Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CAGED COMPOUND DELIVERY AND RELATED COMPOSITIONS, METHODS AND SYSTEMS
Document Type and Number:
WIPO Patent Application WO/2013/086457
Kind Code:
A1
Abstract:
Methods are described and related devices, compositions, and systems, in which a caged compound is administered to a biological environment, the caged compound being caged with a long wavelength absorber, the long wavelength being a wavelength greater than or equal to 750 nm; and irradiating the biological environment to excite the long wavelength absorber with light at a wavelength in a range from 900-1100 nm, thus decaging the compound.

Inventors:
DOUGHERTY DENNIS A (US)
GRUBBS ROBERT H (US)
HUMAYUN MARK (US)
REGAN CLINTON J (US)
EMAMI-NEYESTANAK AZITA (US)
Application Number:
PCT/US2012/068626
Publication Date:
June 13, 2013
Filing Date:
December 07, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CALIFORNIA INST OF TECHN (US)
UNIV SOUTHERN CALIFORNIA (US)
International Classes:
A61K49/06; A61K47/30; A61K47/48; A61K49/08
Domestic Patent References:
WO2011057028A12011-05-12
WO1999010046A11999-03-04
WO2010039886A12010-04-08
Foreign References:
US6274086B12001-08-14
US20100035290A12010-02-11
Attorney, Agent or Firm:
BASS, Jonathan Y. et al. (155 N. Lake Ave.Suite 70, Pasadena CA, US)
Download PDF:
Claims:
CLAIMS

1. A method comprising administering a caged compound to a biological environment, the caged compound being caged with a long wavelength absorber, the long wavelengt being a wavelength greater than or equal to 750 am; and irradiating the biological environment to excite the long wavelength absorber with light at a wavelength in a range from 900-1100 nm, thus decaging the compound.

2. The method of claim 1, wherein the caged compound is comprised within a photoacid compound, the photoacid compound comprising a light absorbing moiety attaching the caged compound as a pay load moiety through a linker moiety, wherem the linker moiety is an organic moiety comprising a geminal diaikyi moiety linked to an ester group having a carbonyl oxygen, the ester group attaching the payload moiety; the light absorbing moiety is an organic moiety attaching the linker moiety in ortho position to a hydroxy 1 group; and the linker is configured to present the carbonyl oxygen for reaction with the hydroxyl group.

3. The method of claim 2, wherem the light-absorbing moiety is a substituted or unsubstituted polyeyclic aromatic hydrocarbon, a susbstituted or unsubstituted closed chain cyanine or a susbstituted or unsubstituted hemicyanine.

4. The method of claim 2 or 3 , wherein the light absorbing moiety is able to absorb light at a wavelength of from about 900 nm to about 1 100 nm.

5. The method of any one of claims 2 to 4, wherein the linker moiety is a monoalkoxy or a dialkoxy moiety in which an oxy group forms part of the ester group having the carbonyl oxygen and attaching the payload moiety, . The method of any one of claims 2 to 5, wherein the payload moiety is a substituted or unsubstituted alkyl, aryi, heieroaryl, ammoalkyl, or oxyalkyl moiety.

7. The method of any one of claims 1 to 6, in which the caged compound is comprised within a photoacid compound according to formula (I):

wherein:

R4 is a light-absorbing moiety presenting a hydroxyl group for interaction with the carbonyl oxygen of R3(CQ)Q, wherein the light- absorbing moiety is a substituted or unsubstituted polycyclic aromatic hydrocarbon, a substituted or unsubstituted closed chain cyanine, or a substituted or unsubstituted he icyanme, and wherein the hydroxy! group is covaiently bonded to the polycyclic aromatic hydrocarbon or closed chain cyanine or hemicyanme and is ortho to X1;

R3 is a payload moiety, wherein the payload molecule is a substituted or unsubstituted alkyl, aryi, heteroaryl, ammoalkyl, or oxyalkyl moiety;

Xf is independently selected from the group consisting of C and O; m is between 0 and 3; and

R1 and " are independently Ci-Ce alkyl groups, cycloalkyl, or substituted or unsubstituted hydrocarbylene groups wherein when R1 and R6 are substituted or unsubstituted hydrocarbyl groups they are linked together to form a cyclic moiety.

8. The method of claim 7 wherein Rl and R2 are methyl groups,

9. The method of anyone of any one claims 1 to 8, wherein the biological environment is a body pari of an individual and the administering is performed by systemic or topic administration of the caged compound.

10. The method of anyone of claims 1 to 9, wherein the biological environment exhibit inflammation or an inflammatory response.

11. The method of anyone of claims 1 to 10. wherein the caged compound is a drug or imaging agent.

12. The method of anyone of claims 1 to 1 1 , wherein the irradiating is performed by a wearable infrared emitting device suitable to worn around a body part of an individual

13. A system for delivering caged compound to a biological environment the system comprising two or more of: one or more caged compound, the caged compound being caged with a long wavelength absorber, the long wavelength being a wavelength greater than or equal to 750 nin; and a light emitting device adapted to irradiate light and excite the long wavelength absorber at a wavelength of from about 900rsm to about 1 l OOnm for simultaneous combined or sequential use in the method of any one of claims 1 to 12.

14. The system of claim 13, wherein the long wavelength absorber and the compound are combined in a photoacid having formula (I).

15. The system of claim 13 or 14, wherein the light emitting device is a wearable infrared emitting device suitable to worn around a body part of an individual

16. A wearable infrared-emitting device comprising; a support adapted to be worn around a body part of an individual; an array of infrared transmitters and infrared detectors arranged on the support, the infrared transmitters con figured to emit infrared light at a wavelengt in a 900-1 ,100 nanometer range, the infrared detectors configured to detect the infrared light transmitted through the body pari of the individual; and a control circuit to control duty cycle and time-gating of the array of the infrared transmitters, wherein the duty cycle and the time-gating are configured to trigger and release caged compound in the brain tissue.

