Sunteți pe pagina 1din 14

Nanotechnology and its Role in Cancer Treatment

By: Vanessa Isla and Japun Padda Humber College REGA 505 Biotechnology Peivand Pirouzi December 6, 2012

Isla and Padda 1

Introduction The World Health Organization (WHO) claims that cancer is the leading cause of death worldwide accounting for about 7.9 million deaths in 2007, and deaths are estimated to rise to about 12 million by the year 2030 (WHO 2011). The rates for new cancer cases and deaths in the U.S. for the year 2007, reported by the Center for Disease Control and Prevention, were 543.2 new cases/100, 000 and 217.8 deaths/100, 000 for men, and 409.4 new cases/100, 000 and 150.9 deaths/100, 000 women (CDC 2011). In Canada, the incidence rate for all cancers in the year 2007 was reported to be 405.2 per 100, 000 for men and women (PHAC 2011). It is evident that cancer is a worldwide concern, and will continue to be a major public health concern especially since the incidence rates are considerably. Cancer has always been a focus of research where there is always a quest for new ways to detect and treat cancer, but the improvement and development of treatment strategies seemed to remain constant over the past 30 years with no major innovations (Farrell et al. 2011). The application of nanotechnology to medicine may be the solution to finding innovative ways to treat cancer more effectively at the molecular level. Nanotechnology is defined as intentional design, characterization, production, and applications of materials, structures, devices, and systems by controlling their size and shape in the nanoscale range (1 to 100 nm). (Gonsalves et al. 2008; Kim et al. 2010). The medical application of nanotechnology led to the emergence of a new field known as nanomedicine, which can serve as a way to improve medical diagnosis, treatment, and prevention of diseases using molecular tools and knowledge of the human body (Gonsalves et al. 2008). Nanomedicine does not only entail the therapeutic products themselves but also the promise of combining its ability to be able to deliver the therapeutic component successfully to its target cell(s) and release it as a response to a physiological event thereby

Isla and Padda 2

performing a specific task (Chan 2006). The promising attributes of applying nanotechnology to cancer therapy through nanomedicine holds great potential for improving the health of the public while possibly finding innovative ways to effectively treat cancer. What makes Nanomedicine attractive for cancer therapy? In hopes of fostering further new ways to approach cancer research and care, in 2004, the National Cancer Institutes (NCI) Alliance for Nanotechnology in Cancer was initiated for the purpose of: (1) providing researchers with the opportunity to study and manipulate macromolecules during the earlier stages of cancer, (2) detecting cancer-related molecules in a sensitive and rapid manner, (3) providing support for novel and highly effective therapeutic agents (Farrell et al. 2011; Amiji 2010). The uses of nanotechnology applications for cancer therapeutics are focused on improving drug delivery and toxicities associated with cytotoxic chemotherapies (Amiji 2010). The multifunctionality of nanoparticles is what makes them quite ideal in treating cancers. Nanoparticles have the ability to deliver the therapeutic component directly to the tumour site while selectively eradicating cancerous cells without forfeiting healthy cells in the process (Farell et al. 2011). This allows for fewer side effects that are commonly associated with chemotherapies for cancer. Currently, the primary treatment options for cancers are: surgical procedures to remove tumour(s), radiation, and chemotherapy (Goncalves et al. 2008). It is important to bear in mind that not all cancers will possess the same characteristics; therefore not all chemotherapeutics will have desirable effects, or selectivity, towards all types of cancer (Li et al. 2011). This nonspecificity that some therapeutics may have can result in poor therapeutic efficacy and also damage to normal tissue, such as bone marrow, skin, kidney, and gastrointestinal mucosa (Nalwa

