical Sciences
University of Toronto
Toronto, Ontario, Canada

Natalya Rapoport
Department of Bioengineering
University of Utah
Salt Lake City, Utah

Stephan T. Stern
Nanotechnology Characterization
Laboratory
NCI Frederick
Frederick, Maryland

Mike Andrew Rauth


Department of Pharmaceutical Sciences
University of Toronto
Toronto, Ontario, Canada
L. Harivardan Reddy
Pharmacy Department
The M.S. University of Baroda
Vadodara, India
Fredika M. Robertson
College of Medicine and Comprehensive
Cancer Center
The Ohio State University
Columbus, Ohio
q 2006 by Taylor & Francis Group, LLC

S. (Thai) Thayumanavan
Department of Chemistry
University of Massachusetts at Amherst
Amherst, Massachusetts
Sandip B. Tiwari
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts
Werner Tjarks
College of Pharmacy
The Ohio State University
Columbus, Ohio

Vladimir P. Torchilin
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts
Anagha Vaidya
Departments of Pharmaceutics and
Pharmaceutical Chemistry
University of Utah
Salt Lake City, Utah
Geoffrey von Maltzahn
HarvardMIT Division of Health Sciences
and Technology
Massachusetts Institute of Technology
Cambridge, Massachusetts
S. P. Vyas
Department of Pharmaceutical Sciences
Dr. H. S. Gour University
Sagar, India
Sidney Wallace
Division of Diagnostic Imaging
The University of Texas M. D. Anderson
Cancer Center
Houston, Texas
Yanli Wang
Departments of Pharmaceutics and
Pharmaceutical Chemistry
University of Utah
Salt Lake City, Utah
Volkmar Weissig
Department of Pharmaceutical Sciences
Northeastern University
Boston, Massachusetts

q 2006 by Taylor & Francis Group, LLC

Ho Lun Wong
Department of Pharmaceutical Sciences
University of Toronto
Toronto, Ontario, Canada
Gong Wu
Department of Pathology
The Ohio State University
Columbus, Ohio
Xiao Yu Wu
Department of Pharmaceutical Sciences
University of Toronto
Toronto, Ontario, Canada
Xiao-Bing Xiong
Faculty of Pharmacy and Pharmaceutical
Sciences
University of Alberta
Edmonton, Alberta, Canada
Weilian Yang
Department of Pathology
The Ohio State University
Columbus, Ohio
Furong Ye
Departments of Pharmaceutics and
Pharmaceutical Chemistry
University of Utah
Salt Lake City, Utah
Guodong Zhang
M. D. Anderson
Cancer Center
The University of Texas
Houston, Texas
Xiaobin B. Zhao
NCI Comprehensive Cancer Center
The Ohio State University
Columbus, Ohio
.

Table of Contents
Section 1
Nanotechnology and Cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Chapter 1

Introduction and Rationale for Nanotechnology


in Cancer Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3

Fredika M. Robertson and Mauro Ferrari


Chapter 2

Passive Targeting of Solid Tumors: Pathophysiological


Principles and Physicochemical Aspects of
Delivery Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11

S. M. Moghimi
Chapter 3

Active Targeting Strategies in Cancer with a Focus on


Potential Nanotechnology Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

Randall J. Mrsny
Chapter 4

Pharmacokinetics of Nanocarrier-Mediated
Drug and Gene Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43

Yuriko Higuchi, Shigeru Kawakami, and Mitsuru Hashida


Chapter 5

Multifunctional Nanoparticles for Cancer Therapy. . . . . . . . . . . . . . . . . . . . . . . 59

Todd J. Harris, Geoffrey von Maltzahn, and Sangeeta N. Bhatia


Chapter 6

Neutron Capture Therapy of Cancer: Nanoparticles


and High Molecular Weight Boron Delivery Agents. . . . . . . . . . . . . . . . . . . . . . 77

Gong Wu, Rolf F. Barth, Weilian Yang, Robert J. Lee, Werner Tjarks, Marina V. Backer, and
Joseph M. Backer
Chapter 7

Preclinical Characterization of Engineered Nanoparticles


Intended for Cancer Therapeutics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105

Anil K. Patri, Marina A. Dobrovolskaia, Stephan T. Stern, and Scott E. McNeil


Chapter 8

Nanotechnology: Regulatory Perspective for Drug


Development in Cancer Therapeutics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139

N. Sadrieh and T. J. Miller

Section 2
Polymer Conjugates. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 157
Chapter 9

Polymeric Conjugates for Angiogenesis Targeted Tumor


Imaging and Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159

Amitava Mitra, Anjan Nan, Bruce R. Line, and Hamidreza Ghandehari


q 2006 by Taylor & Francis Group, LLC

Chapter 10

Poly(L-Glutamic Acid): Efficient Carrier of Cancer Therapeutics and


Diagnostics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185

Guodong Zhang, Edward F. Jackson, Sidney Wallace, and Chun Li


Chapter 11

Noninvasive Visualization of In Vivo Drug Delivery


of Paramagnetic Polymer Conjugates with MRI. . . . . . . . . . . . . . . . . . . . . . . 201

Zheng-Rong Lu, Yanli Wang, Furong Ye, Anagha Vaidya, and Eun-Kee Jeong
Section 3
Polymeric Nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213
Chapter 12