17. The device of claim 16, wherein the infrared transmitters and infrared detectors are located within a plurali ty of modules, each module comprising an infrared transmitter and an infrared detector.

18. The device of claim 16, wherein each infrared transmitter comprises an infrared light emitting device (LED) and an associated variable resistor or transistor.

19. The device of claim 18, wherein each transistor is controlled by a dedicated control signal.

20. The device of any one of claims 16 to 19, wherein at least some infrared transmitters of the array of infrared transmitters are coupled with a respective lens to allow transmission of the infrared light through the lens.

21. The device of claim 20, wherein the lens is a bi-concave lens.

22. The device of any one of claims 16 to 21, where the garment is worn around the head or hands.

23. The device of claim 16 to 22, wherein the caged compound is a caged drug.

24. The device of claim 16 to 23, wherein the control circuit comprises a gating signal and a clock circuit.

Description:
CROSS REFERENCE TO RELATED APPLICATIONS

[0001 ] The present application claims priority to U.S. Provisional Application No. 61/568,046, entitled "Long Wavelength activation of bioactive compound" filed on December 7, 201 1 with docket number QT-6041-P, and is related to PCT Application No. entitled "Photoacid compounds, and related compositions methods and systems" filed on December 7, 2012, with docket number P1 30-PCT, each of which is incorporated herein by reference in its entirety.

FIELD

[0002] The present disclosure relates to compound delivery in a biological environment and in particular to caged compound delivery, and related compositions, methods and systems

BACKGROUND

[0003] Molecular delivery has been a chal lenge in the medical field as well as in the field of biological molecule analysis, in particular when aimed at obtaining controlled delivery of analytes of interest to specific environments. Whether for medical applications or for fundamental biology studies, several methods are commonly used for the delivery of various classes of biom.ateri.als and biomolecules.

[0004] Controlled delivery of targets to specific environments, e.g. specific cell types and or tissues of individuals in vitro and/or in vivo is currently still challenging, especially when directed at providing controlled release of the target in a controllable conformation, typical ly associated to a biological activity.

SUMMARY

[0005] Described herein are methods and systems for controlled delivery of compounds in a biological environment. In particular, in embodiments herein described controlled delivery of a compound of interest is performed by providing a compound of interest as a caged compound to a biological environment and then uncaging the caged compound through light irradiation at a suitable wavelength.

[0006] According to a first aspect, an embodiment describes the administering of a caged compound to a biological environment, the caged compound being caged with a long wavelength absorber, the long wavelength being a wavelength greater than or equal to about 750 nm; and irradiating the biological environment to excite the long wavelength absorber with light at a wavelength in a range from about 900 to about 1 1 OOnm, thus decaging the compound.

[0007] According to a second aspect, an embodiment describes of a system for delivering caged compound to a biological environment the system comprising, a long wavelength absorber, a compound of interest, a light emitting device adapted to irradiate light and excite the long wavelength absorber at a wavelength of from about 900nm to about 1 1 OOnm for simultaneous combined or sequential use in a method of the disclosure.

[0008] According to a third aspect, an embodiment describes a wearable infrared- emitting device comprising: a support adapted to be worn, in use, on a body part of an individual; an array of infrared transmitters and infrared detectors arranged on the support, the infrared transmitters configured to emit infrared light at a wavelength in a 900-1 ,100 nanometer range, the infrared detectors configured to detect the infrared light transmitted through the individual's body part; and a control circuit to control duty cycle and time-gating of the array of the infrared transmitters, wherein the duty cycle and the time-gating are configured to trigger and release caged compound in the brain tissue.

[0009] Methods, systems and related devices herein described allow in some embodiments delivery of chemical conrpounds to biological environments in a conlrolled fashion possibly directed to targeted activation of compounds of interest following delivery.

[0010] Caged compounds and long wavelength absorbers and related methods and systems herein described can be used in connection with applications wherein control led delivery of a compound of interest is desired. Exemplary applications includes application in medical, biological, chemical and pharmacological fields, wherein a controlled release of a compound of interested in a selected biological environment and/or verification and/or validation of a desired chemical biologica and/or pharmacologic activity (e.g. pharmacokinetics, pharmacodynamics, and tocieokinetics) for treatment, imaging or experimental purpose is desired.

[0011] The details of one or more embodiments of the disclosure are set forth in the accompanying drawings and the description below. Other features and objects wil l be apparent from the description and drawings, and from the claims.

BRIEF DESCRIPTION OF DRAWINGS

[0012] The accompanying drawings, which are incorporated into and constitute a part of this specification, illustrate one or more embodiments of the present disclosure and, together with the description of example embodiments, serve to explain the principles an d impl ementation s of the disclosure.

[0013] FIG._i shows a schematic illustration exemplifying a reaction scheme for the deliver}' of a caged drug compound linked to a Near Infrared Range (NIK) absorber.

[0014] FIG. 2 shows a schematic illustration of three pathways (alternative, lectin, and classical pathway) associated with Age-related Macular Degeneration which can be targeted with a controlled deliver}' of a caged compound according to embodiments herein described.

[001 S] FIG. 3 shows a schematic illustration of a cascade of events upon primary brain injury which can be targeted with a con trolled delivery of a caged compound according to embodiments herein described.

[0016] FIG. 4 show a schematic illustration of a diagram of penetration of infrared wavelength into brain tissue (FIG..4A) a CT scan of a patient with traumatic brain injury (FIG. 4.B) as well as a design to intravenous administer a caged-drug complex that is photonic-ally decaged in areas of brain injury by a wearable time-gated LED emitter array contoured to fit the skull (FJG.JC).