Isla and Padda 3

and Webster 2007). Nanomedicines are believed to be multifunctional, which could resolve the issue that other therapeutics have with biological barriers. In regards to nanotechnology, PopeHarman et al. (2007) suggests that selective targeted therapy will be enhanced through the use of multimodal targeting, in which therapeutics are localized physically by more than one mechanism. Potential solutions that nanomedicine may have to surpass biological barriers are to provide therapeutic agent(s) that have the ability to comprise of co-localized delivery. Also, an alternative means of administration besides the use of injection, which can be uncomfortable for some patients. Instead, such materials can be inhaled or swallowed (Pope-Harman et al. 2007). Longer dosing intervals can also be made possible using nanomaterials, and it could potentially increase the amount of drug that reaches malignant cells, therefore potentially minimizing adverse effects (Pope-Harman et al. 2007). All of these potential factors that are contributed by using nanomedicine can play a major role in improving patient compliance, which could overall improve the efficacy of the therapeutic agent in use. What should be considered when developing an effective nanoparticle drug delivery system is drug targeting, which may guarantee preferential detection and killing of cancer cells while minimizing the effects on healthy cells (Nalwa and Webster 2007; Loomis et al 2011). Nanoparticles have unique physical properties (size, charge, biocompatibility, solubility) which can be manipulated to increase circulation half-life. In turn, this can lead to increased accumulation of particles and associated drug cargo at the tumor site (Farrell et al. 2011). Figure 1 illustrates a multifunctional nanocarrier which is an ideal design for a nanoparticle for use as a drug delivery shuttle for a cancer therapeutic (See Appendix A). Ways in which the multifunctionality of the nanoparticle can be engineered so that it may serve as a mediate drug delivery system could be to (1) incorporate a coating of a neutral polymer, such as poly(ethylene

Isla and Padda 4

glycol), which would allow for evasion of the mononuclear phagocyte system resulting in prolonged circulation in the bloodstream. This would also help stabilize the particle as well as decrease its likelihood of being cleared from the plasma (Loomis et al. 2011). (2) Incorporation of imaging agents into the core of the nanoparticle can also be done, which would allow for real time visualization of the nanoparticles biodistribution. These agents could play a role in cancer diagnosis and prediction of therapy (Loomis et al. 2011). (3) Incorporation of a capsule containing a payload of the therapeutics in use can be done, in which the nanoparticle will release upon interaction with the selected cell (Loomis et al. 2011). (4) Targeting ligands can also be incorporated onto the surface of the nanoparticle which would allow for receptor mediated endocytosis into the selected cell, thereby increasing delivery specificity (Loomis et al. 2011). (5) A fusogenic lipid can also be incorporated to trigger release of the nanoparticles contents, for example it can do this once placed in an acidic environment (Loomis et al. 2011). Trigger release is significant in the success and functionality of the nanoparticle if it were to serve as a drug carrier. The reason for this is solely due to the requirement of its contents to be released in order for the drug to be available and cause an effect. The ways in which the drug release is timed as well as the degree of its release are important for tumour therapy (Loomis et al. 2011). Strategies that convey the ability to control the trigger to release the contents of the nanoparticle are essential in developing an effective therapeutic drug delivery system (Loomis et al. 2011). Examples of triggers are heat, pH change, ultrasound, and enzymes, which all may control the release of the nanoparticles contents.

Isla and Padda 5

Technical Aspects of the use of Nanomedicine for Cancer Treatment In order for nanoparticles to be used for biomedical applications it needs to be biocompatible and biodegradable so that the nanoparticles can be excreted by the kidneys or bile, and does not have toxic effects on the individual (Lamprecht 2009). Nanoparticles are ideal because they are able to cross biological membranes and access cells, tissues, and organs that larger particles cannot, such as molecules of various drugs (Pathak et al. 2007). Drug payloads can be quite large, due to large surface-to-volume ratios at the nanoscale (Farrell et al. 2011). Engineers are able to choose a variety of ways to dress the surface of a nanoparticle that could make it more biocompatible and selective in its targeting of biological molecules (Kim et al. 2010). This can be done by coating it with polymers or biorecognition molecules as described in figure 1. With nanomaterials, the high ratio of surface area to volume permits high surface loading of therapeutic agents (Kim et al. 2010). The next question to be asked is how all these characteristics of a nanomedicine work together in treating cancer: Tumours normally lack blood vessels of their own and therefore steal their nutrients (i.e. oxygen and glucose) from the surrounding tissues during the earliest stages of cancer (Grossman and McNeil 2012). More of the nutrients are directed towards the cells at the tumours periphery than its core, therefore starving the core (Grossman and McNeil 2012). These starving cells of the tumours core releases proteins, which signal the tumours oxygen-starved state, that diffuse outward until reaching nearby blood vessels. Growth of new blood vessels are stimulated and can then supply the tumour with oxygen and other nutrients thereby sustaining rapid cell growth and replication. This whole process is known as angiogenesis and is the hallmark of cancer (Hanahan and Weinberg 2000). The rapid growth of these angiogenic cells cause these cells to be

