You are on page 1of 10

Advanced Review

Functionalized radioactive gold nanoparticles in tumor therapy


Raghuraman Kannan,1 Ajit Zambre,1 Nripen Chanda,1 Rajesh Kulkarni,1 Ravi Shukla,1 Kavita Katti,1 Anandhi Upendran,2 Cathy Cutler,3 Evan Boote1 and Kattesh V. Katti1
The development of new treatment modalities that offer clinicians the ability to reduce sizes of tumor prior to surgical resection or to achieve complete ablation without surgery would be a signicant medical breakthrough in the overall care and treatment of prostate cancer patients. The goal of our investigation is aimed at validating the hypothesis that Gum Arabic-functionalized radioactive gold nanoparticles (GA-198 AuNP) have high afnity toward tumor vasculature. We hypothesized further that intratumoral delivery of the GA-198 AuNP agent within prostate tumor will allow optimal therapeutic payload that will signicantly or completely ablate tumor without side effects, in patients with hormone refractory prostate cancer. In order to evaluate the therapeutic efcacy of this new nanoceutical, GA-198 AuNP was produced by stabilization of radioactive gold nanoparticles (198 Au) with the FDA-approved glycoprotein, GA. This review will describe basic and clinical translation studies toward realization of the therapeutic potential and myriad of clinical applications of GA-198 AuNP agent in treating prostate and various solid tumors in human cancer patients. 2011 Wiley
Periodicals, Inc.

How to cite this article:

WIREs Nanomed Nanobiotechnol 2012, 4:4251. doi: 10.1002/wnan.161

INTRODUCTION
rostate cancer in men is the second deadliest cancer after lung cancer. Approximately 217,730 men are diagnosed with prostate cancer resulting in loss of life for 32,050 men.1 Prostate cancer diagnosis begins either with a digital rectal exam during which a doctor feels the prostate to check for irregularity or a blood test to check the level of prostate-specic antigen (PSA) or both.2,3 Recent studies have shown that regular PSA screening did not increase the survival rate over a period of 10 years.4,5 Early detection of prostate tumor by common imaging modalities such as ultrasound (US), computer tomography (CT) is difcult, as the prostate is deep inside the pelvis and harder to access. These clinical impediments will
Correspondence to: KattiK@health.missouri.edu, Kannanr@health.missouri.edu 1 Department of Radiology, University of Missouri, Columbia, MO, USA 2 Nanoparticle Biochem, Inc., Columbia, MO, USA 3 Missouri

University Research Reactor, University of Missouri, Columbia, MO, USA

continue to hinder accurate and early detection of prostate cancer resulting in more cases of androgen dependent and hormone independent prostate cancers and thus posing some of the most vexing questions in medicine.6 The widely used brachy therapy utilize agents of 50100 m in size including 125 I or 103 Pd radioactive seeds, and Y-90 immobilized glass microspheres (Therasphere) to achieve selective internal radiation therapy (SIRT).79 The limited natural afnity of these microspheres toward tumor vasculature coupled with signicantly larger (50100 m) sizes as compared to the porosity of tumor vasculature (150300 nm) causes limited retention and signicant leakage of therapeutic doses away from the tumor site. These problems result in decreased efcacy, acute toxic side effects and lower tumoricidal activity of brachy therapy agents. A central goal of chemo or radiation therapy while treating patients with prostate (and other solid tumors) cancer is to achieve maximum dose intensity at the tumor site while managing drugrelated toxicities to the minimum to non-target
Volume 4, January/February 2012

42

2011 Wiley Periodicals, Inc.

WIREs Nanomedicine and Nanobiotechnology

GA-198 AuNP in tumor therapy

organs. For a vast majority of chemo therapeuticand radiation-induced agents, it is still a signicant challenge to optimize critically important clinical parameters such as plasma concentration and the time of exposure to the diseased tissue to chemotherapy or radiotherapy agents as the manipulation of variables such as dose regimens of drugs and dosing schedules have often failed to achieve the optimum therapeutic payloads. Various targeted strategies being currently employed are of limited benet as the drugs used are rapidly eliminated from the plasma compartment, metabolized into inactive species thus obliterating therapy, and rapidly released from tumor cells due to poor tumor cell/tissue specicity. Therefore, concerted efforts are being focused on the development of clinical modalities to maximize drug exposure to tumor sites following systemic or intratumoral administration. Recently, new approaches using nanoparticle-based delivery systems for the administration of inherently therapeutic radioactive isotopes are being explored.1012 Wang et al. have demonstrated therapeutic effects of emission of 186 Re carried by liposomes into the tumor remnants in a nude rat squamous cell carcinoma xenograft model.13 Following the partial resection of tumor xenografts, the animals were intratumorally injected with 186 Re-labeled or unlabeled (control) and neutrally charged or positively charged 100-nmdiameter liposomes. Tumor efcacy studies indicated that the neutral (n = 4) and cationic (n = 4) liposome treated control groups showed an increase in tumor growth, whereas the 186 Re-neutral-liposome group (n = 8) and the 186 Re-cationic-liposome group (n = 8) presented with a signicantly reduced average nal tumor volume at the end of the study (Day 35). These results demonstrate that the intraoperative use of liposomal therapeutic radionuclides may play an important role in the management of positive surgical margins in advanced head and neck squamous cell carcinoma. French et al. have presented another example of the utility of liposomes as delivery vehicles for radioactive nuclides which involves the application of liposome carrying emitting 186 Re to treat head and neck cancer by direct intratumoral infusion in nude rats. In this study, the authors report average tumor volume reduction for the 186 Re-liposome group on posttreatment on Day 14 to 87.7%.14 This study further provides hope that liposomal 186 Re particles can be effectively used for treating head and neck cancer with minimal side effects after convectionenhanced interventional delivery. Zavaleta et al. have reported biodistribution and pharmacokinetics of 186 Re encapsulated in biotin-liposomes containing
Volume 4, January/February 2012