[0017] FIG. uuu 5 depicts approaches directed to identify caged compounds and related methods and systems for targeted delivery according to embodiments herein described. In particular, FIG. 5A shows a block diagram related to a method to identify and design a caged compound to be delivered in the brain using traumatic brain injuries (TBI) models according to embodiments of methods and systems herein described. FIG, SB shows a schematic illustration of a method for therapy development from human cadaveric work to in vivo testing performed using caged compounds herein described, according to embodiments herein described. The illustration of FIG. SB is provided by the University of Pittsburgh.

[0018] FIG, A shows a plurality/array of infrared (IR) transmitters and IR detectors arranged as a grid on a patient's head. FIG. B shows a possible modular configuration of the IR transmitters and detectors of FIG. 6A, each module corresponding an IR transmitter and an IR detector.

[0019] FIGS. 7A amd 7B exemplify circuits designed to emit near infrared wavelength, the circuits comprising infrared LEDs and potentiometers. The arrangement in FIG. 7B al lows for adjustment of LED parameters (e.g., power output).

[0020] FIG. 8 shows an exemplary circuital arrangement for switching and gating of infrared LEDs as transmitters

[0021] FIG. uu 9 shows an embodiment where both LEDs and lenses are provided, and in particular how a lens placed in front of an LED alters the behavior of transmitted light.

[0022] FIG. . 10 exemplifies the electrical configuration of an infrared detector capable of collecting backscatter from LED emitters.

[0023] FIG. 11 shows a simulated EM field intensity from an array of LED (half circle outline shows skull) resulting from preliminary 2D EM simulations

[0024] FIG. 12 shows illumination intensity across cross-section of skull resulting from preliminary 2D EM simulations

DETAILED DESCRIPTION

[0025] Method and systems and related compositions and devices are described herein that allow delivery of a compound in a biological environment.

[0026] The term "compound" as used herein indicates chemical substance consisting of two or more different chemical elements linked by chemical bonds, that can be separated into simpler substances by chemical reactions. In compounds in the sense of the present disclosure the chemical bonds can be covalent bonds, ionic bonds, dipole-dipole interactions, London dispersion forces and hydrogen bonds. The compounds herein described are typically characterized by the ability to perform biological and/or chemical reactions of interest in a biological environment.

[0027] The wording "biological environment" as used herein indicates an environment comprising a cell and/or a cellular component. Biological environments in the sense of the present disclosure comprise in vitro biological environments such as cells derived from multicellular organisms (e.g. cell culture or tissue culture), subcellular components and/or organelles (e.g. mitochondria or ribosomes), cellular or subcellular extracts (e.g. wheat gerrn or reticulocyte extracts), or purified molecules in the test tube (e.g. proteins, DNA, or RNA, individually or in combination); ex vivo biological environments such as tissues or portion thereof, organs or portion thereof, biopsies, or cadavers or portion thereof; and "in vivo " biological environments such as living organisms, animals, or human patients. Exemplar}? biological environments in the sense of the present disclosure further comprise an aqueous solution of cellular proteins and organelles from a lysed cell, as well as a cell populatio immobilized on a solid surface or suspended in a culture a mass or population of cells forming a tissue or a portion thereof, an abnormal cellular growth, a tumor, a tissue and/or a body part or portion thereof and additional environment identifiable by a skilled person upon reading of the present disclosure.

[0028] In particular, in embodiments herein described a compound of interest is delivered to a biological environment as a caged compound. The term "cage" as used herein relates to the interaction between a first chemical moiety and a second chemical moiety that minimizes the participation in physical chemical or biological reactions of the second chemical moiety. In particular, caging can occur for example through binding of the first chemical moiety to the second chemical moiety through a chemical bond or physical containment of a second chemical moiety by a first chemical moiety (such as a carbon nanotube). Accordingly, a "caged compound" as used herein, relates to a chemical moiety in a cage interaction with another chemical moiety, and can be reversed by means suitable to modify the cage interaction and decage the second moiety. Exemplary means comprise a drug compound caged via a covaient bond to a separate molecule that is inert; when decaged, the drug compound becomes active and can participate in drug-related chemical reactions such as substrate binding or neuromoduiatioii. The term "decage" or "uncage" but herein is defined as a modification of a caging interaction between a first and second chemical moiety to the chemical moiety, liberating the second molecule to participate in chemical or biological reactions. In particular, decaging can be performed through cleavage of a chemical bond, a stereochemical change of the caged compound or physical release of the caged compound. Decaging of a compound can be detected by identifying the decaged activity of the uncaged compound or by LC-MS spectroscopy to identify either the caged compound, the first moiety, or the second moiety (see Example 2).

[0029] In embodiments herein described the compound to be delivered is caged by a long wavelength absorber, which is a chemical moiety able to switch from a ground state to an excited slate upon light irradiation at a long wavelength, (a wavelength greater than or equal to about 750 nm). The term "excited state" as used herein refers to an electronic state of a moiety in which the molecule has absorbed light energy and been promoted to a higher energy state. This process is referred to as "excitation." The term "ground state" refers to the electronic state of a moiety in which the electrons are in their lowest energy molecular orbitals. In photoacid compounds, the excitation can be accomplished, for example, by irradiating a photoacid molecule with light of energy equal to the difference in energy between the ground state and the excited state. The energy of the light is determined by the wavelength of the light according the relationship E = hc/λ, where E is the energy of the photon, h is Plank's constant, and λ is the wavelength of the light. In particular, in some embodiments, the light used to effect the excitation is infrared or near infrared light,

[0030] The term "infrared light" as used herein refers to li ght in the infrared region of the electromagnetic spectrum from approximately 0.75 iim to approximately 1000 μηι. The term "near infrared" refers to a region of the infrared spectrum from approximately 0,75 μη (750 nm) to approximately 1 .4 μηι (1400 nm).

