Sie sind auf Seite 1von 9

Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101 109 www.nanomedjournal.

com

Review: Cancer

Nanotechnology, nanomedicine, and the development of new, effective therapies for cancer
Ernest S. Kawasaki, PhD,T Audrey Player, PhD
National Cancer Institute, Gaithersburg, Maryland Received 1 March 2005; accepted 25 March 2005

Abstract

Cancer is the leading cause of death in the United States among people younger than 85 years, and for the first time has surpassed heart disease as the number one killer. This worrisome statistic has resulted not from an increase in the incidence of cancer, but because deaths from heart disease have dropped nearly in half while the number of cancer-related deaths has remained about the same. This fact accentuates the need for a new generation of more effective therapies for cancer. In this review, the development of new therapies will be discussed in the context of advances in nanotechnologies related to cancer detection, analysis, diagnosis, and therapeutic intervention. First, several nanoanalytical methods, such as the use of quantum dots in detection and imaging of cancer, will be described. These techniques will be essential to the process of precisely describing cancer at the level of the cell and whole organism. Second, examples of how nanotechnologies can be used in the development of new therapies will be given, including methods that might allow for more efficient and accurate drug delivery and rationally designed, targeted drugs. Finally, a new initiative the National Cancer Institute Alliance for Nanotechnology in Cancer will be described and discussed with respect to the scientific issues, policies, and funding. D 2005 Elsevier Inc. All rights reserved.
Nanotechnology; Nanomedicine; Nanoparticles; Quantum dots; Drug delivery; Cancer therapy

Key words:

The bwar on cancer Q is now in its fourth decade since the National Cancer Act was passed in 1971. Although much progress has been made in cataloging the environmental causes and cellular and molecular biological basis for this dreaded disease, we still do not have a precise understanding of the differences between a cancer cell and its normal counterpart. If we do not understand cancer, we cannot control, conquer, and eliminate it. The completion of the human genome sequence in 2001 [1,2] and subsequent improvements in the sequence data [3] are important steps toward our goal to fully comprehend cancer cell biology. We are now closer to being able to fully characterize the differences between the normal and the tumor cell. Coupled with the use of microdissection techniques [4], it is also

No financial conflict of interest was reported by the authors of this paper. T Reprint requests. NCI Microarray Facility, National Cancer Institute, Advanced Technology Center, 8717 Grovemont Circle, Bethesda, MD 20892-4605. E-mail address: ke@mail.nih.gov (E.S. Kawasaki). 1549-9634/$ see front matter D 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.nano.2005.03.002

possible to interrogate the genetic make-up of individual cell types. The hope is that use of such technologies will accelerate the progress in identifying the differences between normal and tumor cells, which in turn will lead to development of new therapies that will specifically target the cancer. The ultimate goal of these strategies is to eliminate the tumor with limited effect on normal tissue. At about the same time that the human genome was being sequenced, a new, novel focus of research evolved from the convergence and coalescence of many diverse scientific disciplines. This new area of research, for the sake of simplicity in this article, will be called bnanotechnology Q as a general term for the creation, manipulation, and application of structures in the nanometer size range. The term bnanoscienceQ is used to infer the study of the phenomena associated with objects somewhat arbitrarily defined as having dimensions between 1 to 100 nm. Indeed, the prefix bnanoQ is now being used in so many fields of research that it has become a source of confusion [5]. In this article, nanomedicine can be thought of as a subdiscipline contained within nanotechnology or the nanosciences. Here

102

E.S. Kawasaki, A. Player / Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101109

the nanomedicine aspects of nanotechnology will be stressed and will cover areas such as bioimaging, drug delivery systems, and new drug therapies as they relate to cancer. This will not be an exhaustive review, as only a few examples will be given, so the authors wish to apologize for the many excellent articles in the field of nanomedicine that will not be cited.

Discussion Nanotechnologies in bioimaging and detection Molecular imaging of live cells and whole organisms is an important tool for studying cancer biology and determining the efficacy of tumor therapies. This type of visualization has been helped tremendously by the development of fluorescent probes called quantum dots [6-8]. Semiconductor quantum dots (QDs) are nanocrystals or nanoparticles in the range of 1 to 10 nm with unique photochemical and photophysical properties not available with common organic dyes and fluorescent proteins [9]. Depending on their size, the QDs, excited at appropriate wavelengths, will emit light in different colors. In general, a smaller particle will emit a shorter wavelength of light than the next larger size. Their stability, increased fluorescence capability, and multicolor fluorescence emission has made QDs excellent reagents for in vivo imaging at the molecular and cellular levels as well as at the whole organ and animal level. There are other types of nanoparticles that can be used for this purpose [10-14], but at the moment QDs appear to be the most versatile for in vivo cancer studies. Modified QDs can be used as robust immunofluorescent probes for detection of cancer markers such as Her2 and other cellular targets on putative tumor cells [15]. Derivatized QDs can be linked to immunoglobulins for development of fluorescent imaging assays for the detection of specific antigens on fixed or live cells. There are several advantages to using QDs: the intensity of signal is much brighter, the signal to noise ratio is higher, and photostability is much better than for common fluorescent molecules [15,16]. In addition, QDs can potentially be used for long-term labeling of live cells [17-19]. The QDs can be directed to cell surface markers via conjugated antibodies and/or they can be internalized into the cells by endocytic uptake [17]. If internalized by endocytosis, the particles can be directed out of the endosome to the cytosol or nucleus by using amphipathic peptides that facilitate uptake. Because the particles are stable, the cells may be followed for weeks and the fluorescence data recorded at multiple time points. Another interesting and useful application of QDs is their use in boptical codingQ of live cells for use in drug compound screens, for multiplexed assays, or for following individual cells in mixed cell populations [20,21]. For analysis of mixed cell populations, cells are transfected with different-sized QDs or different proportions of the particles. The cell types can then be distinguished by the wave-