patent blue dye, and injected intraperitoneally (IP) with avidin in an OVCAR-3 ovarian cancer xenograft model. This study has demonstrated a 30% decrease in the growth of tumor volumes as compared to control groups. This is a compelling evidence for the potential of 186 Re-blue-biotin-liposome/avidin system in treating advanced ovarian cancer involving peritoneal carcinomatosis.15 Amplication of therapeutic doses of X-rays due to encapsulation of tissue mimics on gold foil appears to provide clinical benets. Recent studies have shown radiation dose enhancement up to more than a factor of 100 in an environment of tissue-equivalent polymethylmethacrylate (PMMA) close to the surface of a thin metallic gold foil. These studies involved evaluation of enhancement factors for heavily ltered X-rays (40120 kV tube potential) under backscatter conditions, using thin-lm radiation detectors with sub-micrometer resolution. Under in vitro models, enhanced biological effects have been observed using monolayers of C3H 10T1/2 mouse embryo broblasts exposed in intimate contact with the gold surface. Dosimetry calculations have indicated that a dose of 100 mGy, 80 kV X-rays (measured in homogeneous PMMA) caused a frequency at an inserted gold surface comparable to that obtained with a dose of about 4.5 Gy of 60 Co rays in homogeneous PMMA suggesting that nanoparticles of gold which offer superior surface area may show even better dosimetry characteristics.16 Recently, Balogh and coworkers have reported the development of poly {198 Au} radioactive gold dendrimer composite nanodevices of sizes between 10 and 29 nm and demonstrated their utility in targeted radiopharmaceutical dose delivery to tumor.17 Their studies have shown that single intratumoral injection of poly {198 Au0}d = 22 nm composite nanodevices in phosphate-buffered saline (PBS) delivering a dose of 74 Ci, after 8 days, resulted in a statistically signicant 45% reduction in tumor volume when compared with untreated groups. As part of our ongoing efforts toward the application of nanotechnology for the design and development of sophisticated molecular imaging and therapy agents, we have recently discovered biocompatible radioactive gold nanoparticles.1836 The development of the -emitting radioactive Gum Arabic glycoprotein stabilized biocompatible gold nanoparticulate agent (GA-198 AuNP) is an innovative approach for use in treating hormone refractory prostate and other inoperable cancers.37 Nanoparticles of gold are inherently multifunctional in their diagnostic and therapeutic capabilities.30,38 198 Au, provides a desirable
43

2011 Wiley Periodicals, Inc.

Advanced Review

wires.wiley.com/nanomed

-energy emission and half-life for effective destruction of tumor cells/tissue (max = 0.96 MeV; half-life of 2.7 days).37 The range of the 198 Au -particle is sufciently long to provide cross-re effects of radiation dose delivered to cells within the prostate gland and short enough to minimize signicant radiation dose to critical tissues near the periphery of the capsule. The 2.7 day half-life of 198 Au allows time for transport of the doses of the GA-198 AuNP agent from central radiopharmacies to the clinical sites and is amenable for delivery of single GA-198 AuNP administered doses to provide a 105 Gy dose to prostate tumor. In this review, we will discuss the overall clinical translation efforts of GA-198 AuNP therapeutic agent including (1) production of therapeutic radioactive gold nanoparticles; (2) intra-prostate tumoral delivery, retention, and therapeutic efcacy studies; (3) 198 AuNP dosimetry considerations; and (4) the overall oncological relevance of this new GA-198 AuNP therapeutic agent in the context of its potential applications in treating prostate and various other solid tumors.

THPAL (Initiator)

CH3 HO O N H HO O P N H NH

CH3 OH O

CH3

Gum Arabic (stabilizer)

NaAuCI4

GA-AuNP

TEM of GA-AuNP

FIGURE 1 | Synthesis of GA-AuNP.