[0031 ] In embodiments herein described the long wavelength absorber has a ground state at a condition compatible with viability of the cell and/or ceil component i the biological environment of interest and is capable of assuming an excited state upon absorbance of the light at the long wavelength in the biological environment. In embodiments herein described, promotion from the ground state to a suitable excited state of the long wavelength absorber results in decaging of the compound of interest from the wavelength absorber, following a reaction also identified as photoionic decaging.

[0032] In some embodiments, a preparation of caged compounds herein described is designed to have a photoacid compound comprising a light absorbing moiety attaching a payload moiety through a linker moiety wherein the linker moiety is an organic moiety comprising a geminal diaikyl moiety linked to an ester group having a carboiiyl oxygen, the carboiiyl group of the ester attaching the payload moiety; the light absorbing moiety is an organic moiety attaching the linker moiety in ortho position to a hydroxy! group; and the linker is configured to present the carboiiyl oxygen for reaction with the hydroxy! group.

[0033] In some embodiments, the light-absorbing moiety of a caged compound can be a substituted or unsubstituted poiycyclic aromatic hydrocarbon or closed chain cyanine or hemicyanine and in particular substituted or unsubstituted naphthol.

[0034] In some embodiments, the linker moiety of the caged compound can be a monoa!koxy or a dialkoxy moiety in which the oxy group is linked to the ester group having the carboiiyl oxygen. [0035] In some embodiments, the payload moiety is a substituted or unsubstituted alkyl, aryl, heteroaryl, amirsoalkyl, or oxyalkyl moiety.

[0036] In some embodiments, a preparation of caged compounds herein described is provided in which the linker moiety presents the carbonyl through an the ester, and in particular a derivative of a t-butyl ester, a class that is especially sensitive to acid- catalyzed ester cleavage.

[0037] In some embodiments, a preparation of caged compoimds herein described in which the cleavage process releases a generic entity R^CO jH.a carbox lic acid in which 1 3 is the compound of interested to be delivered,

[0038] In particular, in embodiments herein described photoacid compound herein described are substituted to present hydrophilic substituents (e.g. sulfonate, and in particular poiysulfonat.es such as polysulfonate peptides or oligopeptides, as well as polyethylene glycol groups). In some embodiments water solubility can be imparted by a hydrophilic payload or suitable substituents comprised in the payload to achieve water solubility.

[0039] In particular, in some embodiments, a photoacid herein described has of the general structure according to formula (I):

(1) wherein:

R " is a light-absorbing moiety presenting a hydroxyl group for interaction with the carbonyl oxygen of the R 3 (CO)0 group, wherein the light-absorbing moieiy is a substituted or unsubstituted poiycyclic aromatic hydrocarbon, a substituted or unsubstituted closed chain cyanine, or a substituted or unsubstituted hemieyanine, and wherein the hydroxyl group is cova!ently bonded to the polyey ic aromatic hydrocarbon, the closed chain cyanine, or the hemieyanine, and the hydroxyl group is in a position ortho to X 1 ;

R 3 is a payload moiety, wherein the payload moiety is a substituted or unsubstituted alkyl, aryl, heteroar l, alkoxy, alkylamino, or dialkylamino moiety; and

X 1 is independently selected from the group consisting of C and O; rn is between 0 and 3; and

R 5 and R y' are independently { ' : C ' ( . alkyl groups, cycloalkyl, or substituted or unsubstituted hydrocarbylene groups wherein when R 1 and R 2 are substituted or unsubstituted hydrocarbylene groups they are linked together to form a cyclic moiety.

[0040] In particular, in the photoacid compound of fonnuia (I) and other embodiments herein described, the moiety

(D)

is the linker.

[0041] In particular, in some embodiments, R 1 can be a moiety of Fonnuia (111);

wherein n is between 0 and 5. In particular,

where n is between 0 and 5, and m is between 1 an.

In particular, in some embodiments, R can be a moiety of Formula (V):

wherein R a and R b are independently H, alkyl, or Q-aikyi; Y is N, O, or S; and p is between 1 and 4,

In particular, ula (VI):

wherein R c and R d are independently alkyl and q is between 1 and 4.

In particular, in some embodiments, R can be a moiety of Formula (VII):

wherein r is between I and 4

6] In particular, in some embodiments, R 4 can be a moiety of Formula (VIII):

[0047] A skilled person wil l understand, upon a reading of the present disclosure, that the light absorbing moiety and herein described can be substituted or unsubstitiited and in particular have additional substituents which can be added to impart additional functionalities such as, for example, hydrophilic substituents (e.g. sulfonate, and in particular polysulfonates such as po!ysulfonate peptides or oligopeptides, as wel l as polyethylene glycol groups), and functional groups and/or moieties to connect the photoacid compounds herein described to other molecules and/or substances (e.g. for connection to carbon nanotubes, fullerenes, antibodies, polymers, proteins, lipids, carbohydrates, and others identifiable to a skilled person),

[0048] In embodiments herein described wherein the caged compound is caged within a photoacid the caged compound can be comprised as a payload R J in a photoacid of Fonnuia (I).

[0049] In particular, R J can be an organic moiety such as, for example a substituted or unsubstituted alkyl, aryl, heteroar i, alkoxy, alkylamino, or dialkylamino moiety. In some embodiments, and in particular in embodiments where R J is a substituted or unsubstituted alkyl, aryl, beteroaryl molecule, R 3 is adapted to exist in a carboxylic acid form wherein the carboxylic acid form can be used to provide the carbonyl group of the linker of Formula (II).

[0050] In particular, in some embodiments, R 3 can be a moiety of Fonnuia (IX):

wherein q is between 0 and 5, R a is FI, or substituted or urs substituted alkyl, alkylamino, alkoxy, aryl, arylammo, aryloxy, heteroaryl, hetroarylamino, and iieteroaiyloxy; X is C or N; and wherein when q is greater than 1 , each R" ~ is independent of the other R" ~ substiiuents.