length(s) of their emitted light. It is possible to create greater than 100 different codes, a process that will be useful in development of a wide variety of multiplex assays. In conjunction with single-cell analysis, the ability to visualize whole organs or tumor masses in live organisms in situ is crucial for studies in cancer biology and therapeutics [21-24]. In an early study [25], QDs were coated with a lung-targeting peptide to demonstrate localization and accumulation of the probes in the lungs after intravenous injection, while 2 different peptides were used to direct probes to blood or lymphatic vessels associated with tumors. This study demonstrated the feasibility of using QDs for the in vivo targeting of specific organs or tissues, which could potentially be useful for detection of specific diseases and drug delivery. In a study of metastasis [26], B16F10 tumor cells labeled with QDs were used to track cellular extravasation into lung tissue in mice injected with these cells. The investigators demonstrated the utility of QDs in characterizing the metastatic behavior and potential of these cell types in vivo. Their study showed that the biological behavior of the labeled cells was indistinguishable from unlabeled cells, and that by labeling each cell with a different QD, up to 5 different populations of cells could be followed. An important aspect of diagnosis and prognosis of many cancers is the determination of the extent of involvement of the lymph nodes in the disease. The sentinel lymph node (the first node in the lymphatic basin into which the primary tumor drains) [27] is often a target of surgical resection by the oncologist but may not always be easy to detect or visualize. A novel nanoparticle, called the near-infrared fluorescent type II quantum dot, has been used to label the sentinel nodes for mapping and imaging [28]. Near-infrared QDs are similar to those described above except that their emission spectrum is just outside of human color detection in the infrared. The advantage of these particles is that infrared can penetrate much deeper into living tissue, and thus the near-infrared QDs are able to detect and image objects that are not detectable by QDs that emit in the visible range. This type of QD technology provides the surgeon with an image of the lymph node, allows real-time visualization of organ during surgery, and can be used for confirmation that the operation has been complete by observing the loss of concentrated QD fluorescence in the lymph node area. One of the most promising applications of QDs is for the detection and diagnosis of cancers in vivo. Preliminary experiments in mice have shown that multifunctional nanoparticle probes can home in on cancer targets in these animals and allow for visualization of the tumors in vivo [29]. The particles were protected from degradation in vivo by coating with a copolymer, contained multiple polyethylene glycol (PEG) molecules for improved biocompatibility, and contained monoclonal antibodies for recognition of prostate tumor antigens. By using specific QDs, the authors were able to easily image prostate cancer tumor xenografts

E.S. Kawasaki, A. Player / Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101109

103

in the whole animal. The QDs used in these studies were used for detection of subdermal xenografts but would not be able to image tumors internally. However, with the nearinfrared type particle described above [28], imaging reagents called near-infrared-emissive polymersomes [30], or perhaps with magnetic nanoparticles [11,13,31], deeper tumors can be visualized, eliminating the need for more invasive procedures. The multifunctional QDs described in this study may be used for drug delivery as well, providing a versatile platform for diagnosis and therapy. Prevention is the best bcureQ for cancer, but failing that, early detection will greatly increase survival rates with the reasonable assumption that an in situ tumor will be easier to eradicate than one that has metastasized [32]. RNA or DNA markers from exfoliated tumor cells in circulation can be analyzed by polymerase chain reaction (PCR) or other amplification technologies, but tumor protein markers may be more informative [33]. The major difficulty with protein analysis is that there are few tumor-specific molecules in the plasma at early stages of the cancer, and direct amplification methods are not available for proteins. Detection of these proteins by established protocols is not feasible, but a new method called bnanoparticle-based bio-bar codesQ allows for the ultrasensitive detection of any protein where appropriate antibodies are available [34 -36]. The assay uses 2 types of probes: a magnetic 1-Am diameter microparticle functionalized with a tumor antigen monoclonal antibody, and 10- to 30-nm gold nanoparticles containing oligonucleotide bar codes and polyclonal antibodies to the antigen in question. The 2 particles essentially are used to perform a sandwich assay in solution, and after processing, the bar code oligonucleotides can be amplified by PCR to increase the sensitivity of the assay by a million-fold. By this method, as few as 30 molecules of protein can be detected, which hopefully will be sufficiently sensitive to diagnose early stages of cancer. The low-molecular-weight (LMW) region of the blood proteome appears to contain important biomarkers for the detection, diagnosis, and prognosis of cancers [33,37]. These LMW proteins are usually in very low abundance and difficult to detect even by mass spectrometry. However, new micro- or nano-fabrication methods may allow the production of nanoparticles that can be used as bnanoharvestersQ to collect and concentrate LMW proteins so that they may be easily analyzed by sensitive mass spectrometric techniques. This is another example of how nanotechnologies will enable the researcher and clinician to analyze cancer markers that were previously undetectable. Nanotechnologies in drug delivery Perhaps the greatest immediate impact of nanotechnologies in cancer therapy will be in the realm of drug delivery. The therapeutic index of nearly all drugs currently being used would be improved if they were more efficiently delivered to their biological targets through appropriate application of nanotechnologies [38,39]. Some drugs