PRODUCTION OF THERAPEUTIC RADIOACTIVE GOLD NANOPARTICLES


A novel approach for manufacturing therapeutic radioactive AuNPs has been developed at the University of Missouri Research Reactor (MURR). Traditional production methods for AuNPs do not work at tracer levels for the generation of corresponding radioactive gold nanoparticles. Each gold nanoparticle contains several atoms of gold, a number of which could be radioactive 198 Au/199 Au. This should result in a substantial increase in therapeutic dose to the tumor site. In addition, they can be easily tagged with oligonucleotides, tumor-specic proteins, and peptides that are selective for receptors over expressed by cancerous cells/tissue. Traditional gold nanoparticle formulation methods utilize interaction of gold salts with sodium borohydride (NaBH4 ), hydrazine, and other reducing agents for the production of AuNPs at macroscopic levels. Because of kinetic considerations, such methods fail when used at the tracer level to produce radioactive gold nanoparticles. A recent discovery in our laboratory has demonstrated that a trimeric alanine conjugate (P(CH2 NHCH(CH3 )COOH)3 ; THPAL) upon mixing with NaAuCl4 results in the formation of gold nanoparticles of well-dened particulate size (1520 nm) (Figure 1).28,33 The nanoparticle initiator (P(CH2 NHCH(CH3 )COOH)3 ; THPAL) is nontoxic and the gold nanoparticle formation by this
44

method proceeds in aqueous media resulting in biologically benign alanine and phosphoric acid as by products. It is important to note that the reaction outlined in Figure 1 is efcient for the production of nanoparticulate gold using radioactive precursors (198 Au/199 Au) at signicantly low concentrations. For the production of 198 Au, gold foil 530 mg are irradiated at a ux of 8 1013 n/cm2 /s. The radioactive foil is subsequently dissolved with aquaregia, dried down, and reconstituted in 0.51 mL of 0.05 N HCl to form H198 AuCl4 . The radioactive gold solution is added to aqueous solutions of GA, followed by solution of reducing agent, THPAL (P(CH2 NHCH(CH3 )COOH)3 ), for formation of radioactive gold nanoparticles of well-dened particle sizes (1520 nm).28,33 The change in color from pale yellow to purple is diagnostic of plasmon plasmon transition and characteristic of radioactive gold nanoparticle formation, and the solutions at the tracer levels are characterized by UVVis spectrophotometry.

AuNP DOSIMETRY CONSIDERATIONS


Nanoparticles based on -particle emitting isotopes of gold offer a possible means by which effective radiotherapy may be performed for the treatment of prostate cancer without complicating effects and urinary morbidity. For 198 Au, the combination of
TABLE 1 Dosimetry for Different Radioactive Isotopes
Isotope
125 103 131

198

T1/2
17 days

Emission Principle EC, 21 keV photons

I Pd Cs

59.4 days EC, 2735 keV photons 9.7 days EC, 434 keV photons 2.7 days (mean energy = 312 keV), 412 keV photon

198 Au

2011 Wiley Periodicals, Inc.

Volume 4, January/February 2012

WIREs Nanomedicine and Nanobiotechnology

GA-198 AuNP in tumor therapy

TABLE 2 Effectiveness of 198 Au Therapy Compared with Other


Isotopes Used for Prostate Brachy therapy

Radioisotope Indices Target dose, Dtotal (Gy) BED (Gy) TCP (%)
125

DOSIMETRY FOR PROSTATE TUMOR TREATMENT USING RADIOACTIVE GOLD NANOPARTICLES


Brahme40 provides a rationale for an energyindependent stopping power for low energy relativistic electrons, therefore, the range of the electrons is limited by the stopping power in water (1 g/cm3 ). For a maximum -energy of 0.96 MeV (for 198 Au), the maximum range is 4 mm (0.4 g/cm2 )/(1 g/cm3 ). A simple approach to 198 Au dosimetry would be to assume a uniform distribution of activity within a volume that has a diameter greater than the completely slowing down approximation (CDSA) of 4 mm. With such a distribution, charged particle equilibrium is achieved. The dose rate within the volume is then a summation of deposited energy from all the nanoparticles in the volume, which will, in turn, be dependent upon the total activity per unit mass. A dose rate for 198 Au particles will be based on the 0.317 MeV average energy. Assume that a volume of water is approximately 1 cm in diameter; the total mass will be 4/3 r3 0.5 g. On the basis of Attix41 effective dose calculations, the dose rate is D = 3600s/h (3.7 107 disintigrations/s) 0.317 MeV (1.602 1010 Gyg/MeV) 0.5 g

103

Pd

131

Cs

198

Au

145.0 101.7 79 236.2 135.8 6.36

125.0 112.7 95.5 94.1 122.3 2.15

120.0 115.7 97.1 61 118.5 1.27

105.0 124.33 99.2 21.358 104.6 0.41

Teff (day) D (Teff ) (Gy)


Wasted dose (%)

-particle emission with relatively high energy rays results in a low dose, because the short range of the electrons in tissue restricts the dose to the local volume of tissue. In addition, the relatively short half-life of 198 Au (Table 1) presents some intriguing possibilities for increased relative biological effectiveness (RBE). These properties are compared with other radioisotopes used for either low dose rate (LDR) or high dose rate (HDR) prostate brachy therapy in Table 1. 198 Au had been used as early as the 1950s for clinical brachy therapy.39 The effectiveness of 198 Au is shown in Table 2. 198 Au isotope offers superior radiation dose rate as compared to the slower (longer T1/2 ) isotopes. 125 I and 103 Pd are commonly used seed implanted emitters with relatively low (<100 keV) rays. In Table 2, wasted dose is the difference between the total dose delivered (Dtotal ) and the dose over the effective treatment time (Teff ) versus the total dose delivered, e.g. the dose that is the result of the total decay of radioisotope after it has been implanted in the tumor. There is an obvious inverse correlation between BED/TCP and T1/2 . In clinical applications, radioactive gold is encapsulated within 0.1 mm thick platinum, which attenuates the -particle spectrum and thus the dose is due to the high energy emissions. Our new approach which involves implementation of radioactive gold nanoparticles for therapy is reliant upon in vivo stabilization through nanoparticle formation. Therefore, the energy of the -emission by gold is unaffected and leads to high dose rates in vivo. The ability to reliably and accurately predict dose to tissues is a critical aspect in the successful implementation of radiation therapy. Restriction of dose to relevant tumor tissues is important to avoid undesirable side effects. Calculations of dose and dose rate in the treatment volume is vital in understanding the biological effective dose (BED), effective dose(D(Teff ), T1/2 dependent) and the tumor control probability (TCP) based on radiobiological survival models.
Volume 4, January/February 2012

