Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS FOR USING CERIUM OXIDE NANOPARTICLES TO MITIGATE OR PROTECT AGAINST RADIATION INJURY
Document Type and Number:
WIPO Patent Application WO/2013/151698
Kind Code:
A1
Abstract:
Methods of mitigating the effects of radiation exposure or providing a radioprotective effect in a subject are provided herein. The methods include administering a therapeutically effective amount of cerium oxide nanoparticles to the subject to mitigate the effects of radiation exposure or to offer a radioprotective effect to the subject. The cerium oxide nanoparticles are suitably less than 20nm in diameter and have over 50% of the cerium in the 34+ oxidation state.

Inventors:
VUJASKOVIC ZELJKO (US)
JACKSON ISABEL L (US)
SEAL SUDIPTA (US)
DAS SOUMEN (US)
Application Number:
PCT/US2013/030811
Publication Date:
October 10, 2013
Filing Date:
March 13, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV DUKE (US)
UNIV CENTRAL FLORIDA RES FOUND (US)
International Classes:
A61K9/14; A61K33/244
Foreign References:
US20100166821A12010-07-01
US20050066571A12005-03-31
US20100242342A12010-09-30
US20040009349A12004-01-15
Other References:
See also references of EP 2841055A4
Attorney, Agent or Firm:
VANHEYNINGEN, Tambryn, K. et al. (Sceales Starke & Sawall, LLP,100 East Wisconsin Avenue, Suite 110, Milwaukee WI, US)
Download PDF:
Claims:
CLAIMS

claim:

1. A method of mitigating the effects of radiation exposure in a subject comprising

administering a therapeutically effective amount of a composition comprising cerium oxide nanoparticles to the subject.

2 , The method of claim 1, wherein the composition is administered at the same time as the radi ati on exposure .

3. The method of claim 1, wherem the composition is administered after the radiation

exposure of the subject

4. The method of claim 3, wherein the composition is administered between i hour and 24 hours after the radiation exposure.

5.. The method of claim 3, wherein the composition, is administered between .1 day and .1 week after the radiation exposure,

6. The .method of any one of the preceding claims, wherein the composition is administered daily, every other day, twice a week, weekl or bimonthly for a period of at least one month.

7. The method of any one of the preceding claims, wherein the cerium oxide nanoparticles are administered at a dosage of between about 0.00005 rag/kg and. about 0,5 nig/lcg.

8. The method of claim 7, w herein the dosage is between about 0.005 mg/kg and about 0.05 rag/kg.

9. The method of any one of the preceding claims, wherein the composition comprises between about Ι ηΜ and ΙΟμΜ cerium oxide nanoparticles,

10. The method of claim 9, wherein the composition comprises between about I GOnM and ! μ cerium oxide nanoparticles.

1.1.. The method of any one of the preceding claims, wherein the cerium oxide nanoparticles are between 1 nm and 20 nm.

12. The method of any one of the preceding claims, wherein greater than 80% of the cerium in the cerium oxide -nanoparticles is in the 3+ oxidation state.

13, The method of any one of the preceding claims, wherein the composition was made by dissolving ceriu nitrate hexahydrate in water followed by addition of hydrogen peroxide..

Π

14. The method of any one of the preceding claims, wherein the composition further comprises polyethylene glycol,

15. The method of an one of the preceding claims, wherein the composition is administered by injection or via an osmotic pump.

16. The method of claim 15. wherein, the injection route is selected from intraperitoneal.,, parenteral, intravenous, intramuscular, subcutaneous, intrathecal, transcutaneous.

1.7. The method of any one of the preceding claims, wherein mitigation of radiation, damage includes increased survival, reduced morbidity, decreased lung damage or increased lung function.

1 S. The method of claim 17, wherein the reduction in morbidity ma be monitored by

monitoring respirator function of the subject.

1 . The method of any one of the preceding claims, wherein the composition increases survival, of a subject after exposure to radiation by at least 30%.

20. The method of an one of the preceding claims, wherein the composition, increases survival of a subject after exposure to radiation by at. least 40%.

2 L The method of an one of the preceding claims, wherein the composition increases survival of a subject after exposure to radiation by at least 50%.

22. The method of any one of the preceding claims, wherein the composition is administered before radiation exposure as a radioprotective agent.