that have previously failed clinical trials might also be re-examined using nanotechnological approaches. A number of obstacles may be overcome with various novel applications of nanodrug delivery. For example, many drugs are not very soluble, making it difficult to administer therapeutic doses [40- 42]. These compounds can be bsolubilizedQ by formulating them into crystalline nanosuspensions that are stabilized by surfactants [42], or by combining them with organic or lipid nanoparticles that keep them in circulation for longer periods [43- 45]. If an efficacious compound has a short half-life in the circulation, its stability can be increased tremendously by encasing it within nanosized liposomes as a drug carrier [44]. In the case of central nervous system cancers, many drugs have difficulty in crossing the bloodbrain barrier to attack the tumor. Drug-loaded nanoparticles are able to penetrate this barrier, and have been shown to greatly increase therapeutic concentrations of anticancer drugs in brain tumors [46,47]. The best way to increase the efficacy and reduce the toxicity of a cancer drug is to direct the drug to its target and maintain its concentration at the site for a sufficient time for therapeutic action to take effect [39,48]. For example, lipid cationic nanoparticles coupled to an integrin-targeting ligand were shown to deliver genes selectively to angiogenic blood vessels in tumor-bearing mice. As the therapeutic part of the nanocomplex, a mutant Raf gene was coupled to the particle for transfection and expression in the tumor cells; expression of this mutant gene was shown to block angiogenesis in this model. The directed nanoparticle caused apoptosis in the tumors and a sustained regression of established primary and metastatic tumors [49]. Liposomes (phospholipid spheres of ~100 nm in diameter that are bi-layered in structure) are excellent carriers for a variety of drugs [44] and are being tested extensively for use in gene therapy protocols and targeted drug delivery in cancer. With current liposome technologies it is not difficult to transfect a cell for purposes of gene therapy, but the therapeutic gene may be degraded if it is not able to traffic out of the endosome. To enhance the efficacy of gene therapy, synthetic pH-sensitive histidylated oligolysine can be added to a drugliposome complex to aid in escaping from the endosome [50]. This protocol was shown to improve the transfection efficiency in prostate and pancreatic cancer cell lines by 39-fold, and elevated the expression of the transgene in a human prostate cancer xenograft model in athymic nude mice without increasing toxicity. Monoclonal antibodies are good targeting vehicles for nanoparticles but other bioconjugates are being tested with varying degrees of success. Nucleic acid ligands called aptamers that mimic antibodies are potential replacements because they can be designed to bind to practically any antigen in an in vitro system. The aptamers are generated by evolutionary methods in vitro, and the molecules with high affinity are used for targeting antigens in vivo. This strategy

104

E.S. Kawasaki, A. Player / Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101109

has been applied to directing PEG-coated nanoparticles to home in on prostate-specific membrane antigen in prostate cancer cells. The aptamer conjugated particles were shown to have a 77-fold increase in binding versus the control particles, and a large increase in uptake of drug-encapsulated particles [51]. There are numerous examples of similar type targeting of nanoparticles [39,48,52-54], and this area of research promises to provide important weapons in the arsenal for developing a cure for cancer. Nanomedicine and new drug therapies All drug molecules are, in essence, products of natural or synthetic nanoengineering. For example, the most widely used and effective drug, the common aspirin, is only about 0.6 nm [55]. On the blargeQ side are therapeutic monoclonal antibodies that are approximately 30 nm in length. Many of the globular proteins, such as hemoglobin, are 5 nm in diameter, and natural proteins of similar size may be used as a therapeutic. Double-stranded DNA in the nucleus is about 2.5 nm in width, but totals 2 meters in length in the mammalian cell, an astounding example of bnanopackingQ in a 2- to 5-Amdiameter nucleus. Medicinal and structural chemists have been creating and manipulating nanometer- and subnanometer-sized components of drugs for decades, and will continue to do so into the foreseeable future. The difference is that they will now be joined by a wide variety of scientists from a number of disciplines normally not involved in drug research. A recent article written by physicists in Physical Review Letters [56], titled bElectronic Structure and Bonding of Au on a SiO2 Cluster: A Nanobullet for Tumors,Q is a good example of how research from diverse sources can and should be brought to bear on the problem of cancer. Success in this endeavor, however, will require a concerted effort to integrate and coordinate the research in an approach that might now be described as bsystems biologyQ [57,58]. What are the requirements for an effective and safe cancer drug [59]? There must be an adequate drug concentration in the body to allow for an effective dose at the tumor site. The target must be strongly inhibited, with the targetTs function essential for tumor cell viability. The drug must have a high differential toxicity toward the tumor or a favorable therapeutic window. Research in nanomedicine will be addressing all these points, and a few examples in the drug development arena will be given below. Monoclonal antibodies will be an essential component of the new wave of cancer treatments developed through nanotechnologies. They are being used as imaging vehicles, for drug targeting, as drug carriers, and as the drug itself. There are 9 or more FDA-approved antibodies approved for clinical use in cancer [60-62], and approximately 20 more are being evaluated in clinical trials. The mechanism of action of the antibodies includes receptor ligand binding competition; interference with receptor function; antibody-dependent, cell-mediated cytoxicity; complementdependent cellular cytoxicity; or, perhaps, a combination of

the above. This activity can also be combined with toxins directed at the cancer cell to produce an even more efficacious drug. One of the holy grails of drug research is to be able to rationally design and produce effective small-molecule inhibitors of protein function [63-69]. Development of many drugs will be the result of application of nanotechnologies that have been in place for many years. For example, nuclear magnetic resonance and x-ray crystal structures of target proteins and their ligands or substrates are being used as the template for rational design of new drugs. The target may be enzymes or receptorligand proteins. Inhibitors of enzymatic activity are, in general, easier to design than blockers of proteinprotein interactions. As an example, the first successful drug approved for the treatment of chronic myeloid leukemia (CML), Gleevec, is an inhibitor of the tyrosine kinase mutant, BCR-ABL [70,71]. For many patients, administration of the drug results in what appears to be complete remission, but Gleevec-resistant leukemia often returns through mutations in the active site. By careful study of the mutations and the structure of the kinase, new small molecular inhibitors were designed that could block the mutant strains and appear to be more efficacious than the original drug [72-74]. The 2 drugs are reported to work synergistically, which will, hopefully, result in a complete cure of CML. Drugs derived from nucleic acids are beginning to make an impact on the nanomedicine scene. Antisense technology [75] exploits the use of oligonucleotides in the range of 15 to 20 nucleotides to block the function of an RNA target. This technology has made rapid progress after experiencing initial difficulties in showing efficacy for in vivo models of disease. Ongoing clinical trials using antisense drugs include prostate, breast, pancreatic, lung, colorectal, melanoma, and brain cancers. A novel approach for this technology is to use oligonucleotides for sensitizing tumor cells to chemotherapy. The oligonucleotides are being combined with nanoliposomes to target and deliver the nucleic acids to the cancer cells [76] and block production of the alpha folate receptor. This block was shown to decrease cell survival of breast cancer cell lines, and sensitized a cell line by 5-fold to doxorubicin. This is a good example of how nanotechnologies can be used to increase the effectiveness of existing drugs, facilitating the use of lower dosages to decrease toxicity. The latest additions to the repertoire of gene-specific silencing agents are the bsmall interferingQ RNAs or siRNAs [77-79], which are the effector molecules of the RNA interference (RNAi) pathways found in most eukaryotes. The siRNAs are small double-stranded molecules of about 21 nucleotides in length that result in specific degradation of mRNAs containing the complementary sequence. This specificity makes siRNAs attractive candidates as nanodrugs for blocking the expression of wayward genes in cancer, but their application can be hindered by instability in the blood and poor uptake into the tar-