[0051] In particular, in some embodiments wherein R 3 is according to Formula (IX), R J can be selected from the group consisting of Formulas (X)~(XO):

[0052] In particular, in some embodiments, R ~ can be a moiety of Formula wherein n is between 1 and 5, R'\ R^, and R !' are independentfy H, or substituted or unsubstituted alkyl, aikyiamino, alkoxy, aryl, arylamino, aryloxy, heteroaryl, hetroarylammo, and heteroaryloxy; and wherein when n is greater than 1. the R and of each ( H k' : ) unit are independent of the R" and R? of the other units.

[0053] In particular, in some embodiments wherein R 3 is according to Formula (XIII), R 3 can be selected from the group consisting of Formulas (XIV) and (XV :

n particular, in some embodiments, R' can be a moiety of Formula (XVI):

wherein p is between 1 and 5, R 0' , R P , and R r are independently H, or substituted or unsubstituted alkyl, aikyiamino, alkoxy, aryl, arylamino, aryloxy, heteroaryl, hetroarylaroino, and heteroaryloxy; wherein R 5 is H, substituted or unsubstituted alkyl, acyl, aryl; and wherein when p is greater than 1 , the R a and R' j of each C(R )(R p ) unit are independent of the R" and of the other units.

[0055] In particular, in some embodiments wherein R " ' is according to Formula (XVI), R 3 can be of Formula (XVII):

[0056] in particular, in some embodiments wherein R is according to Formula (XVI) and R 6 is acyl, R J can be a peptide or oligopeptide such that the peptide or oligopeptide is attached to the linker via the N-tennimis of the peptide or oligopeptide.

[0057] In particular, in some embodiments, R can be a moiety of Formula (XVIO):

wherein m is between 1 and 5, R 'J , R , and R Y are independently H, or substituted or unsubstituted. alkyl, alkylammo, alkoxy, aryl, arylamino, aryloxy, heteroaryl, hetroarylarnino, and heteroaryloxy; and wherein when m is greater than 1 , the R a and R^ of each C(R")(R |:i ) unit are independent of the R" and R^ of the other units.

8] In particular, in some embo e a moiety of Formula (X

wherein q is between 0 and 4, R a is , or substituted or unsubstituted alkyl, alkylamino, alkoxy, and, arylamino, aryloxy, heteroaryl, hetroarylamino, and heteroaryloxy; ε is a substituted or unsubstituted hydrocarbyiene, X is C or N; and wherein when q is greater th an 1 , each R" is i ndependent of the other R f substituents.

] In embodiments, of methods and systems herein described the administered compou d is uncaged by irradiating the biological environment at a wavelength suitable to excite the light absorbing moiety of the photoacids herein described thus triggering the acid base reaction between the hydroxy, group of the light absorbing moiety and the carbonyl group of the linker thus releasing and therefore uncaging the compound (see Example 1). The term "irradiate" or "illuminate" as used herein relates to the exposure of the target compound and minimal surrounding area to appropriate levels of radiation, in the form of emitting light to result in a desired wavelength in the biological environment where caged compound is to located and intended to be released.

[0060] In some embodiments, suitable wavelength ranges can be determined based on the features of the biological environment that is targeted. For example in applications where irradiation of a brain portion located 4-6 cm into the brain across an intact skull is desired a suitable light radiation can be set at a wavelength between about 900 ran to about 1100 nm. Other suitable wavelengths can be identified by a skilled person in view of the distance and light scattering between the light source and the biological environment to be targeted, as well as physical chemical and biological features of the biological environment that affect light diffusion through the biological environment.

[0061] In some embodiments, in methods and systems herein described administering of a caged compound to a biological emdrorsrnent, is performed with a caged compound caged with a long wavelength absorber, where the long wavelength is a wavelength greater than or equal to about 750 nm.

[0062] In particular the administering can be performed according to techniques and procedures that are identifiable by a skilled person based on the targeted biological environment, desired amount of compound of interest to be administered and uncaged as well as the experimental design and/or conditions of choice which are functional to the desired amount and chemical and/or biological properties of a caged compound to be delivered.

[0CI63] For example, in embodiments, wherein the biological environment is in vitro, administering of the caged compound can be performed by providing the caged compound to the media surrounding the biological environment in a suitable amount and using additional tech iques and approaches identifiable by a skilled person.

[0064] In embodiments, wherein the biological environment comprises an ex vivo specimen administering the caged compound can be performed by providing the caged compound in a suitable formulation to be administered using routes which can be identified by a skilled person based on the specific specimen at issue ,

[0065] In embodiments herein described where the administering is performed in an individual in vivo, the administering can be performed by systemic and/or topical administration.

[0066] The wording "systemic administration" as used herein indicates a route of administration by which a caged compound is brought in contact with the body of the individual, so that the desired effect is systemic (i.e. non limited to the specific tissue where the chemical and/or biological activity of the caged compound is desired). The term "individual" as used herein in the context of treatment includes a single biological organism, including but not limited to, animals and in particular higher animals and in particular vertebrates such as mammals and in particular human beings. The wording "topical administration" as used herein indicates a route of administration by which a caged compound is brought in contact with the specific tissue or other biological environment where the chemical and/or biological activity of the caged compound is desired.

[0067] Exemplary systemic administration routes include enteral and parenteral administration. Enteral administration is a systemic route of administration where the substance is given via the digestive tract, and includes but is not limited to oral administration, administration by gastric feeding tube, administration by duodenal feeding tube, gastrostomy, enteral nutrition, and rectal administration. Parenteral administration is a systemic route of administration where the substance is given by route other than the digestive tract and includes but is not limited to intravenous administration, intra-arteria! administration, intramuscular administration, subcutaneous administration, intradermal, administration, intraperitoneal administration, and intravesical infusion. In some embodiments, administration can be performed intravenously by introducing a liquid formulation including a caged compound in a vein of an individual using intravenous access methods identifiable by a skilled person, including access through the skin into a peripheral vein. In some embodiments, administration of a caged compound can be performed in raperitoneally, by injecting a caged compound in the peritoneum of an individual, and in particular of animals or humans. Intraperitoneal administration is generally preferred when large amounts of blood replacement fluids are needed, or when low blood pressure or other problems prevent the use of a suitable blood vessel for intravenous injection. In some embodiments administration can be performed intragastrically, including administration through a feeding tube, In some embodiments, administration of a caged compound can be performed intracranially.