Description:
METHODS OF USING CERIUM OX IDE NA OPARTICLES TO MITIGATE OR

PROTECT AGAINST RADIATION INJURY

CROSS-REFERENCE TO RELATED APPLICATIONS

This patent application c l aims the benefit of priority of United States Provisional Patent Application No. 61/620,219, filed April 4, 2012,. which is incorporated herein by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

This invention was made with United States government support awarded by the National Institutes of Health grant number IRC! AIOS1290-01. The United States may have certain rights in this invention.

INTRODUCTION

The present invention relates to the field of nanotechnology and in particular to eeria naiioparticies and methods of using the same to mitigate the effect of radiation.

Nanotechnology is rapidly emerging as an important tool in the field of medicine. Among various nanopariiel.es. cerium oxide nanopartic!es have a unique regenerative antioxidant property and can efficiently scavenge reactive oxygen/nitrogen species species (H2O2, (¾ > OH: and 'NO). One of the cerium oxide nanoparticle (Ce . 2; CMP) formulations, CMP- 18, was reported well-tolerated, non-imimmogenic and without overt pathology in small animal in viva models.

Lung injury is the major cause of death following acute radiatio exposure to the thoracic region. Anecdotal evidence described in the literature suggests radiation accident victims receivin a whole body dose in excess of 10 Gy may experience partial hematological recovery with excellent supportive care. However, in these individuals death due to acute respiratory distress occurred three to six months after exposure due to radiation induced lung injury. At the time of death, the lungs were congested and edemic suggesti ve of interstitial pneumonia with diffuse alveolar damage and. in some cases, fibrous lesions were present.. There is no FDA approved medical counter-measure (MC ) to mitigate lung damage and improve survival following acute radiation exposure. SUMMARY

Provided herein are methods of improving survival and .mitigating the damage, in particular lung damage, caused by acute radiation exposure. In particolar, methods of mitigating the effects of radiation exposure in subjects exposed to radiation are provided. The methods include administering a composition comprising cerium oxide nanopartieles (CNPs) to the subject in an amount effective to mitigate the effects of radiation exposure. In particular, the methods mitigate the lung damaging effects of radiation even when the composition is provided after radiation exposure.

BRIEF .DESCRIPTION OF THE DRA WINGS

Figure 1 Is a set of photographs of transmission electron micrographs of the nanoceria panicles when made in water (referred to as CNP-I8 or CNP-WB) and. polyethylene glycol (also referred to as CNP-300 or CNP-ME).

Figure 2 is a graph showing the percent survival of mice provided the indicated treatments two hours after irradiation over time.

Figure 3 is a graph showing the Penh. levels (an indicator of imminent morbidity) over time in mice treated with the indicated treatments two hours after irradiation.

Figure 4 is a graph showing the change in respiratory .rates over time in mice treated with the indicated treatments two hours after irradiation.

Figure 5 is a set of photographs showing the histopatlio! ogy of lung tissue in non- irradiated controls, irradiated controls and in irradiated and treated animals (lOOnM CNP-18).

Figure 6 A. is a photograph of a transmission electron micrograph showing the

nanopartieles.

Figure 6B is a photograph of a high, magnification image of a nanopartiele showing the structure.

Figure 6 ' C is a graph showing the size range of the nanopartieles used.

Figure 6I> is a graph showing the results of a ΜΤΓ cell viability assay after the cells were incubated with the indicated concentrations of CNP-18 for 24 hours, No cytotoxicity was observed. DETAILED DESCRIPTION

Since September i 1. 2001 , the U.S. government has placed increasing emphasis on developing arid stockpiling medical eountermeasures (MC s) to treat acute radiation syndrome (A S) in the event of a nuclear or radiological attack on U.S. soil. At this time there is no approved MCM for treading- the delayed effects of acute .radiation exposure, specifically acute and delayed lung injury (DEA E-lung). One of the primary concerns associated with thoracic irradiation is an acute hut delayed onset of radiation, pneumonitis with an incidence that rise very steeply at relatively low radiation doses. Previous studies indicate that the initial oxidative stress produced during the radiation event is amplified through continuous production of reactive oxygen/nitrogen species (ROS/RNS) leading to an imbalance between oxygen derived free radicals and antioxidant capacity. The continuous generation of R.OS plays a central role in the underlying processes of radiation injury ihrough.activatio.n/de-activation of key redox-regulated signaling pathways leading to acute pneumonitis as an early event and or chronic fibrosis as a late event. Antioxidant therapies may be useful to reduce inflammation and fibrosis in rodent models of radiation-induced lung injur}'. Therefore, we hypothesized tha the potent antioxidant properties of specially designed nanoceria may improve survival after radiation injury by restoring the redox balance of the cell.