E.S. Kawasaki, A. Player / Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101109

105

get cells. To overcome these problems, siRNA can be complexed with nanoparticles containing a homing sequence that directs the complex to the tumor site [54]. By this method, siRNA directed against vascular, endothelial, growth factor receptor-2, and VEGF R2 was able to block expression of the growth factor in tumor-bearing mice, and inhibit tumor angiogenesis and growth rate. RNAi drugs combined with nanodelivery agents will be an important addition to cancer therapies and other areas of nanomedicine. A fascinating approach for eradicating tumors is through the application of cancer immunotherapy or vaccines [80-82]. It is thought that the body can eliminate small tumors by an appropriate immune response, but at some point a malignant cancer is able to evade the response by developing mechanisms that blind the host to its presence. If methods can be found to activate the immune system against these tumors, the body should be able to destroy the cancer, and all forms of cancer might be susceptible to this type of immunotherapy. Tumor antigens by themselves are not very immunogenic and require some type of adjuvant to boost the immune response against the cancer. A new vaccine design has been developed that couples the antigens to solid-core nanobeads [83]. For effectiveness, the beads have to be of narrowly defined size (40 nm to 50 nm), which allows them to localize to dendritic cells in the draining lymph nodes. Conjugation of the antigens to the nanobeads induced responses that were 2- to 10-fold higher than other bead sizes, and higher than currently used immunizing adjuvants. A single dose of the antigen-coated beads protected mice from tumors in 2 different model challenges, and was even able to cause rapid clearance of established tumors. This is a good example of how nanotechnologies may provide a major breakthrough in cancer therapy and the effectiveness of vaccines in general. Cancer immunotherapy may provide a relatively benign, nonchemotherapeutic method of destroying tumors. Another promising and, perhaps, complementary approach is the thermal ablation method of tumor destruction. Treatment of solid tumors with hyperthermia has been an option for some time [84], but has some drawbacks. For deep, underlying tumors, the energy source can harm the intervening and surrounding healthy tissue even when focused beams are used. To overcome this problem methods have been developed to selectively heat the tumors using nearinfraredabsorbing gold nanoparticles called nanoshells [85-87]. Nanoshells, in this case, are composed of a silica core surrounded by a thin gold metal shell, and will absorb energy (heat up) when exposed to the appropriate wavelength of light. The near-infrared characteristics were chosen because absorption by tissues is minimal and penetration of the light is optimal at this wavelength. The nanoshells were injected into mice, and the nanoparticles were simply allowed to accumulate in implanted tumors. This can occur as most tumor vasculature is bleaky,Q and will allow nanosized particles to penetrate into the tumor while normal tissue or organs are not affected. The tumors

are then illuminated with a near-infrared diode laser to heat the tumor and cause cellular destruction. By this protocol all the tumors were ablated, and the mice remained tumor-free for many months. Although not used in these studies, the efficacy of the procedure could be improved by attachment of tumor-homing or -targeting molecules to the nanoshells for increasing concentration at the site of heating. In appropriate settings, thermal ablation methods could be used to replace surgical resection of tumors, and targeted therapies and immunotherapy as a substitute for toxic chemotherapy. It is becoming increasingly clear that a bone-two punchQ is needed to eradicate tumors, and the nanomedicinal methods being developed have a good chance of achieving this goal with much less damage to normal tissue than existing therapeutic protocols. National Cancer Institute Nanotechnology Initiative National Cancer Institute (NCI) director, Dr. Andrew C. von Eschenbach, has set the goal of ending suffering from cancer by the year 2015. To meet this ambitious goal, clinicians will need new insights, tools, and approaches for the prevention, detection, and treatment of primary cancer and metastases. A major component of this complex endeavor will be the newly launched $144.3-million, 5-year initiative named the bNCI Alliance for Nanotechnology in CancerQ [88]. The initiative is meant to bring together the physical, biological, and medical science communities in a coordinated effort to direct the benefits of nanotechnology to cancer patients. A more detailed discussion of the nanotechnology initiative can be found at the NCI website (http://nano.cancer.gov). One of the AllianceTs first steps was to establish the Nanotechnology Characterization Laboratory (NCL), which will work in concert with the National Institute of Standards and Technology (NIST) and the Food and Drug Administration (FDA). With NCL in the lead, the organizations will develop standards for the characterization of nanoscale materials and devices, and for their preclinical testing as well. The close links among the NCL, NIST, and FDA will provide a boost for the development of cancer nanotechnologies and facilitate the accelerated regulatory review and translation of therapies into the clinical realm. The major goals of the NCL will include (1) standardizing preclinical testing and characterizing nanoscale materials or devices to speed the regulatory review of novel cancer diagnostics, therapeutics, and prevention strategies; (2) performing preclinical toxicology, pharmacology, and efficacy testing for intramural and extramural NCI investigators as well as the private sector; (3) facilitating collaborations among the NCI, academia, and the private sector to facilitate the translation of basic nanoresearch into the clinical arena; (4) providing a focus for multidisciplinary research, development, and clinical applications of nanotechnology; and (5) collaborating with other government agencies to capitalize on existing resources and expertise to accelerate the development of nanotechnological