[0068] In some embodiments a caged compound can be administered topically by applying the caged compound usually included in an appropriate formulation directly where its action is desired. Topical administration include but is not limited to epicutaneous administration, inhalational administration (e.g. in asthma medications), enema, eye drops (E.G. onto the conjunctiva), ear drops, intranasal route (e.g. decongestant nasal sprays), and vaginal administration.

[0069] For example, in embodiments wherein the biological environment is a body- organ, of an indi vidual administering can be performed by injection or ingestion designed for de lively a desired amount in the organ to be targeted. In embodiments wherein the biological environment is skin, administration can be performed injection or topical absorption. In embodiments wherein the biological environment is central nervous system of an individual, administration can be performed by injection or ingestion. In embodiments wherein the biological environment is a lung, administration can be performed by inhalation or injection. In some embodiments, wherein the biological environment is an eye, administration of the caged compound can be performed through topical absoiption of drops or direct intraocular injection. In some embodiments, the biological environment comprises a retinal ganglion cells and the administering can be performed injection. In this embodiment, the compound can be injected iiitraocuiarly into the vitreous cavity targeting retinal ganglion. In some embodiments, the biological environment is a brain and the administering can be performed by injection or ingestion (see Example 3).

[0070] In methods and systems herein described the administered compound can then be uncaged by irradiating the biological environment comprising the caged compound to excite the long wavelength absorber within the biological environment with light at a wavelength in a range from about 900 to about 1100 nm, thus decaging the compound,

[0071] In some embodiments, the irradiating can be performed from a light source that is configured to excite the long wavelength absorber of the caged compound of interest possibly preceded by determining the related parameters which vary in view of the specific light source used and are identifiable by a skilled person upon reading of the present disclosure. For example, in an embodiment wherein the light source is an LED array, parameters include: number of LEDs, power per LED, the distance of the array from the biological environment, separation between LEDs, and LED/lens viewing half angle

[0072] In some embodiments, irradiation parameters such as distance, intensity, and continuity of irradiation, con be predetermined in view of the specific biological environment to be irradiated according to techniques and approaches identifiable by a skilled person. For example, predetermination can be performed by thermoacoustic, photoacoustic imaging, or by availably known near infrared ( R) windows based on known absorption coefficients and/or absorption coefficient spectrums. Thermoacoustic imaging can be performed for example by a device delivering electromagnetic radiation to the biological environment and an acoustic detector placed to the outside surface to measure the strength of the thermoacoustic waves. Photoacoustic imaging can be performed for example by a device delivering electromagnetic radiation to the biological environment and an ultrasonic detector placed to the outside surface to measure the strength of the ultrasonic emission. In addition, the molar extinction coefficient of deoxyhemoglobin has its highest absorption peak at 420 nm and a second peak at 580 nm, and its spectrum then gradually decreases as light wavelength increases. Oxyhemoglobin shows its highest absorption peak at 410 nm, and two secondary peaks at about 550 nm and about 600 nm. The absorption spectrum of water can be in the range from about 250 to about 1000 nm. The absorption spectrum for arteries (Sa0 2 ~ 98%) has λαάη of about 686 nm and NIR. window has a range of about 634 - about 756 am, The absorption spectrum for veins (Sv0 2 ~ 60%) has X mis of 730 nm and a NIR window of about 664 - about 934 nm. The absorption spectrum for brain tissue (St.0 2 ~ 70%). λ 11Ί1ϊ! = about 730 nm and a NIR window of about 656 - about 916 nm. Infrared illumination between 900-1 lOOnm wavel en gth has depth of penetration of 4-6 cm in to the brain across an intact skull (FIG, 11 and FIG. 12).

[0073] In some embodiments, the parameters for irradiation necessary from the light source to excite a long wavelength absorber to perform the chemical change can be determined by a skil led person in a laboratory setting for example by exposing the compound to a one or more wavelength and then detecting presence of an excited long wavelength absorber through techniques such as liquid chromatography-mass spectrometry (LC-MS), infrared (IR), nuclear magnetic (NMR) spectroscopy and additional techniques identifiable by a skilled person. Alternatively, or in addition, the skilled person can determine the irradiation associated to a desired delivery of a compound of interest by caging the long wavelength absorber to a compound, irradiating the resulting caged compound at one or more settings and detecting presence of a decaged compound in the one or more settings to identify a setting wherein a desired presence of the compound of interest is detected. Samples that contain the caged compound, the decaged compound, and the long wavelength absorber, can be for example collected and analyzed through LC-MS, IR, or NMR spectroscopy (see Example 2). In some embodiments, detection of decaging can be performed by labeling the caged compound with a suitable label configured to emit a labeling signal upon decaging of the compound.

[0074] The terms "label" and "labeled molecule" as used herein as a component of a complex or molecule referring to a molecule capable of detection, including but not limited to radioactive isotopes, ffuorophores, chemiiuminescent dyes, chromophores, enzymes, enzymes substrates, enzyme cefaclors, enzyme inhibitors, dyes, metal ions, nanoparticles, metal sols, ligands (such as biotin, avidin, streptavidin or haptens) and the like. The term "fluorophore" refers to a substance or a portion thereof which is capable of exhibiting fluorescence in a detectable image. As a consequence, the wording "labeling signal" as used herein indicates the signal emitted from the label that allows detection of the label, including but not limited to radioactivity, fluorescence, chemiluminescence, production of a compound in outcome of an enzymatic reaction and the like.