In the Examples, we demonstrate mitigation of latig injury and improvement in survival after treatment with LOO.rs CKP- 18 starting 2 hours after acute radiation exposure to the thorax. Thus nanoceria are capable of improving survival and reducing lung damage when provided to subjects after irradiation. Thus, methods of mitigating the effects of radiation exposure in subjects exposed to radiation, are provided. The methods include administering a composition comprising CNPs to the subject in an amount effective to mitigate the effects of radiation exposure. In particular, the methods mitigate the i ng damaging effects of radiation even when the composition is provided after radiation exposure. The CNPs were made and characterized using methods described more folly in the Examples.

The radiation exposure level used in the Examples was a 90% lethal dose over 160 days. Tower or sub-lethal doses of radiation also cause similar effects on subjects and may lead to decreased, lung performance or increased lung damage after exposure which may be mitigated by treatment with the cerium oxide nanoparticl.es as. described herein. Mitigating the effects of radiation includes, but is not limited to, improving survival after radiation exposure, reducing morbidity, reducing lung damage, improving respiratory or limg functions, which may be measured by monitoring the respiratory function, of the subject, for example, b monitoring the breathing frequency, enhanced pause (Penh), tidal volume (TV), peak respiratory flow (PIP), peak expiratory flow (PEP), and inflammation in the lungs. Treating a subject as used herein refers to any type of treatment that imparts a benefit to the subject, including improvement in the condition of the subject (e.g., in one or mor symptoms), delay in the progression of the condition, delay the onset of symptoms or slowin the progression of symptoms, etc.

Although some compositions have been demonstrated to offer radioprotection if given to the subject prior to radia tion exposure, compositions capable of mitigating the effects of radiation exposure when administered after the exposure are not available. The compositions provided herein are capable of mitigating the effects of radiation exposure when administered at the same time as or after radiation exposure. This surprisin effect is demonstrated in the Examples in a mouse model. The composition comprising cerium oxide nanoparticles may be administered 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, 24 hours, 2 days, 3 days, 4 days, 5 days, 6 days or even i or 2 weeks after exposure to the radiation. The composition may be administered multiple times at intervals of I day, 2 days. 3 days, bi weekly, weekly, bimonthly or monthly.

An effective amount or a iherapeutieally effective amount as used herein means the amount of the composition, thai, when administered to a subject for treating radiation exposure is sufficient to effect a treatment (as defined above). The therapeutically effective amount will vary depending on the formation of the composition, formulation or combination, the severity and timing of exposure to radiation and the age, weight, physical condition and responsiveness of the subject to be treated. Subjects include mammals, such as humans, mice, rats, dogs, cats, cows, pigs and non-mammals such as chickens, turkeys or other animals.

In the Examples, the cerium oxide nanoparticles were demonstrated to protect 90% of the mice when given two times a week fo four weeks at 7pg kg commencing two hours after exposure to a 90% lethal dose of radiation. Thus in the Examples, the nanoeeria was capable of increasing survi val by 80%. Suitably, the methods described herein are capable of increasing the survi val chance of a subject after exposure to radiation by at. least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or even. more. The compositions comprising cerium oxide nanoparticies may be made by methods known to those of sk ll in the ar See, for example, U.S. Patent Nos. 7,347,987 ant! 7,727,559; U.S. Patent Application No. 12/874,398, entitled Polycrystalline Natiopartieles of Cerium Oxide and Associated Methods; and arakoti et al. Naiioceria as Antioxidant; Synthesis and

Biomedical Applications JOM (2008) 60:33-37 for methods of preparing cerium oxide nanoparticies, each of which are incorporated herein by reference in their entireties. Suitably the cerium oxide nanoparticies have particle diameters that are between. nrn and 20nm, suitably between, him and lOnm, suitably between lnm and 3nm. Transmission electron micrographs of the nanopartic es are shown in Figures 1 and 6A and B. Figure 6C shows the size distribution of the nanoparticies.