Isla and Padda 6

irregular and leaky resulting in more and larger gaps in their walls compared to healthy blood vessels (Grossman and McNeil 2012). Depending on where the tumour is located in the body and what stage of development it is in the size of the gaps vary but range from a few hundred nanometers to a few microns (Grossman and McNeil 2012). Nanoparticles that are between 10nm to 300nm in diameter can significantly pass through these gaps in the blood vessels supplying tumours but not significantly penetrate healthy tissue (where the pores in normal blood vessels are 2-6nm in size) (Grossman and McNeil 2012). Nanoparticles are also able to selectively accumulate in tumour tissue by means of a phenomenon called the enhanced permeability and retention (EPR) effect (Grossman and McNeil 2012). The drug payload is then released by an associated trigger and eventually the cancer cells of the tumor undergo apoptosis. Approved FDA Nano-drugs There are already FDA-approved nano-drugs on the market: Abraxane and Doxil (illustrated in Figure 2, See Appendix B.) both have proven to be beneficial to cancer patients (Farell et al. 2011, Grossman and McNeil 2012). Abraxane is a nanoparticle of the drug paclitaxel, which is a chemotherapeutic that is poorly soluble in water. Abraxane uses a nanoparticle that is bound by the blood protein albumin to encapsulate and solubilize paclitaxel. Abraxane has proven to be more effective and less toxic than is comparator Taxol (Grossman and McNeil 2012). Doxil is a nanosized liposome composed of crystals of the drug doxorubicin encapsulated in a lipid layer and coated with polyethylene glycol (PEG). Free doxorubicin is commonly known to be toxic to the heart and cause damage to the hearts cardiac muscles. The nanoparticle delivery system of Doxil allows for the distribution of the drug in the body with minimal distribution to

Isla and Padda 7

the heart. Doxil is known to cause ulcerations to the skin due to the distribution of the drug to the skin but this adverse side effect may be more preferable than cardiac toxicity (Grossman and McNeil 2012). Based on testings done to compare the side-by-side efficacy of the nanodrug to the free drug, nanodrugs were more efficacious and allowed for the use of a lower dosage (Farrell et al. 2011). Today there are about 82 ongoing clinical trials involving nanoparticles to treat cancer (refer to Table 1 in the Appendix for examples of nanomaterials in clinical use for cancer therapy) (Grossman and McNeil 2012). Existing Concerns About the use of Nanomedicine Existing concerns about nanomedicine and its use for cancer therapy involve the confusion and disagreement with the current definition of Nanotechnology. It is considered an umbrella term used to define products, processes and properties at the nano scale. In terms of safety and regulations of nanotechnologies in the use of cancer treatment, issues of biodistribution and toxicity must be addressed. Biodistribution and cellular uptake of nanoparticles depends on its size, shape and other properties (Farrell et al. 2011). There is also the concern of mass producing nanomedicines. The ability to reproducibly manufacture nanomedicines at large scales with high levels of control over the physicochemical properties remains a major hurdle (Grossman and McNeil 2012). Though many labs can make nanomedicines at the milligram levels for proof-ofconcept in vitro studies, the costs and manufacturing challenges associated with making largescale batches of the same quality remain great (Grossman and McNeil 2012). With this in mind, nanomedicines are indeed expensive. Using the two FDA-approved nanomedicines in the market for cancer therapy as an example, average per-dose cost of Abraxane and Doxil exceed $5000 in 2009 compared with less than $500 for Taxol and less than $200 for doxorubicin (Grossman and McNeil 2012). However, nanomedicines are documented to be less toxic to