12 Gy/h/mCi The total dose is then: Dtotal = 1.44D0 T1/2 where D0 is the initial dose rate, Dtotal is the total dose. The initial dose rate will be based on the size of the prostate volume under treatment. Another helpful means to compare radiation treatment is to employ biophysical models. For permanent prostate implants (using gold nanoparticles), the tumor cells are subjected to continuous irradiation during the period which activity is present. The instantaneous dose rates will vary over time. Parameters for prostate cancer are applied for calculations of the radiobiological indices based on the linear-quadratic cell-inactivation model. In this case, we use the recommended values by Wang et al.42 = 0.15/Gy, = 0.05/Gy2 , Tp = 42 days and the repair half-life = 0.27h. Tp is the potential doubling time for prostate tumor cells. Teff = Tavg ln Dtotal TD T1/2
45

2011 Wiley Periodicals, Inc.

Advanced Review

wires.wiley.com/nanomed

Teff is dened as the time at which the cellinactivation rate equals the rate of cell repopulation for any hypothetically remaining cell. The Dale BED model43 uses a two-critical target of repair of sublethal radiation damage. The repair capability is modeled by a time constant for sublethal damage repair (this is inversely proportional to the repair half-time), and used in the following equation for repair effectiveness (RE). In this equation, is the decay constant for the radionuclide in question. RE(Teff ) = 1 + D0 1 ( ) 1 eTeff 2 (1 e(+)Teff ) +

fact that a general TCP formalism tends to underestimate tumor cure rate when tumor cell repopulation occurs during treatment. As this is less likely to happen with the high dose rates seen in 198 AuNP therapy, we present TCP here as one more possible parameter to consider. TCP = exp[N0 e(BED) ] N0 is the number of tumor cells initially present (e.g., 106 cells in a tumor).

1 e2Teff

IN VIVO TUMOR RETENTION CHARACTERISTICS OF GA-198 AuNP


In order to understand the retention and clearance characteristics of radioactive GA-198 AuNP within tumor, we have performed a detailed in vivo investigation involving intratumoral administration of GA198 AuNP (3.5 Ci/tumor) in SCID mice (n = 5) bearing human prostate cancer xenografts (Figure 2). These studies involved analysis of 198 Au in various organs post euthanasia of animals at 30 min, 1, 2, 4, and 24 h. The blood, various organs, and tumors were collected and counted to measure the amount of radioactivity (Figure 2). Analysis of 198 Au radioactivity revealed that over 75% of the injected dose (ID) of GA-198 AuNP was retained in prostate tumors at 24 h,

The repair effectiveness is then used to calculate the BED: BED = D(Teff ) RE(Teff ) ln 2 Teff Tp

In this model, any dose deposited after Teff is considered to be a wasted dose. The difference between Dtotal and D(Teff ) is the amount of wasted dose and should be as small as possible. Tumor control probability (TCP) is determined by the Poisson probability of inactivating all tumor cells. The BED is the more accepted model in the community, based on the
300
30 min 1 hour

2 hour

4 hour

24 hour

250

200 Avg %D / gm

150

100

50

0
o Bl od ea H rt Lu ng Li ve r l Sp ee n St ac om h d Ki ne y us M cl e n Bo e ad Bl de r Br ai n nc Pa re as or m Tu A or m Tu B

Organs

FIGURE 2 | Biodistribution prole showing retention of GA-198 AuNP in prostate tumor after intratumoral injection.
46
2011 Wiley Periodicals, Inc.

Volume 4, January/February 2012

WIREs Nanomedicine and Nanobiotechnology

GA-198 AuNP in tumor therapy

and was nearly constant from 30 min to 24 h. The GA-198 AuNP nanoparticles exhibited slow clearance (leakage) into the blood with only 0.06%ID/g at 24 h. The uptake of GA-198 AuNP agent in various organs, as shown in Figure 2, at different postinjection time points have further indicated that there is a signicant accumulation of nanotherapeutic agent in the kidneys (10.68 1.72%ID/g) after 24 h, and the main clearance route of 198 Au activity was via urine with 16.77 0.13%ID at 24 h. Lungs, pancreas, and liver exhibited very low uptake at 24 h. These pharmacokinetics features conrmed the excellent retention of therapeutic payloads of GA-198 AuNP nanoparticles within prostate tumors with only minor leakage to non-target organs. Intratumoral administrations of the nonradioactive surrogate of GA-198 AuNP agent (GA-AuNP) in very low concentrations have been investigated in order to ascertain if GA-198 AuNP agent distributes homogeneously within the prostatic tumor (Figure 3). In these experiments, nonradioactive analog, GAAuNP, was administered at 12 g/g of prostate