Suitably the cerium, oxide uan.opartkl.es are prepared, such that mor than 50% of the cerium in the nanoparticies is in the 3 oxidation, state. Suitably, at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85% 90% or even more of the nanoparticies in the composition arc in the 3+ oxidation state in. the composition. The nanoparticies may be prepared in water or short polar solvents compatible with biological systems. The cerium oxide nanoparticies may complexed or coated with a biologically compatible polymer or polysaccharide such as polyethylene glycol or dextran. in the Examples, the nanoparticies were prepared in. water or as a microemutsion. The nanoparticies prepared in water were shown to be more effective than those prepared, as a microemulsion in our preliminary experiments shown in the Examples. The compositions may also comprise other pharmaceuticals useful in mitigating the effects of radiation exposure such as other pharmaceuticals such as antioxidants or may aiso include pharmaceutically acceptable additives or exeipients.

The compositions described herein may be administered by any means known to those skil led in the art, including, but not limited to, oral, topical, intranasal, intraperitoneal, parenteral, intravenous, intramuscular, subcutaneous, intrathecal., transcutaneous, nasopharyngeal, or trausmucosaL In the Examples, the CNiP were provided via intraperitoneal injection. Thus the compositions may be formulated, as an mgestable, injectable, or topical formulation or for delivery via an osmotic pump. The compositions could also be provided as an. aerosol formulation for direct delivery to the lung vi inhalation. The compositions may also be delivered within a liposomal or time-release vehicle. Administration of the compositions to a subject in accordance with the invention appears to exhibit beneficial effects in a dose-dependent maimer. Thus, within broad limits, administmtioa of larger quantities of the- compounds is expected to achieve increased beneficial biological effects than, administration of a smaller amount Moreover, efficacy is also contemplated at dosages below dm level at which toxicity is seen.

it will be appreciated that the specific dosage administered ' in any given, case- will be adjusted in accordance with the compositions being administered, the formulation of the composition, the disease to be treated or inhibited, the condition of the subject, and other relevant, medical factors that may modify the activity of the composition or the response of the subject, as is well known by those skilled in the art. For example, the specific dose for a particular subject depends on age, body weight, general state of health, diet, the timing and mode of

administration, the rate of excretion, medicaments used in combination, and the severity of the particular disorder to which the therapy is applied. Dosages for a given patient can be

determined using conventional considerations, e.g., by customary comparison of the differential activities of the compositions and of a known agent, such as by means of an appropriate conventional pharmacological or prophylactic protocol.

The maximal, dosage for a subject is the highest, dosage that, does not cause undesirable or intolerable side effects. As shown in Figure 6D, no toxicity is detected in vitro at any of the doses tested up to ΙΟμΜ. In the Examples, a l OOnM dose o CNP-18 was effective to

significantly reduce both morbidity and mortality after a lethal dose of radiation was

administered. The number of variables in regard to an individual prophylactic or treatment regimen is large, and a considerable range of doses is expected. The route of administration will also impact the dosage requirements. It is anticipated that dosages of the composition will reduce morbidity and/or mortality by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% compared to morbidit or mortality if the radiation exposure is left, untreated. It is specifically contemplated thai pharmaceutical, preparations and compositions ma palliate or alleviate symptoms of radiation exposure without providing a cure, or, in some embodiments, ma be used to reverse the effects of radiation exposure.

The cerium oxide nanopartieles may be administered at a dosage between about 0.00005 mg/kg and 0.5 rag/kg. Suitably, the dosage is between about 0.005 mg/kg and 0,05 mg. kg. In the examples, the dosages used were between about 0.00007 mg/kg and 0.007 mg/kg. The composition may be formulated such that the cerium oxide nao.opartic-S.es are between lOnM and 10 μΜ, Suitably, the cerium oxide nanoparticles are present at between 1 0 «M and Ι μΜ in the composition.