[0075] The terms "detect" or "detection" as used herein indicates the determination of the existence, presence or fact of a target in a limited portion of space, including but not limited to a sample, a reaction mixture, a molecular complex and a substrate. The "detect" or "detection" as used herein can comprise determination of chemical and/or biological properties of the target, including but not limited to ability to interact, and in particular bind, other compounds, ability to activate another compound and additional properties identifiable by a skilled person upon reading of the present disclosure. The detection can be quantitative or qualitative. A detection is "quantitative" when it refers, relates to, or involves the measurement of quantity or amount of the target or signal (also referred as quantitation), which includes but is not limited to any analysis designed to determine the amounts or proportions of the target or signal. A detection is "qualitative" when it refers, relates to, or involves identification of a quality or kind of the target or signal in terms of relati ve abundance to another target or signal , which is not quantified.

[0076] For example, in some embodiments the caged compound can be labeled with fluorescent/color indicating molecule that only fluoresces/ changes color when decaged and detectable fluorescence/ color change is a positive indicator which wavelength is necessary to excite the long wavelength absorber. As a result, the fluorescence can be identified either through fluorescence microscopy or spectroscopy..

[0077] In some embodiments, in vitro experiments of the long wavelength absorber or the caged compound can be performed to identify wavelengths and/or settings for a desired in vitro biological environment and/or preliminary data directed to prepare in vivo experiments and in particular to minimize experimentation in the in vivo model. In an in vivo animal model experiment, a skilled person can use a light source as previously described (e.g., 1R LED, an LCD panel, and additional sources identifiable by a skilled person) to irradiate animals that contain an administered compound caged by a long wavelength absorber. Based on data from corresponding in vitro experiments, the skilled person can adjust the specific parameters (e.g. distance and power) necessary for a Song wavelength absorber to be excited by tracking the activity of the liberated uncaged compound (e.g. fluorescence, downstream drag effects, side-effects, death) or mass spectroscopic analysis of samples collected. For use on tissue and cell types in the body, a skilled person is aware of the different unique light absorption characteristics known in the medical community. A skilled person can empirically determine light absorption for example by placing probes (e.g., an optical probe for collecting returned light, or other device capable of same) at different sites of the tissue or body part; the probes at the cite measure and read out the wavelength absorbed when exposed to light in the location where the probe is placed. After determining the setting necessar for irradiation based on probe read outs, the skilled person is able to validate decaging from the irradiation from by tracking the activity of the liberated uncaged compound (e.g. fluorescence, downstream drag effects, side-effects, and death) or be mass spectroscopy sample analysis when available.

[0078] In some embodiments, the skilled perso can adjust the dista ce of the light source to the target in conjunction with changes to the power settings of the Sight source. In embodiments, when the distance of the light source is not fixed, separate measurements for the power of irradiation can be made for each tested range of distance related to the specific lo g wavelength absorber used. In embodiments, when the distance of the light source is fixed, measurements for the power of irradiation are only for that range and directly related to the specific long wavelength absorber used. These experiments are typically done first in vitro, followed by non-human in vivo samples, and then ex vivo samples (see Examples 2, 3, 4). Experiments with varying distances and power of the light source irradiating human patients are performed in clinical trials under strict regulation and oversight. After clinical trials determine safe conditions, irradiation settings coupled with administering the caged compound is ready for general use.

[0079] In some embodiments, the particular wavelength range is known not to produce reactive oxygen species in biological environments. [0080] In some embodiments, the irradiating can be performed following the administering of the caged compound at a time that is dependent on a window of time for a caged compound to reach a specific area of the biological environment from administration and for its clearance (through means not limited to diffusion, transport, secretion, or metabolic changes) in order to couple control release via irradiation. In some of those embodiments the window of time can be determined based on the feature of the biological environment at issue, for example in an in vitro environment of ceil components, in which there is instantaneous entry and no exit for the compound upon administration the window of time is dependent on parameters such as solubility and coefficient of diffusion of the caged compound in the coiture media, amount administered and additional parameters identifiable by a skil led person. In an in vivo biological environment wherein the administering is performed systemically to an individual, additional factors can also be taken into account including, clearance time (e.g. through metabolism and/or secretion), cell uptake and blood flow as well as additional parameters identifiable by a skilled person. In an in vitro environment of cell lines or an i vivo environment comprising of microorganisms, the window of time can be empirically determined by performing cell uptake assays with labeled substrates, LC-MS assays, and/or metabolite assays (see Example 2).

[0081 ] In some embodiments, where the biological environment is formed by in vivo or ex vivo environments larger than microorganisms, a window of time between administering and irradiating can be identified by pharmacokinetic approaches identifiable by a skilled person, which can be performed for example to establish a desired time for a compound to be absorbed, distributed, and eliminated or clearance (through metabolism or secretion) in the biological environment (see Example 2, Example 3, and Example 4). In particular, absorption refers to the ability and process of a dosage reaching the bloodstream and the related time can be measured according to approaches identifiable by a skilled person. For example compounds administered intravenously typically do not require absorption since they immediately reach the vascular system. Absorption can be for example determined by in vitro models such as membrane-based .models (PAMPA), cell culture -based models (Caco-2), and transporter assays. Determining distribution of a compound can be performed by calculating the volume in which the drug is distributed is a product of the drug ' s dose divided by the plasma concentration ((Vd) = dose/plasma concentration) which is dependent on half-life of the compound. The half-life of the compound can be determined by in vitro systems to mimic the in vivo environments such as hepatocyte uptake assays or commercially available microsome systems utilizing cytochrome P450 enzymes.