Isla and Padda 8

healthy tissues and provide a significantly better quality of life than the molecular counterparts but with only modest improvements in overall survival (Grossman and McNeil 2012). With this in mind, since nanomedicines are considerably more expensive than their small-molecule competitors, they must show a dramatic increase in patient survival in their clinical data (Grossman and McNeil 2012). Improvement in Nanotechnology FDA Regulations Currently, FDAs regulatory regime is based on the main idea that large particle versions of products are considered to be safe, and this can be presumed that nanoversions of the same bulk products would be safe as well. FDA believes that both bulk products and nanoparticles are bioequivalent. This regulatory regime is scientifically flawed. Firstly, not all nanoscale materials are created equal and their toxicities depend on factors such as size, charge, shape, polarity, etc (Bawa, 2011). In addition, as the size of particle decreases, a great proportion of its atoms are located on the surface relative to its core, often rendering the particle more reactive over its conventional bulk counterpart (Bawa, 2011). The decrease in particle size increases reactivity of the particle, dissolution rate, saturation solubility, and in whole, its toxicological risk and a serious public health concern (Bawa, 2011). Hence, one main improvement to take the first step to this serious public health concern would be that FDA needs to start assessing nanoparticles on a case-by-case basis to determine if general trends or themes can be identified and whether new regulatory procedures are needed to replace the current ones on hand (Bawa, 2011). Conclusion There is tremendous potential for the use of nanoparticles in cancer treatment and diagnosis, even though many nanoparticles that have been developed are currently undergoing research,

Isla and Padda 9

and still remain in clinical trials. There are already FDA-approved patented nanoparticles out in the market, such as iron oxide nanoparticle-based magnetic resonance imaging agents (e.g. Feridex), which are routinely used in the clinic, as well as Doxil (doxorubicin in long circulating liposomes) and Abraxane (paclitaxel in albumin nanoparticles) from the therapeutic stand point (Amiji 2011). If there is a way of minimizing the toxicity of certain types of oncology drugs, as well as minimizing the harmful effects that the drugs may impose on an individual then attention should really be made on the development of these nanoparticle drug delivery systems. Once the knowledge of these nanoparticles improve over time the greater the chance of these nanoparticles to actually improve its efficacy and have the potential to serve as a new means of treating cancer.

Appendices Appendix A

Figure 1 Multifunctional nanoparticle (in this case a liposome) (1) has a stealth coating increasing stability and decreasing plasma clearance, (2) has imaging agents allow for real time visualization of nanoparticle biodistribution, (3) carries a payload of therapeutics, (4) actively targets specific cells via a targeting ligand, (5) can be triggered to release its contents (Loomis et al. 2011).

Isla and Padda 10

Appendix B

Figure 2. Abraxane and Doxil are FDA-approved nanomedicines for cancer treatment on the market today. (a) Abraxane, produced by Celgene Corp. is a nanoparticle of the drug paclitaxel bound by the blood protein albumin. (b) Doxil is a Johnson and Johnson product composed of crystals of the drug doxorubicin encapsulated in a lipid layer and coated with polyethylene glycol (PEG) (Grossman and McNeil 2012). Appendix C Table 1. Examples of Nanomaterials in Clinical Use for Cancer Therapy Nanomaterial Trade Name Metallic Iron Oxide Gold Nanoshells Organic Protein Liposome Target Adverse Effects Acute urinary retention Fever Under investigation Cytopenia Manufacturer Current Status MagForce CytImmune Sciences Nanospectra Biosciences Abraxis Bioscience Ortho Biotech In phase 3 clinical trials In phase 2 clinical trials In phase 1 clinical trials FDA Approved FDA Approved FDA Approved In phase 2 clinical trials

NanoTherm Aurimmune Auroshell

Various forms Various Forms Head and neck Breast Various forms

Abraxane Doxil/Caelyx

Polymer

Oncasper

CALAA-01 (Kim et al. 2010)

Hand-foot syndrome, stomatitis Acute Urticaria, lymphoblastic rash leukemia Various Mild renal forms toxicity