The composit ons may be administered as. a single dose or as multiple doses. Suitable effective total dosage amounts for administering the compositions may be determined by those of skill in the art, but typically range from about 50 nanograms to about 1 milligram per kilogram of body weight, although they are typically about 1-100 micrograms per kilogram of bod weight. The dosage used in the Examples and shown to be effective was about 56p.g/kg given in 8 doses of 7 ig. kg in each dose. Large doses may be required for therapeutic effect and toxicity of the composition is low. in some embodiments, the effective dosage amount ranges from about I to about 100 micrograms per kilogram of body weight weekly. In another embodiment, the effective dosage amount ranges from about 5 to about 5,000 microgram per kilogram of body weight over the course of treatment. Notably the dose of 7ug/kg in 8 total doses used in the Examples was effective to offer substantial protection against a lethal radiation dose when administration, began after radia tion exposure. Those of skill in the art will appreciate that Sower doses may be effective if the radiation exposure is sub-lethal or if the compositions described herein are provided, in advance of radiation exposure as a radioprotective agent and not solely to mitigate previous radiation exposure. The effective dosage amounts described herein refer to total amounts administered, that is, if more than one dosage is administered in a period of time, the effective dosage amounts correspond to the total amount administered.

If the composition is administered as more than one dose or as divided doses, the dosage rate and amount may be modified accordingly. For example, the composition may be

administered two or more times separated by 4 hours, 6 hours, 8 hours, 12 hours, a day, two days, three days, four days, one week, two weeks, or by three or more weeks. In the Examples, the compositions were given two times per week for a period of four weeks. The compositions may be provided for longer tha four weeks, for example for two, three, four months or more.

The cerium oxide nanoparticles may be co-administered with other pharmaceuticals or compositions either in a unitary composition or as two separate compositions, Co-administration of cerium oxide nanoparticles with other compositions may be admini stered in any order, at the same time or as part of a unitary composition. The two may be administered such that one is administered before the other with a difference in administration time of 1. hour, 2 hours, 4 hours, 8 hours, 12 hours, 1.6 hours, 20 hours, I. day, 2 days, 4 days, 7 days, 2 weeks, 4 weeks or more. The following examples are meant only to be illustrative and are not meant as limitations on the scope of the invention or of the appended claims. All references cited herein are hereby incorporated by reference in their entireties, EXAMPLES

Radiation-induced lung injury has traditionally been divided into two phases: the early inflammatory phase and the late flbro-pr iiferative phase. Histologically, the pneumonitis reaction is characterized by edema, atypia and desquamation of alveolar epithelial cells, intimal proliferation and medial thickening, mononuclear cell infiltrates, giant cell, formation, and inflammatory exudates with an absence of polymoi honuclear leukocytes. Hyaline membranes are also seen. Progressive interstitial fibrosis with fibro-obliteration of capillaries, thickened alveolar walls, and lipid-laden foamy macrophages are. seen during the late phase.

Under normal circumstances the cell is able to maintain an adequate homeostasis between the formation of ROS and its removal through particular enzymatic pathways or via antioxidants. However, following radiation exposure to the lung, there is an immediate and continuous increase in chronic oxidative stress that leads to vascular dysfunction., tissue hypoxia., and.

inflammation, and fibro-proliferation. Therefore, we hypothesized suppressing chronic oxidative stress may remove the primary stimulant facilitating the pathogenesis of radiation-induced injury.

We previously characterized the pulmonary dose-response and pathology of injury among six of the most common murine strains utilized as models of radiation-induced Sung injury. See Jackson, Vujaskovie, and Down, Revisiting strain-related differences in radiation sensitivity of the mouse lung: recognizing and avoiding the confounding effects of pleural effusions. R di t R s 2010, 173, { 1), 10-20 and Jackson, Vujaskovie, and Down, A Further Comparison of Pathologies after Thoracic Irradiation among Different Mouse Strains: Finding the Best Preclinical Model for Evaluating Therapies Directed Against Radiation-Induced Lung Damage. Radiat Res 2011, both of which are incorporated herein by reference in their entireties, The dose-response relationship for lethal pneumonitis in CBA/i mice has been characterized using a single radiation dose of 1 1.75- 14.75 Gy to the whole thorax. Mortality in the CBA/J strain strongly correlates with increased lung weight, an indicator of edema and congestion, and elevated breathing rates, a marker for functional damage. We evaluated whether CNP could mitigate limg injury in female CBA/J mice when delivered starting two hours afte -whole thorax irradiation with a single dose of 15 Gy

(LD90/160) of 320 kV x-rays (Dose rate: 0.69 Gy/rnin, HVL~2.<K) mm A!, 75 cm SSD). Two different formulations of cerium oxide nanopariicles, CNP- 18 (Cerium oxide nanoparticles prepared in water; CNP-DW) and CNP-ME (a microemuision of CNP; CNP-300) were analyzed. See, for example, U.S. Patent Nos, 7,347,987 and 7,727,559 or U.S. Patent Application No. 12/874398 and Karakoii et al. Nanoceria as Antioxidant: Synthesis and Biomedical Applications JOM (2008) 60:33-37 for methods of preparing cerium oxide nanoparticles, each of which is incorporated herein by reference in its entirety.