106

E.S. Kawasaki, A. Player / Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101109

applications and establish a partnership with industry to bring them to market. The emphasis of the Alliance will be directed toward the following areas, not strictly defined, but meant mainly as guidelines for research. A summary of the Cancer Nanotechnology Plan or CNPlan is given below. ! Prevention and control of cancer. Nanoscale devices that can deliver cancer prevention and treatment agents will be created. These might include the nanoparticles/liposomes described above for drug delivery. Multicomponent anticancer vaccines will be tested using nanoscale delivery vehicles as described above in the cancer immunotherapy section. Early detection and proteomics. Implantable, stable molecular sensors for detection of cancer-associated biomarkers will be developed; the sensors can be analyzed in situ or collected ex vivo with the results transmitted by wireless technology to the physician. To facilitate mass spectropic analysis of cancer biomarkers, bsmartQ collection platforms will be designed. Imaging diagnostics. For sensitive and precise imaging, bsmartQ injectable, targeted nanoparticles will be developed that allow for single-cell resolution of tumors. Nanoscale devices will be engineered to address the biological diversity of cancer cells that make up a particular tumor. Multifunctional therapeutics. There is a need for nanoscale devices that integrate diagnostic and therapeutic functions. An example of this is the QD nanoparticles that can be used for imaging and drug delivery. In a related area, bsmartQ therapeutic devices will be tested that can control the spatial and temporal release of drugs while monitoring the efficacy of the treatment. Quality of life enhancement in cancer care. Nanotechnologies will be brought to bear on this important aspect of cancer treatment. Nanoscale devices will be developed to optimally deliver medications for conditions such as pain, nausea, loss of appetite, depression, and so on. Interdisciplinary training. Success of the nanotechnology initiative will hinge on the ability of researchers from diverse disciplines to communicate and interact effectively. Thus, there will be extensive efforts to provide cross-training in molecular and systems biology to nanotechnology engineers, physicists, and chemists, and in nanotechnology to cancer researchers. In addition, there will be the creation of new interdisciplinary coursework/degree programs to train scientists skilled in both cancer biology and nanotechnology.

Excellence (CCNEs). The CCNEs will represent the core units for the NCI Alliance for Nanotechnology, and will be a national resource that will integrate the basic and clinical sciences with engineering to develop and apply nanotechnology to cancer research to accelerate the application of this science to the clinic. The CCNEs will work closely and provide expertise for existing programs, such as the nationTs Comprehensive Cancer Centers, and Specialized Programs of Research Excellence, and large population infrastructures, such as the breast and colon cancer family registries. The NCI will commit approximately $90.8 million for the FY 20052009 period, and about $20 million in 2005 alone. Second, the CNPlan will fund cross-disciplinary training programs as a means to encourage the creation of multidisciplinary teams needed to integrate nanotechnology and cancer biology. The grants will support the career development of individuals from the basic, biomedical, clinical, information, and engineering sciences who are pursuing research that applies advances in nanotechnology to the prevention, detection, diagnosis, and treatment of cancer. The NCI is committing about $15.5 million over a 3-year period to fund a total of 36 new grants. Third, the CNPlan will fund focused nanotechnology development initiatives that will be milestone driven and product oriented, with an emphasis on commercialization through both small and large companies in the private sector. The proposed projects that will be eligible for consideration include molecular imaging and early detection, in vivo imaging, reporters of therapeutic efficacy, multifunctional therapeutics, prevention and control of cancer, and enablers of research. The NCI is scheduled to commit $7 million for FY 2005, and an applicant may be awarded up to $1 million a year for a 5-year project. Fourth, the CNPlan will fund projects that apply nanotechnology in cancer biology and translational research through basic research project grants and other mechanisms. As R01 grants have not been the best mechanism for funding investigator-initiated technology and development projects, the NCI will make also make use of program announcements and requests for applications or proposals, as well as a variety of program management and funding mechanisms that have been shown to succeed in previous technology development programs. The NCI will also consider opportunities through the Small Business Innovation Research/Small Business Technology Transfer programs, as well as administrative supplements to existing grants to expedite integration of medical nanotechnologies into the NCI research program. Summary The good news this year was that, for the first time, deaths caused by heart disease were fewer than those by cancer for people younger than 85 [89]. The bad news was that there were more deaths caused by cancer than from heart disease.

Funding for the nanotechnology initiative or CNPlan will be provided in 4 areas. The first is for the development of up to 5 Centers of Cancer Nanotechnology

E.S. Kawasaki, A. Player / Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101109