tumor in mice with prostate tumor xenografts. Postintratumoral administration of GA-AuNP, homogeneity in distribution of this agent was monitored by CT imaging of tumor regions at various times postinjection. The pre- and postadministration images shown in Figure 3 clearly demonstrate that GA198 AuNP agent permeates and distributes homogeneously in prostate tumors within 1 h postadministration and the injected dose (ID) of optimum therapeutic payloads resides within tumors for over 24 h. Encouraged by the excellent tumor retention features of GA-198 AuNP, we have explored therapeutic efcacy studies in order to evaluate tumor ablation properties.

TUMOR ABLATION CHARACTERISTICS OF GA-198 AuNP


The therapeutic efcacy of GA-198 AuNP has been evaluated using prostate tumor-bearing SCID mice models. In this in vivo investigation, solid tumors were allowed to grow for 3 weeks, and animals were randomized (denoted Day 0) into control and treatment groups (n = 7) with no signicant differences in tumor volume. On Day 8, 408 Ci (30 L) of GA198 AuNP was directly administered into the prostate tumor to deliver an estimated dose of 70 Gy. Control SCID mice received 30 L Dulbeccos PBS. Tumor growth was monitored twice each week. Complete in vivo therapeutic data are depicted in Figure 4.37 Tumor growth in the treated animals showed a drastic decrease within 1 week (Day 14) post-treatment with respect to the control group. Tumor volumes
Tumor

Pre-scan

1 hour post dose

24 hours post dose

FIGURE 3 | CT imaging of tumors in pre and post intratumoral


administration of GA-AuNP in prostate tumor bearing mice. Red Arrow indicates the homogeneity in distribution of GA-AuNP agent in prostate tumor within 1 h and 24 hrs post administration.

1.0

PC3 tumor volume (cm3)

0.8

Treatment vs. Control ** p = 0.005; Day 17 * p < 0.0001; Day 2128 n = 57; means sem Day 8 0.41 mCi Au-198 NP, i.t.

Control group

0.6

Control
Tumor

Treatment

0.4

0.2

FIGURE 4 | Therapeutic efcacy studies of


GA-198 AuNP in prostate tumor-bearing SCID mice. (Reprinted with permission from Ref 37. Copyright 2010 Elsevier)

**
0.0 0

*
30

Treated group

5 10 15 20 25 Days after palpable tumors randomized

Volume 4, January/February 2012

2011 Wiley Periodicals, Inc.

47

Advanced Review

wires.wiley.com/nanomed

were twofold lower (P < 0.005) for the treated animals compared with the control group; 9 days posttreatment with GA-198 AuNP (Day 17). This signicant therapeutic effect was maintained throughout the 30 days long study.37 Three weeks after GA-198 AuNP administration, tumor volumes for the control animals were vefold greater with respect to those for the radiotherapy group (P < 0.0001; 0.86 0.08 vs 0.17 0.02 cm3 ). Several animals in the control group had to be euthanized because of continued weight loss, deteriorating overall health status, and risk of tumor ulceration. By contrast, none of the seven animals in the treatment group reached early termination criteria. They did exhibit a transient weight loss that peaked at 17.6 2.4% on Day 17, but recovered to 10.6 2.9% by Day 31. Liver contained 0.91 0.26%ID, kidney 0.13 0.01%ID, and small intestines 0.09 0.00%ID. Levels of radioactivity noted for blood, heart, lung, spleen, stomach, and pancreas were barely distinguishable from background. Insignicant/no radioactivity in liver, intestine, and various nontarget organs unequivocally established that the therapeutic payload indeed resided within the tumor site throughout the 30-day long treatment regimen. The therapeutic efcacy and concomitant in vivo tolerance of GA-198 AuNP further provides evidence on the potential for clinical translation of this agent. Reduction in tumor volume has a direct bearing on the efcacy of chemotherapy and immunotherapy and has been shown to stimulate a natural immune response. The signicant reduction in tumor volume (>70%), as shown by GA-198 AuNP in prostate tumor-bearing SCID mice indicates the potential for clinical translation of this new nanotherapeutic agent for clinical applications in reducing the size of tumor prior to surgical resection, and perhaps to reduce/eliminate the need for surgical resection in certain circumstances.

efcacy studies section, intratumoral delivery of GA198 AuNP nanoparticles within prostate tumors in mice not only resulted in effective control on the growth of tumors, the analysis of tumor tissue unambiguously demonstrated necrotic features. Therefore, the ultra efcient retention of GA-198 AuNPs within prostate tumors has the potential to effectively destroy tumor cells within localized areas and thus may prevent cell propagation and recruitment of tumor cells (and stem tumor cells) into bone marrowa pathway to decelerate and stop metastases of prostate and other solid tumors.