RhonePoulenc Rorer Calando

Isla and Padda 11

References: Amiji, M. M. 2011. Nanomedicine for Cancer Therapy [online]. Pharm Res. 28: 181-186. Doi: 10.1007/s11095-010-0261-0. Bawa, R. (2011). Regulating nanomedicine - can the fda handle it?. Current drug delivery, 8, 227-234. Bhattacharyya, S., Kudgus, R. A., Bhattacharyya, R., and Mukherjee, P. 2011. Inorganic Nanoparticles in Cancer Therapy [online]. Pharm Res. 28: 237-259. Doi 10.1007/s11095010-0318-0 BreastCancer.org. 2008. Accessed site at: http://www.breastcancer.org/dictionary/m/metastaticcancer_t.jsp. [Accessed on 11 March 2011] Brewer, M., Zhang, T., Dong, W., Rutherford, M., and Tian, R. 2007. Future approaches of nanomedicine in clinical science. Med Clin N Am. 91(5): 963-1016. CDC. 2011. Rate for New Cancer Cases and Deaths by Race/Ethnicity and Sex. Accessed at: http://www.cdc.gov/features/dsCancerDisparities/. [Accessed site on 14 March 2011]. Chan, V.S.W. 2006. Nanomedicine: An unresolved regulatory issue. Regulatory Toxicology and Pharmacology, 46:218224. doi:10.1016/j.yrtph.2006.04.009 Chanda, N., Shukla, R, Zambre, A., Mekapothula, S., Kulkami, R. R., Katti, K., Bhattacharyya, K., Fent, G. M., Casteel, S. W., Boote, E. J., Viator, J. A., Upendran, A., Kannan, R., and Katti, K. V. 2011. An Effective Strategy for the Synthesis of Biocompatible Gold Nanoparticles Using Cinnamon Phytochemicals for Phantom CT Imaging and Photoacoustic Detection of Cancerous Cells [online]. Pharm Res. 28: 279-291. Doi: 10.1007/s11095-010-0276-6 Choi, Y., Kwak, J., and Park, J. W. 2010. Nanotechnology for Early Cancer Detection [online]. Sensors. 10: 428-455. Doi:10.3390/s100100428 Drbohlavova, J., Adam, V., Kizek, R., and Hubalek, J. 2009. Quantum Dots Characterization, Preparation and Usage in Biological Systems [online]. International Journal of Molecular Sciences. 10: 656-673. Doi:10.3390/ijms10020656. Farrell, D., Ptak, K., Panaro, N. J., Grodzinski, P. 2011. Nanotechnology-Based Cancer Therapeutics Promise and Challenge Lessons Learned Through the NCI Alliance for Nanotechnology in Cancer [online]. Pharm Res. 28: 273-278. Doi: 10.1007/s11095-0100214-7. Gonsalves, K.E., Halberstadt, C.R., Laurencin, C.T., and Nair, L. S. 2008. Biomedical nanostructures. John Wiley & Sons, Inc. New Jersey. pp 49, 409-437. Grossman, J. H. and McNeil, S. E. 2012. Nanotechnology in Cancer Medicine. Physics Today. 65(8): 38-42. Doi: 10.1063/PT.3.1678

Isla and Padda 12

Hanahan, D. and Weinberg, R.A. 2000. The hallmarks of cancer. Cell. 100(1): 57-70 Heidel, J. D. and Davis, M. E. 2011. Clinical Developments in Nanotechnology for Cancer Therapy [online]. Pharm Res. 28: 187-199. Doi 10.1007/s11095-010-0178-7. Kateb, B., Chiu, K., Black, K. L., Yamamoto, V., Khalsa, B., Ljubimova, J. Y., Ding, H., Patil, R., Portilla-Arias, J. A., Modo, M., Moore, D. F., Farahani, K., Okun, M. S., Prakash, N., Neman, J., Ahdoot, D., Grundfest, W., Nikzad, S. and Heiss, J.D. 2011. Nanoplatforms for constructing new approaches to cancer treatment, imaging, and drug delivery: What should be the policy? [online]. NeuroImage. 54: S106-S124. Doi:10.1016/j.neuroimage.2010.01.105 Kim, B.Y.S., Rutka, J.T. and Chan, W.C.W. 2010. Nanomedicine. The New England Journal of Medicine. 363: 2434-43. Lamprecht, A. 2009. Nanotherapeutics: Drug delivery concepts in nanoscience. Pan Stanford Publishing Pte. Ltd. Singapore. Pp. 4, 18-19, 94-95, Li, N., Wang, J., Yang, X., and Li, L. 2011. Novel nanogels as drug delivery systems for poorly soluble anticancer drugs [online]. Colloids and Surfaces B: Biointerfaces. 83: 237-244. Doi:10.1016/j.colsurfb.2010.11.027. Liu, C., Yang, T., Wang, C., Chien, C., Chen, S. Wang, C., Leng, W., Hwu, Y., Lin, H., Lee, Y., Cheng, C., Je, J. H., and Margaritondo, G. 2009. Enhanced photocatalysis, colloidal stability and cytotoxicity of synchrontron X-ray synthesized Au/TiO2 nanoparticles [online]. Materials Chemistry and Physics. 117: 74-79. Doi:10.1016/j.matchemphys.2009.05.030. Loomis, K., McNeeley, K., and Bellamkonda, R. V. 2011. Nanoparticles with targeting, triggered, release, imaging functionality for cancer applications [online]. Soft Matter. 7: 839-856. Doi: 10.1039/c0sm00534g. Mitra, M., Dilnawaz, F., Misra, R., Harilal, A., Verma, R. S., Sahoo, S. K., and Krishnakumar, S. 2010. Toxicogenomics of nanoparticulate delivery of etoposide: potential impact on nanotechnology in retinoblastoma therapy [online]. Cancer Nano. pp. 1-16. Doi: 10.1007/s12645-010-0010-4. Morrow, K. J., Bawa, R., and Wei, C. 2007. Recent Advances in Basic and Clinical Nanomedicine. Medical Clinics of North America 91(5): 825. Nalwa, H. S. and Webster, T. J. 2007. Cancer Nanotechnology: Nanomaterials for Cancer Diagnosis and Therapy. American Scientific Publishers. Stevenson Ranch, CA. pp. 3, 8185, 333-344. Oxford Reference Online. 2011. Accessed at: http://www.oxfordreference.com.proxy.lib.uwaterloo.ca/views/SEARCH_RESULTS.htm l?y=7&q=chemotherapy&x=20&ssid=263689462&scope=global&time=0.85616269261 3289. Oxford University Press [accessed on 12 March 2011].