107

If the incidence of deaths from cancer had dropped as much as heart disease, cancer would be approaching the status of a rare disease. Instead, overall cancer mortality has changed little during this period, while deaths from heart disease have plummeted almost by half. Although cancer may be more complex than cardiovascular disease (an arguable opinion), it is not inconceivable that lifestyle changes (smoking cessation) and new drugs developed from nanotechnological and other medical advances could create the same laudable statistic for cancer as heart disease in the next decade. Nanotechnological studies are not new. In essence, all drug molecules can be considered as nanoengineered structures. What is new is the inclusion of a number of other nano-based approaches to medicinal studies. For example, the antibody-conjugated QDs show promise in in vivo targeting that can be potentially useful for detecting disease, while both QDs and nanosized liposomes demonstrate significant improvement over conventional, lessdirected drug delivery protocols. Monoclonal antibodies and vaccines directed against tumors have been extensively studied, while antisense oligonucleotides and siRNAs are more recent additions to the nanomedicine repertoire. Tumor destruction via the use of nanoshells for thermal ablation is also being examined and shows promise as a nonsurgical method for tumor removal. Precise knowledge of the normal and cancer genome is at hand, and the structure and function of all genes are now within grasp of the medicinal chemist and drug developers. This will allow the creation of nontoxic, targeted small-molecule drugs for use in the oncology clinic. Due to the complexity of cancers, a combination of approaches will likely be needed for the effective elimination of all tumor cells. The NCI is spearheading a multidisciplinary approach to the study of nanomedicine in an attempt to expedite its progress and direct the benefits to cancer patients. This is a 5-year multi-million-dollar scientific venture, with emphasis on prevention, detection, therapeutics, patient quality of life, and training of researchers, to accelerate the development of nanotechnology for the ultimate defeat of cancer. In conclusion, there is good reason to feel cautious optimism for the cure of cancer by 2015 [90]. An entire knowledge base is focusing its efforts on the disease, which is more than has been done for any other disease. Furthermore, nanotechnologies, as estimated by the National Science Foundation, will become a $1 trillion industry by 2015, and much of this economic effort will be directed toward the healthcare and cancer therapy sectors. For further information into the fascinating field of nanotechnologies, the reader is directed to the following very useful websites: US government: www.nano.gov National Institutes of Health: http://nihroadmap.nih.gov/ nanomedicine National Cancer Institute: http://nano.cancer.gov National Science Foundation: http://nsf.gov/crssprgm/ nano

National Aeronautics and Space Administration: http:// ipt.arc.nasa.gov NanoScience and Technology Institute: www.nsti.org United Kingdom: http://www.nanotec.org.uk Nanotech Info and Directory: www.nanotech-now.com

References
[1] International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature 2001;409: 860 - 921. [2] Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, Sutton GG, et al. The sequence of the human genome. Science 2001;291: 1304 - 51. [3] International Human Genome Sequencing Consortium. Finishing the euchromatic sequence of the human genome. Nature 2004;431: 931 - 45. [4] Player A, Barrett JC, Kawasaki ES. Laser capture microdissection, microarrays and the precise definition of a cancer cell. Expert Rev Mol Diagn 2004;4:831- 40. [5] Joachim C, Emerich DF, Wu X, Liu H, Liu J, Haley KN, et al. To be nano or not to be nano? Nat Mater 2005;4:107 - 9. [6] Alivisatos AP. Semiconductor clustors, nanocrystals, and quantum dots. Science 1996;271:933 - 7. [7] Alivisatos P. The use of nanocrystals in biological detection. Nat Biotechnol 2004;22:47 - 52. [8] Bruchez Jr M, Moronne M, Gin P, Weiss S, Alivisatos AP. Semiconductor nanocrystals as fluorescent biological labels. Science 1998;281:2013 - 6. [9] Smith AM, Gao X, Nie S. Quantum dot nanocrystals for in vivo molecular and cellular imaging. Photochem Photobiol 2004;80: 377 - 85. [10] Anderson SA, Glod J, Arbab AS, Noel M, Ashari P, Fine HA, et al. Noninvasive MR imaging of magnetically labeled stem cells to directly identify neovasculature in a glioma model. Blood 2005; 105:420 - 5. [11] Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. Biomaterials 2005;26: 3995 - 4021. [12] Harisinghani MG, Weissleder R. Sensitive, noninvasive detection of lymph node metastases. Public Libr Sci Med 2004;1:e66. [13] Hutten A, Sudfeld D, Ennen I, Reiss G, Hachmann W, Heinzmann U, et al. New magnetic nanoparticles for biotechnology. J Biotechnol 2004;112:47 - 63. [14] Lian W, Litherland SA, Badrane H, Tan W, Wu D, Baker HV, et al. Ultrasensitive detection of biomolecules with fluorescent dye-doped nanoparticles. Anal Biochem 2004;334:135 - 44. [15] Wu X, Liu H, Liu J, Haley KN, Treadway JA, Larson JP, et al. Immunofluorescent labeling of cancer marker Her2 and other cellular targets with semiconductor quantum dots. Nat Biotechnol 2003; 21:41 - 6. [16] Smith AM, Nie S. Chemical analysis and cellular imaging with quantum dots. Analyst 2004;129:672 - 7. [17] Jaiswal JK, Goldman ER, Mattoussi H, Simon SM. Use of quantum dots for live cell imaging. Nat Methods 2004;1:73 - 8. [18] Jaiswal JK, Mattoussi H, Mauro JM, Simon SM. Long-term multiple color imaging of live cells using quantum dot bioconjugates. Nat Biotechnol 2003;21:47 - 51. [19] Michalet X, Pinaud FF, Bentolila LA, Tsay JM, Doose S, Li JJ, et al. Quantum dots for live cells, in vivo imaging, and diagnostics. Science 2005;307:538 - 44. [20] Han M, Gao X, Su JZ, Nie S. Quantum-dot-tagged microbeads for multiplexed optical coding of biomolecules. Nat Biotechnol 2001;19: 631 - 5.