CONCLUSIONS AND OVERALL ONCOLOGICAL IMPLICATIONS OF GA-198 AuNP AGENT


The development and commercialization of the emitting GA-198 AuNP agent is an innovative approach in cancer therapy because of the inherent high afnity of such agents to tumor vasculature. For future applications of this new therapeutic agent in humans, it is important to recognize that intratumoral injections of GA-198 AuNP will not interfere with systemic administration of other therapeutic agents. Therefore, even the large tumors can be effectively treated with homogeneously dispersed injectable radioactive gold nanoparticles. The results of preclinical studies in prostate tumor-bearing mice, as described above, have shown the clinical capabilities of the readily injectable nanotherapeutic agent, GA-198 AuNP in providing optimum therapeutic payloads at the tumor site with no associated side effects. Detailed toxicology studies of GA-AuNP, in small (mice) and large animals (pigs), have clearly demonstrated the nontoxic characteristics of this new therapeutic agent. The in vivo therapeutic efcacy results obtained to date present realistic prospects toward the development of a new generation of homogeneously dispersed radioactive nanoparticles of 198 Au embedded within the nontoxic Gum Arabic plant source (GA-198 AuNP). The clinical applications and oncological relevance of homogeneously distributed and readily injectable nanoparticulate agents go beyond treating prostate cancers. Indeed, the immediate impact of this intratumorally injectable therapeutic agent will have signicant benets to a large number of inoperable human tumors including: cancers of the head and neck, lip, mouth, buccal mucosa, nasopharyngeal, salivary gland, soft palate, tonsilar fossa/pillar, esophageal, respiratory and digestive tract, genitourinary, bladder, urethral, endometrial, cervix, vaginal, retinal, brain, breast, pancreatic, liver, lung cancers, and soft tissue sarcomas.
Volume 4, January/February 2012

ROLE OF LOCALIZED THERAPY TO DECELERATE METASTASES


It is important to note that the residual tumor tissue samples from the above studies contained 52.3% of the ID of GA-198 AuNP after 28 days. The tumors harvested from the treatment group consisted largely of necrotic tissue, indicating extensive tumor cell kill. The propensity with which GA-198 AuNP ablates prostate tumors (Figure 4) is directly related to the excellent tumor retention characteristics of radioactive gold nanoparticles (Figure 2). The unprecedented payload of inherent therapeutic agent within tumor cells has a direct way of curative prospects in achieving desirable necrotic outcomes. As discussed in the therapeutic
48

2011 Wiley Periodicals, Inc.

WIREs Nanomedicine and Nanobiotechnology

GA-198 AuNP in tumor therapy

A number of clinical trials for establishing safety and tolerance using intratumoral administration of chemotherapeutic agents including 2-hydroxyutamide (Licroca Depot),44 -Gal Glycosphingolipids45 , and Ad5CMV-NIS46 administered intraprostatically followed by radioiodine treatment in patients with locally recurrent adenocarcinoma of the prostate are currently in progress within the National Cancer

Institute and other clinical centres across the world. In this context, GA-198 AuNP agent represents a new nanotechnological approach to oncology as it presents an effective clinical pathway to reduce tumor volume through intratumoral administration of predetermined doses in order to achieve optimum therapeutic payloads at the tumor sites.

ACKNOWLEDGMENTS
The work reported in this review has been supported by grants from the National Institutes of Health (NIH)/National Cancer Institute under the Cancer Nanotechnology Platform program (Grant 5R01CA11941201 and NIH 1R21CA128460-01 and NIH-Small Business Innovation Research Contract 241) and by the University of Missouri-Research Board Program C8761 RB 06-030. Some of the results reported in this review are taken from the graduate research project of Rajesh Kulkarni which would be used toward partial fulllment of his PhD degree program. Continuing collaborative support and encouragement from various faculty colleagues including Jeffrey C Smith, John Lever, Silvia Jurrison, Wynn Volkert, and Timothy Hoffman are gratefully acknowledged.