Polymeric Nanoparticles for Tumor-Targeted Drug


Delivery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215

Tania Betancourt, Amber Doiron, and Lisa Brannon-Peppas


Chapter 13

Long-Circulating Polymeric Nanoparticles for Drug and


Gene Delivery to Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231

Sushma Kommareddy, Dinesh B. Shenoy, and Mansoor M. Amiji


Chapter 14

Biodegradable PLGA/PLA Nanoparticles for Anti-cancer


Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 243

Sanjeeb K. Sahoo and Vinod Labhasetwar


Chapter 15

Poly(Alkyl Cyanoacrylate) Nanoparticles for Delivery


of Anti-Cancer Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251

R. S. R. Murthy and L. Harivardhan Reddy


Chapter 16

Aptamers and Cancer Nanotechnology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

Omid C. Farokhzad, Sangyong Jon, and Robert Langer


Section 4
Polymeric Micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315
Chapter 17

Polymeric Micelles for Formulation of Anti-Cancer Drugs. . . . . . . . . . . . . . . 317

Helen Lee, Patrick Lim Soo, Jubo Liu, Mark Butler, and Christine Allen
Chapter 18

PEO-Modified Poly(L-Amino Acid) Micelles for


Drug Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 357

Xiao-Bing Xiong, Hamidreza Montazeri Aliabadi, and Afsaneh Lavasanifar


Chapter 19

Hydrotropic Polymer Micelles for Cancer Therapeutics . . . . . . . . . . . . . . . . . 385

Sang Cheon Lee, Kang Moo Huh, Tooru Ooya, and Kinam Park
Chapter 20

Tumor-Targeted Delivery of Sparingly-Soluble Anti-Cancer


Drugs with Polymeric Lipid-Core Immunomicelles . . . . . . . . . . . . . . . . . . . . 409

Vladimir P. Torchilin
q 2006 by Taylor & Francis Group, LLC

Chapter 21

Combined Cancer Therapy by Micellar-Encapsulated


Drugs and Ultrasound. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421

Natalya Rapoport
Chapter 22

Polymeric Micelles Targeting Tumor pH. . . . . . . . . . . . . . . . . . . . . . . . . . . . 443

Eun Seong Lee and You Han Bae


Chapter 23

cRGD-Encoded, MRI-Visible Polymeric Micelles


for Tumor-Targeted Drug Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465

Jinming Gao, Norased Nasongkla, and Chalermchai Khemtong


Chapter 24

Targeted Antisense Oligonucleotide Micellar


Delivery Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 477

Ji Hoon Jeong, Sun Hwa Kim, and Tae Gwan Park


Section 5
Dendritic Nanocarriers. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 487
Chapter 25

Dendrimers as Drug and Gene Delivery Systems . . . . . . . . . . . . . . . . . . . . . . 489

Tae-il Kim and Jong-Sang Park


Chapter 26

Dendritic Nanostructures for Cancer Therapy . . . . . . . . . . . . . . . . . . . . . . . . 509

Ashootosh V. Ambade, Elamprakash N. Savariar, and S. (Thai) Thayumanavan


Chapter 27

PEGylated Dendritic Nanoparticulate Carriers of


Anti-Cancer Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 523

D. Bhadra, S. Bhadra, and N. K. Jain


Chapter 28

Dendrimer Nanocomposites for Cancer Therapy . . . . . . . . . . . . . . . . . . . . . . 551

Lajos P. Balogh and Mohamed K. Khan


Section 6
Liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 593
Chapter 29

Applications of Liposomal Drug Delivery Systems


to Cancer Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 595

Alberto A. Gabizon
Chapter 30

Positively-Charged Liposomes for Targeting


Tumor Vasculature. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 613

Robert B. Campbell
Chapter 31

Cell Penetrating Peptide (CPP)Modified Liposomal


Nanocarriers for Intracellular Drug and Gene Delivery . . . . . . . . . . . . . . . . . 629

Vladimir P. Torchilin
q 2006 by Taylor & Francis Group, LLC

Chapter 32

RGD-Modified Liposomes for Tumor Targeting . . . . . . . . . . . . . . . . . . . . . . 643

P. K. Dubey, S. Mahor, and S. P. Vyas


Chapter 33

Folate Receptor-Targeted Liposomes for


Cancer Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 663

Xiaobin B. Zhao, Natarajan Muthusamy, John C. Byrd, and Robert J. Lee


Chapter 34

Nanoscale Drug Delivery Vehicles for Solid Tumors:


A New Paradigm for Localized Drug Delivery
Using Temperature Sensitive Liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . 677

David Needham and Ana Ponce

Section 7
Other Lipid Nanostructures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 721
Chapter 35

Nanoemulsion Formulations for Tumor-Targeted Delivery . . . . . . . . . . . . . . . 723

Sandip B. Tiwari and Mansoor M. Amiji


Chapter 36

Solid Lipid Nanoparticles for Antitumor Drug Delivery . . . . . . . . . . . . . . . . . 741

Ho Lun Wong, Yongqiang Li, Reina Bendayan, Mike Andrew Rauth, and Xiao Yu Wu
Chapter 37

Lipoprotein Nanoparticles as Delivery Vehicles


for Anti-Cancer Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 777

Andras G. Lacko, Maya Nair, and Walter J. McConathy


Chapter 38

DQAsomes as Mitochondria-Targeted Nanocarriers


for Anti-Cancer Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 787

Shing-Ming Cheng, Sarathi V. Boddapati, Gerard G. M. DSouza, and Volkmar Weissig

q 2006 by Taylor & Francis Group, LLC

You might also like