108

E.S. Kawasaki, A. Player / Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101109 [44] Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov 2005;4:145 - 60. [45] Wissing SA, Kayser O, Muller RH. Solid lipid nanoparticles for parenteral drug delivery. Adv Drug Deliv Rev 2004;56:1257 - 72. [46] Koziara JM, Lockman PR, Allen DD, Mumper RJ. Paclitaxel nanoparticles for the potential treatment of brain tumors. J Control Release 2004;99:259 - 69. [47] Steiniger SC, Kreuter J, Khalansky AS, Skidan IN, Bobruskin AI, Smirnova ZS, et al. Chemotherapy of glioblastoma in rats using doxorubicin-loaded nanoparticles. Int J Cancer 2004;109: 759 - 67. [48] Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Deliv Rev 2004;56:1649 - 59. [49] Hood JD, Bednarski M, Frausto R, Guccione S, Reisfeld RA, Xiang R, et al. Tumor regression by targeted gene delivery to the neovasculature. Science 2002;296:2404 - 7. [50] Yu W, Pirollo KF, Yu B, Rait A, Xiang L, Huang W, et al. Enhanced transfection efficiency of a systemically delivered tumor-targeting immunolipoplex by inclusion of a pH-sensitive histidylated oligolysine peptide. Nucleic Acids Res 2004;32:e48. [51] Farokhzad OC, Jon S, Khademhosseini A, Tran TN, Lavan DA, Langer R. Nanoparticle-aptamer bioconjugates: a new approach for targeting prostate cancer cells. Cancer Res 2004;64:7668 - 72. [52] Cortez-Retamozo V, Backmann N, Senter PD, Wernery U, De Baetselier P, Muyldermans S, et al. Efficient cancer therapy with a nanobody-based conjugate. Cancer Res 2004;64:2853 - 7. [53] Gillies ER, Frechet JM. Dendrimers and dendritic polymers in drug delivery. Drug Discov Today 2005;10:35 - 43. [54] Schiffelers RM, Ansari A, Xu J, Zhou Q, Tang Q, Storm G, et al. Cancer siRNA therapy by tumor selective delivery with ligandtargeted sterically stabilized nanoparticle. Nucleic Acids Res 2004; 32:e149. [55] Whitesides GM. The brightQ size in nanobiotechnology. Nat Biotechnol 2003;21:1161 - 5. [56] Sun Q, Wang Q, Rao BK, Jena P. Electronic structure and bonding of Au on a SiO2 cluster: a nanobullet for tumors. Phys Rev Lett 2004; 93:186803/1-4. [57] Hood L, Heath JR, Phelps ME, Lin B. Systems biology and new technologies enable predictive and preventative medicine. Science 2004;306:640 - 3. [58] Hood L, Perlmutter RM. The impact of systems approaches on biological problems in drug discovery. Nat Biotechnol 2004;22: 1215 - 7. [59] Kamb A. Opinion: whats wrong with our cancer models? Nat Rev Drug Discov 2005;4:161 - 5. [60] Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 2004;3:391 - 400. [61] Groner B, Hartmann C, Wels W. Therapeutic antibodies. Curr Mol Med 2004;4:539 - 47. [62] von Eschenbach AC. A vision for the National Cancer Program in the United States. Nat Rev Cancer 2004;4:820 - 8. [63] Arkin MR, Wells JA. Small-molecule inhibitors of protein-protein interactions: progressing towards the dream. Nat Rev Drug Discov 2004;3:301 - 17. [64] Brooijmans N, Kuntz ID. Molecular recognition and docking algorithms. Annu Rev Biophys Biomol Struct 2003;32:335 - 73. [65] Card GL, Blasdel L, England BP, Zhang C, Suzuki Y, Gillette S, et al. A family of phosphodiesterase inhibitors discovered by cocrystallography and scaffold-based drug design. Nat Biotechnol 2005;23:201 - 7. [66] Erlanson DA, Wells JA, Braisted AC. Tethering: fragment-based drug discovery. Annu Rev Biophys Biomol Struct 2004;33:199 - 223. [67] Lipinski C, Hopkins A. Navigating chemical space for biology and medicine. Nature 2004;432:855 - 61. [68] Rees DC, Congreve M, Murray CW, Carr R. Fragment-based lead discovery. Nat Rev Drug Discov 2004;3:660 - 72.

[21] Mattheakis LC, Dias JM, Choi YJ, Gong J, Bruchez MP, Liu J, et al. Optical coding of mammalian cells using semiconductor quantum dots. Anal Biochem 2004;327:200 - 8. [22] Jain RK, Stroh M. Zooming in and out with quantum dots. Nat Biotechnol 2004;22:959 - 60. [23] Jiang W, Papa E, Fischer H, Mardyani S, Chan WC. Semiconductor quantum dots as contrast agents for whole animal imaging. Trends Biotechnol 2004;22:607 - 9. [24] Larson DR, Zipfel WR, Williams RM, Clark SW, Bruchez MP, Wise FW, et al. Water-soluble quantum dots for multiphoton fluorescence imaging in vivo. Science 2003;300:1434 - 6. [25] Akerman ME, Chan WC, Laakkonen P, Bhatia SN, Ruoslahti E. Nanocrystal targeting in vivo. Proc Natl Acad Sci U S A 2002;99: 12617 - 21. [26] Voura EB, Jaiswal JK, Mattoussi H, Simon SM. Tracking metastatic tumor cell extravasation with quantum dot nanocrystals and fluorescence emission-scanning microscopy. Nat Med 2004;10: 993 - 8. [27] Jakub JW, Pendas S, Reintgen DS. Current status of sentinel lymph node mapping and biopsy: facts and controversies. Oncologist 2003;8: 59 - 68. [28] Kim S, Lim YT, Soltesz EG, De Grand AM, Lee J, Nakayama A, et al. Near-infrared fluorescent type II quantum dots for sentinel lymph node mapping. Nat Biotechnol 2004;22:93 - 7. [29] Gao X, Cui Y, Levenson RM, Chung LW, Nie S. In vivo cancer targeting and imaging with semiconductor quantum dots. Nat Biotechnol 2004;22:969 - 76. [30] Ghoroghchian PP, Frail PR, Susumu K, Blessington D, Brannan AK, Bates FS, et al. Near-infrared-emissive polymersomes: self-assembled soft matter for in vivo optical imaging. Proc Natl Acad Sci U S A 2005;102:2922 - 7. [31] Petri-Fink A, Chastellain M, Juillerat-Jeanneret L, Ferrari A, Hofmann H. Development of functionalized superparamagnetic iron oxide nanoparticles for interaction with human cancer cells. Biomaterials 2005;26:2685 - 94. [32] Lotze MT, Wang E, Marincola FM, Hanna N, Bugelski PJ, Burns CA, et al. Workshop on cancer biometrics. Identifying biomarkers and surrogates of cancer in patients: a meeting held at the Masur Auditorium, National Institutes of Health. J Immunother 2005;28:79 - 119. [33] Posadas EM, Simpkins F, Liotta LA, MacDonald C, Kohn EC. Proteomic analysis for the early detection and rational treatment of cancerrealistic hope? Ann Oncol 2005;16:16 - 22. [34] Georganopoulou DG, Chang L, Nam JM, Thaxton CS, Mufson EJ, Klein WL, et al. Nanoparticle-based detection in cerebral spinal fluid of a soluble pathogenic biomarker for AlzheimerTs disease. Proc Natl Acad Sci U S A 2005;102:2273 - 6. [35] Keating CD. Nanoscience enables ultrasensitive detection of AlzheimerTs biomarker. Proc Natl Acad Sci U S A 2005;102:2263 - 4. [36] Nam JM, Thaxton CS, Mirkin CA. Nanoparticle-based bio-bar codes for the ultrasensitive detection of proteins. Science 2003;301: 1884 - 6. [37] Liotta LA, Ferrari M, Petricoin E. Clinical proteomics: written in blood. Nature 2003;425:905. [38] Sahoo SK, Labhasetwar V. Nanotech approaches to drug delivery and imaging. Drug Discov Today 2003;8:1112 - 20. [39] Vasir JK, Reddy MK, Labhasetwar V. Nanosystems in drug targeting: opportunities and challenges. Curr Nanosci 2005;1:47 - 64. [40] Kipp JE. The role of solid nanoparticle technology in the parenteral delivery of poorly water-soluble drugs. Int J Pharm 2004;284: 109 - 22. [41] Muller RH, Keck CM. Challenges and solutions for the delivery of biotech drugsa review of drug nanocrystal technology and lipid nanoparticles. J Biotechnol 2004;113:151 - 70. [42] Rabinow BE. Nanosuspensions in drug delivery. Nat Rev Drug Discov 2004;3:785 - 96. [43] Horn D, Rieger J. Organic nanoparticles in the aqueous phase-theory, experiment, and use. Angew Chem Int Ed 2001;40:4330 - 61.