REFERENCES
1. American Cancer Society. Cancer Facts and Figures 2010. Atlanta, GA: American Cancer Society; 2010. 2. Stephan C, Cammann H, Meyer HA, Lein M, Jung K. PSA and new biomarkers within multivariate models to improve early detection of prostate cancer. Cancer Lett 2007, 249:1829. 3. Wilt TJ, Thompson IM. Clinically localised prostate cancer. Br Med J 2006, 333:11021106. 4. Andriole GL, Grubb RL III, Buys SS, Chia D, Church TR, Fouad MN, Gelmann EP, Kvale PA, Reding DJ, Weissfeld JL, et al. Mortality results from a randomized prostate-cancer screening trial. N Engl J Med 2009, 360:13101319. 5. Schroder FH, Hugosson J, Roobol MJ, Tammela TLJ, Ciatto S, Nelen V, Kwiatkowski M, Lujan M, Lilja H, Zappa M, et al. Screening and prostate-cancer mortality in a randomized European study. N Engl J Med 2009, 360:13201328. 6. Fowler JE Jr, Pandey P, Bigler SA, Yee DT, Kolski JM. Trends in diagnosis of stage T1a-b prostate cancer. J Urol 1997, 158:18491852. 7. Stubbs RS, Wickremesekera SK. Selective internal radiation therapy (SIRT): a new modality for treating patients with colorectal liver metastases. HPB (Oxford) 2004, 6:133139. 8. Wollner I, Knutsen C, Smith P, Prieskorn D, Chrisp C, Andrews J, Juni J, Warber S, Klevering J, Crudup J, et al. Effects of hepatic arterial yttrium 90 glass microspheres in dogs. Cancer 1988, 61:13361344. 9. Hilgard P, Muller S, Hamami M, Sauerwein WS, Haberkorn U, Gerken G, Antoch G. Selective internal radiotherapy (radioembolization) and radiation therapy for HCCcurrent status and perspectives. Z Gastroenterol 2009, 47:3754. 10. Buckle T, Chin PT, van Leeuwen FW. (Non-targeted) radioactive/uorescent nanoparticles and their potential in combined pre- and intraoperative imaging during sentinel lymph node resection. Nanotechnology 2010, 21:482001. 11. Matson JB, Grubbs RH. Synthesis of uorine-18 functionalized nanoparticles for use as in vivo molecular imaging agents. J Am Chem Soc 2008, 130:67316733. 12. Phillips WT, Goins BA, Bao A. Radioactive liposomes. WIREs Nanomed Nanobiotechnol 2009, 1:6983. 13. Wang SX, Bao A, Herrera SJ, Phillips WT, Goins B, Santoyo C, Miller FR, Otto RA. Intraoperative 186Reliposome radionuclide therapy in a head and neck squamous cell carcinoma xenograft positive surgical margin model. Clin Cancer Res 2008, 14:39753983. 14. French JT, Goins B, Saenz M, Li S, Garcia-Rojas X, Phillips WT, Otto RA, Bao A. Interventional therapy of head and neck cancer with lipid nanoparticle-carried rhenium 186 radionuclide. J Vasc Interv Radiol 2010, 21:12711279. 15. Zavaleta CL, Goins BA, Bao A, McManus LM, McMahan CA, Phillips WT. Imaging of 186Re-liposome therapy in ovarian cancer xenograft model of peritoneal carcinomatosis. J Drug Target 2008, 16:626637. 16. Regulla DF, Hieber LB, Seidenbusch M. Physical and biological interface dose effects in tissue due to X-rayinduced release of secondary radiation from metallic gold surfaces. Radiat Res 1998, 150:92100. 17. Khan MK, Minc LD, Nigavekar SS, Kariapper MS, Nair BM, Schipper M, Cook AC, Lesniak WG, Balogh

Volume 4, January/February 2012

2011 Wiley Periodicals, Inc.

49

Advanced Review

wires.wiley.com/nanomed

LP. Fabrication of {198Au0} radioactive composite nanodevices and their use for nanobrachytherapy. Nanomedicine 2008, 4:5769. 18. Chanda N, Shukla R, Zambre A, Mekapothula S, Kulkarni RR, Katti K, Bhattacharyya K, Fent GM, Casteel SW, Boote EJ, et al. An effective strategy for the synthesis of biocompatible gold nanoparticles using cinnamon phytochemicals for phantom CT imaging and photoacoustic detection of cancerous cells. Pharm Res 2011, 28:279291. 19. Chanda N, Kattumuri V, Shukla R, Zambre A, Katti K, Upendran A, Kulkarni RR, Kan P, Fent GM, Casteel SW, et al. Bombesin functionalized gold nanoparticles show in vitro and in vivo cancer receptor specicity. Proc Natl Acad Sci U S A 2010, 107:87608765. 20. Afrasiabi Z, Shukla R, Chanda N, Bhaskaran S, Upendran A, Zambre A, Katti KV, Kannan R. Nanoscale sensor design via in situ labeling of gold nanoparticles onto protein scaffolds. J Nanosci Nanotechnol 2010, 10:719725. 21. Chanda N, Shukla R, Katti KV, Kannan R. Gastrin releasing protein receptor specic gold nanorods: breast and prostate tumor avid nanovectors for molecular imaging. Nano Lett 2009, 9:17981805. 22. Katti K, Chanda N, Shukla R, Zambre A, Suibramanian T, Kulkarni RR, Kannan R, Katti KV. Green nanotechnology from cumin phytochemicals: generation of biocompatible gold nanoparticles. Int J Green Nanotechnol Biomed 2009, 1:B39B52. 23. Katti KK, Kattamuri V, Bhaskaran S, Kannan R, Katti KV. Facile and general method for synthesis of sugarcoated gold nanoparticles. Int J Nanotechnol Biomed 2009, 1:B53B59. 24. Nune SK, Chanda N, Shukla R, Katti K, Kulkarni RR, Thilakavathi S, Mekapothula S, Kannan R, Katti KV. Green nanotechnology from tea: phytochemicals in tea as building blocks for production of biocompatible gold nanoparticles. J Mater Chem 2009, 19:29122920. 25. Kattumuri VCM, Guha S, Kannan R, Katti KV, Ghosh K, Patel RJ. Agarose-stabilized gold nanoparticles for surface-enhanced Raman spectroscopic detection of DNA nucleosides. Appl Phys Lett 2006, 88:153114. 26. Katti KV, Kannan R, Rahing V, Cutler C, Pandrapragada R, Katti KK, Kattumuri V, Robertson JD, Casteel SJ, Jurisson S, et al. Hybrid nanoparticles in molecular imaging and therapy. In: Czechoslovak Journal of PhysicsProceedings of the 15th Radiochemical Conference, vol 56, Supplement D, 2006. 27. Kannan R, Katti KK, Barbour LJ, Pillarsetty N, Barnes CL, Katti KV. Characterization of supramolecular (H2O)18 water morphology and water-methanol (H2O)15(CH3OH)3 clusters in a novel phosphorus functionalized trimeric amino acid host. J Am Chem Soc 2003, 125:69556961. 28. Kannan R, Rahing V, Cutler C, Pandrapragada R, Katti KK, Kattumuri V, Robertson JD, Casteel SJ, Jurisson