Briefly, cerium oxide nanoparticles. were prepared using a wet chemical synthesis. A stoichiometric amount of cerium nitrate hexahydrate (99.999%) was dissolved in deionized water (18,2 ΜΩ) by constant slow stirring for 10 minutes. The solution is then preferably filtered using a pre-rkised 20nra filter paper (ANODISC -47, 0.02μτη) to remove undissolved and unwanted impurities. The filtered solution is then transferred to a standard -flask and stirred slowly and the cerium (il l) ions are oxidized using a stoichiometric amount of hydrogen

peroxide as an oxidizer, added dro wise while continuing to stir, to form cerium oxide

nanoparticles. The amount of hydrogen peroxide influences the time required for the seff- assemiy of nanoparticles during the aging of the sol ut ion. In a typical procedure, the mo lar ratio of Ce to hydrogen peroxide varied from 1 :3 to 1 :20. The color and the pH of the solution were monitored continuously during the addition of hydrogen peroxide. The pH of the solution drops below 3.0 by addition of the hydrogen peroxide. The solution is stirred until the typical yellow color of the solution gradually changes to a. dark yellow. The stirring is stopped at this point and the solution is transferred to a bottle. Slow evolution of oxygen in the form of bubbles should be observed, during aging of the solution. The solution is aged for a minimum of 15 days and it changes color from dark yellow to dark orange and. then to Sight yellow (or colorless). During the aging process the oxidation state of the nanoceria undergoes a dynamic reversibilit nd changes from predominately€e : to Ce' "" , The aging allows time dependent oriented

agglomeration, of nanoparticles in solution with low cerium to hydrogen peroxide ratios. The pH is monitored during the process and increases gradually and is maintained, at 1 ,0-4.5 to keep the nanoparticles in suspension. During aging, the natioparticle suspension is stored under ambient laboratory conditions in light, at a temperature between 20-30 ' C and humidity less than 50%. The C Ps can be stored in the same environment after the aging process and may be dialyzed against deionized water prior to use to eliminate the acid or other impurities. The CNPs can be combined with PEG as described in Karakoti ei ai 2009, J Am Client Soc 131 : 14144-5, which is incorporated herein by reference m its entirety,

Mice were randomized into groups to receive saline vehicle or one of two formications of nanoceria, CNP- 18 or CNP-ME. Nanoparticles were given b intraperitoneal injection two times per week for four weeks at a high dose of 100 rM (0.00? mg kg) or a low dose of 1.0 nM

(0,00007 mg kg). Animals were followed for up to twenty-two weeks for survival (primary parameter). Respiratory taction was assessed prior to radiation and every 2 weeks post- radiation, using unrestrained whole body piethysmograph. At the time of necropsy, lung tissue was harvested, weighed, and formalin fixed for paraffin embedding. Tissue sections were stained with hematoxylin and eosin to assess histopathologic damage, and perivascular and alveolar inflammation.

A. .significant percentage (90%, 9/10) of irradiated animals survived in the 100 nM CNP- 18(WB) nanoparticle treated group compared with 10% ( 1/ 10) of animals in the RT + saline and 30% (3/3 ) in RT + 10 nM nanoceria-C P-18 (WB) treated groups (Figure 2), An improvement in multiple respiratory function parameters, including breathing frequency {Figure 4), enhanced pause (Penh) (Figure 3), tidal, volume (TV), peak inspiratory How (Pl ' F), peak expiratory flow (PEP) was observed. These results demonstrated a significant mitigating effect on functional injury with 100 nM CNP-18 treatment Lung histology revealed a significant decrease (p<

0.000! ) in the structural damage in mice receiving 100 «M CNP-.l 8 naaoparticles (Figure 5). In addition, significant reduction of in. inflammation (p-0.0075) was observed in the 100 nM CNP- 1 nanoparticle treated group compared with radiation only control animals (Figure 5).