E.S. Kawasaki, A. Player / Nanomedicine: Nanotechnology, Biology, and Medicine 1 (2005) 101109 [69] Shoichet BK. Virtual screening of chemical libraries. Nature 2004; 432:862 - 5. [70] Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov 2002;1:493 - 502. [71] Druker BJ. Imatinib as a paradigm of targeted therapies. Adv Cancer Res 2004;91:1 - 30. [72] Burgess MR, Skaggs BJ, Shah NP, Lee FY, Sawyers CL. Comparative analysis of two clinically active BCR-ABL kinase inhibitors reveals the role of conformation-specific binding in resistance. Proc Natl Acad Sci U S A 2005. [73] Gumireddy K, Baker SJ, Cosenza SC, John P, Kang AD, Robell KA, et al. A non-ATP-competitive inhibitor of BCR-ABL overrides imatinib resistance. Proc Natl Acad Sci U S A 2005;102:1992 - 7. [74] Shah NP, Tran C, Lee FY, Chen P, Norris D, Sawyers CL. Overriding imatinib resistance with a novel ABL kinase inhibitor. Science 2004;305:399 - 401. [75] Crooke ST. Antisense strategies. Curr Mol Med 2004;4:465 - 87. [76] Jhaveri MS, Rait AS, Chung KN, Trepel JB, Chang EH. Antisense oligonucleotides targeted to the human alpha folate receptor inhibit breast cancer cell growth and sensitize the cells to doxorubicin treatment. Mol Cancer Ther 2004;3:1505 - 12. [77] Dorsett Y, Tuschl T. siRNAs: applications in functional genomics and potential as therapeutics. Nat Rev Drug Discov 2004;3:318 - 29. [78] Downward J. Use of RNA interference libraries to investigate oncogenic signalling in mammalian cells. Oncogene 2004;23:8376 - 83. [79] Fuchs U, Damm-Welk C, Borkhardt A. Silencing of disease-related genes by small interfering RNAs. Curr Mol Med 2004;4:507 - 17.

109

[80] Gilboa E. The promise of cancer vaccines. Nat Rev Cancer 2004; 4:401 - 11. [81] Pardoll D. Does the immune system see tumors as foreign or self? Annu Rev Immunol 2003;21:807 - 39. [82] Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med 2004;10:909 - 15. [83] Fifis T, Gamvrellis A, Crimeen-Irwin B, Pietersz GA, Li J, Mottram PL, et al. Size-dependent immunogenicity: therapeutic and protective properties of nano-vaccines against tumors. J Immunol 2004;173: 3148 - 54. [84] Stauffer PR, Goldberg SN. Introduction: thermal ablation therapy. Int J Hyperthermia 2004;20:671 - 7. [85] Brongersma ML. Nanoscale photonics: nanoshells: gifts in a gold wrapper. Nat Mater 2003;2:296 - 7. [86] Hirsch LR, Stafford RJ, Bankson JA, Sershen SR, Rivera B, Price RE, et al. Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance. Proc Natl Acad Sci U S A 2003; 100:13549 - 54. [87] OTNeal DP, Hirsch LR, Halas NJ, Payne JD, West JL. Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles. Cancer Lett 2004;209:171 - 6. [88] Alper J. US NCI launches nanotechnology plan. Nat Biotechnol 2004;22:1335 - 6. [89] Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, et al. Cancer statistics, 2005. CA Cancer J Clin 2005;55:10 - 30. [90] Blagosklonny MV. How cancer could be cured by 2015. Cell Cycle 2005;4.

Das könnte Ihnen auch gefallen