Isla and Padda 13

Pathak, P., Katiyar, V. K., and Giri, S. 2007. Cancer Research Nanoparticles, Nanobiosensors and their use in cancer research [online]. Journal of Nanotechnology Online. 3: 1-14. Doi: 10.2240/azojono0116. Patra, H. K., Dasgupta, A. K., Sarkar, S., Biswas, I., and Chattopadhyay, A. 2010. Dual role of nanoparticles as drug carrier and drug [online]. Cancer Nano. Doi: 10.1007/s12645-0100011-3. PHAC. 2011. Accessed site at: http://dsol-smed.phac-aspc.gc.ca/dsol-smed/cancer/cgibin/cancerchart2?DATA_TYPE=R&YEAR_FROM=92&YEAR_TO=07&CAUSE=799 &AREA=00&AGE=0&SEX=3&CTIME1=View+Chart&CI=NO&SCALE=LINEAR. [Accessed on 14 March 2011]. Pope-Harman, A., Cheng, M. M., Robertson, F., Sakamoto, J., and Ferrari, M. 2007. Biomedical Nanotechnology for Cancer. Med Clin N Am. 91(5): 899-927. Simon, E. 2010. Biological and chemical sensors for cancer diagnosis [online]. Measurement Science and Technology. 21(112002): 1-24. Doi:10.1088/0957-0233/21/11/112002. Vogel, V. 2009. Nanotechnology. Wiley-VCH. Germany. 5: 58-61. Wang(a), C. Liu, C., Chien, C., Chen, H. Hua, T., Leng, W. Chen, H., Kempson, I. M., Hwu, Y., Hsiao, M., Lai, T., Wang, J., Yang, C., Lin, H., Chen, Y., and Margaritondo, G. 2011. Xray synthesized PEGylated (polyethylene glycol coated) gold nanoparticles in mice strongly accumulate in tumors [online]. Materials Chemistry and Physics. 126: 352-356. Doi:10.1016/j.matchemphys.2010.11.014. Wang(b), L., Liu, Y., Li, W., Jiang, X., Ji, Y., Wu, X., Xu, L., Qiu, Y., Zhao, K., Wei, T., Li, Y., Zhao, Y., and Chen, C. 2011. Selective Targeting of Gold Nanorods at the Mitochondria of Cancer Cells: Implications for Cancer Therapy [online]. Nano Letters. 11: 772-780. Doi: org/10.1021/nl103992v. WHO. 2011. Cancer. Accessed site at: http://www.who.int/cancer/en/. [Accessed site on 14 March 2011]. Webster, T. J. 2011. Nanotechnology Enabled In Situ Sensors for Monitoring Health [online]. Springer Science + Business Media. New York, NY. pp. 10, 12. Doi: 10.1007/978-14419-7291-0. Wei, C. 2007. Medical Clinics of North America: Nanomedicine. Elsevier, Inc. Philadelphia, Pennsylvania. 91(5): xiii-xv.

S-ar putea să vă placă și