S, Smith C, et al. Nanocompatible chemistry toward fabrication of target-specic gold nanoparticles. J Am Chem Soc 2006, 128:1134211343. 29. Katti KV, Kannan R, Chandrasekar M, Srinath A, Kannan R, Katti K, Bhaskaran S, Pandrapragada RK. Optical and photophysical properties of gold nanoparticles. Mol Imaging 2004, 3:278. 30. Fent GM, Casteel SW, Kim DY, Kannan R, Katti K, Chanda N. Biodistribution of maltose and gum arabic hybrid gold nanoparticles after intravenous injection in juvenile swine. Nanomedicine 2009, 5:128135. 31. Shukla R, Nune SK, Chanda N, Katti K, Mekapothula S, Kulkarni RR, Welshons WV, Kannan R, Katti KV. Soybeans as a phytochemical reservoir for the production and stabilization of biocompatible gold nanoparticles. Small 2008, 4:14251436. 32. Katti KV, Kannan R, Katti KK. Stabilized, biocompatible gold nanoparticles and enviro-friendly method for making same. US 2009/0074674 A1 2009. US Patent pending. 33. Kattumuri V, Katti K, Bhaskaran S, Boote EJ, Casteel SW, Fent GM, Robertson DJ, Chandrasekhar M, Kannan R, Katti KV. Gum arabic as a phytochemical construct for the stabilization of gold nanoparticles: in vivo pharmacokinetics and X-ray-contrast-imaging studies. Small 2007, 3:333341. 34. Kannan RK, Katti KV, Katti KK, White HW, Cutler CS. Methods and articles for gold nanoparticle production. US 2007/0051202 A12007. US Patent. 35. Katti KV, Kannan R, Viator J, Chanda N, Shukla R, Zambre A. Pancreatic cancer imaging and therapy using EGFR Peptide conjugated gold nanoparticles. US Patent pending October 2009. 36. Katti KV. Formylation of fucntionalized P-H bonds. A novel approach to the design of synthons for use in biomedicine. Proc Indian Acad Sci Chem Sci 1999, 111:112. 37. Chanda N, Kan P, Watkinson LD, Shukla R, Zambre A, Carmack TL, Engelbrecht H, Lever JR, Katti K, Fent GM, et al. Radioactive gold nanoparticles in cancer therapy: therapeutic efcacy studies of GA198AuNP nanoconstruct in prostate tumor-bearing mice. Nanomedicine 2010, 6:201209. 38. Boote E, Fent G, Kattumuri V, Casteel S, Katti K, Chanda N, Kannan R, Katti K, Churchill R. Gold nanoparticle contrast in a phantom and juvenile swine: models for molecular imaging of human organs using xray computed tomography. Acad Radiol 17:410417. 39. Myers WG, Colmery BH Jr, McLellon WM. Radioactive gold-198 for radiation therapy. Am J Roentgenol Radium Ther Nucl Med 1953, 70:258273. 40. Brahme A, Svensson H. Depth absorbed dose distributions for electrons. Phys Med Biol 1978, 23:788792. 41. Attix FH. Introduction to Radiological Physics and Radiation Dosimetry. New York: John Wiley & Sons; 1986.

50

2011 Wiley Periodicals, Inc.

Volume 4, January/February 2012

WIREs Nanomedicine and Nanobiotechnology

GA-198 AuNP in tumor therapy

42. Wang JZ, Li XA, Yu CX, DiBiase SJ. The low / ratio for prostate cancer: what does the clinical outcome of HDR brachytherapy tell us? Int J Radiat Oncol Biol Phys 2003, 57:11011108. 43. Dale RG. The application of the linear-quadratic doseeffect equation to fractionated and protracted radiotherapy. Br J Radiol 1985, 58:515528. 44. Khan N, Adhami VM, Mukhtar H. Review: green tea polyphenols in chemoprevention of prostate cancer: preclinical and clinical studies. Nutr Cancer 2009, 61:836841.

45. Johnson JJ, Bailey HH, Mukhtar H. Green tea polyphenols for prostate cancer chemoprevention: a translational perspective. Phytomedicine 2010, 17:313. 46. Gene Therapy and Radioactive Iodine in Treating Patients With Locally Recurrent Prostate Cancer That Did Not Respond to External-Beam Radiation Therapy. Available at: http://www.cancer. gov/clinicaltrials/search/view/print?cdrid=618542& protocolsearchid=9255404&version=patient.

Volume 4, January/February 2012

2011 Wiley Periodicals, Inc.

51

You might also like