[0082] In vitro half-life (ti /2 ) can be determined using the equation: t t# = In 2/(-k), where k represents the terminal elimination rate constant and is calculated as the negative slope of the line defined by the linear regression of the natural log loss of compound and incubation time. Clearance is defined as the volume of fluid that is completely cleared of the compound per unit time. Determining clearance of a compound can be performed by calculating the extraction ratio, blood (or serum) flow rates, solubility, and compute aggregation determined by in vitro systems to mimic the in vivo environments such as for example human eryopreserved hepatocytes or liver microsomes of the biological environment. Extraction ratios are calculated as the difference arterial compound concentration and venous drug concentration to the arterial compound concentration at the organ outflow. Flow rates can be calculated by blood pressure measurements. Solubility and compound aggregation is determined by flow cytometry analysis. Flow cytometry analysis can be performed by adding the compound to biologicai buffers that mimic the biologicai environment and measuring the particle size distribution with a flow cyto.met.er.

[0083] In some embodiments, light is emitted through a light source under conditions that irradiate a biological environment to produce a set, known, and consistent wavelength, known or predetermined, in view of the specific environment, the desired effect and the conditions of choice as will be understood by a skil led person. For example, in some embodiments, the light source is selected to irradiate at levels that do not change the physi cal nature of the biological environment whi le in other embodiments the light source is selected to physically alters the biologicai environment (e.g. by melting or excision) in accordance with the specific biologicai environment and the experimental design as will be understood by a skilled person. Similarly, in some embodiments, the light source can irradiate the biological environment in pulses, and in other embodiments, the light source irradiates in a continuous fashion in accordance with the amounts, timing and location of the caged compound that is intended to be released. Exemplary light sources for this purpo.se are light emitting diodes, or LEDs, which are capable of delivering a controlled and set desired wavelength in the range indicated (e.g., 900 nm to 1100 nm) within the biological environment of the practitioner's choice. Exemplary circuit designs to emit near infrared wavelength are depicted in FIGS. 7.4 and 7B.

[0084] In some embodiments, a light source is a Samp, LCD panel, or LED display which is comprised of a non-portable device or a portable device. In some embodiments, the non-portabie device or portable device containing the light source is a microscope, endoscope, flashlights, garments (see FIG. 6), or glasses. The non-portable or portable device comprising light sources, in some embodiments, also comprises image intensifier to further increase the emitted power from such devices in order to activate the preparation of molecules. Turning now to the description of FIG. 6, a wearable ultra-low weight, low power, time-gated, wearable infrared LED grid emitter (600), (610) is shown that transmits the energy deep into the brain across a intact skull, scalp a d hair and triggers the release of caged compounds.

[0085] The LED emitter system according to the present disclosure uses longer wavelengths (900-1100 nm) and therefore reduces absorption and scattering of the energy, resulting in deeper light penetration into brain tissue (4-6 cm). FIGS. 7A and 7B show an exemplary arrangement of and array of infrared LEDs, each LED in series with a corresponding variable resistor. Alternatively, each LED can be put in series with a related transistor, as shown in FIG. 7B, each transistor being controlled by a dedicated control signal ctrll, etri ' 2, . ... Ctrin.

[0086] According to some embodiments of the present disclosure, the grid comprises a lensing arrangement that provides a more efficient energy delivery and allows reduction in the number of LED's. Reference can be made, for example, to FIG. u 9 which shows an improved Sight distribution angle in presence of a lens (e.g. a bi-concave Sens, as shown in the figure).. [0087] The LED and associated systems are configured such that the duty cycle and time-gated functions trigger and release preferentially the caged-drug in brain tissue but not inside brain vessels. In order to do so, a circuit (800) like the o e shown in FJG.J8 can be provided where, upon switching on of a general switch SI and a switch S2, the latter being triggered either by a gating signal (810) or (820) by a clock circuit (830). Therefore, the LED array (840) is powered only when bot switches S I and S2 are on .

[0088] Turning now to the representation of FIG. 6A, each module (610) comprises an infrared transmitter section (650) and an infrared detector (660) section, as better shown in FIG. 6B. The infrared transmitter section (650) comprises, for example, an array of infrared LEDs like those shown in FIGS. 7A and 7B with associated variable resistors or transistors a d one or more lenses like those shown in FIG. uu . On the other hand, a possible embodiment of the infrared detector section (660) of FIG. 6B is shown in FIG, . where the infrared detector section is configured as a PIN photodiode-transimpedance amplifier (TIA) detection module (1000), comprised of a PIN photodiode (1010) and a transimpedance amplifier ( 1020). Each detector section (660) of a module (610) is expected to allow detection of a transmission signal sent intracraiiialiy from the transmitter section (650) of a module (610) located on an opposite side of the head of the patient.

[0089] According to several embodiments of the present disclosure, the circuits employed in the above described device can be designed to require ultra-low power and dissipate little to no heat. By way of example, the LED array can be driven using two small lithium ion batteries with protection circuitry. Additionally, the LED array can be designed to operate in a range such that the maximal permissible temperature rise does not exceed I °C above body temperature, an international standard for biomedical devices.

[0090] If desired, the LED emitter system according to the present disclosure can be configured such that it can work in conjunction with infrared spectroscopy. Backscatter from the emitters can be collected to record medical information such as 0 2 saturation and blood flow. [0091] In particular, according to several embodiments of the present disclosure, an infrared LED array will have one or more following properties: 1) The LED emitter system will use longer wavelengths in the range of 900-1100 nm, which will ensure low absorption and scattering of the energy, resulting in deeper light penetration. 2) The grid will be designed to have one detector and one LED per module. The number of modules in a grid depends on the size of the device. The grid will be designed to uniformly illuminate the brain by use of lenses coupled with LEDs and will be designed to be lightweight and portable. 3) The LED and associated systems will be configured such that the duty cycle and associated circuits (see, e.g., FIGS. 7A, 7B and 8 described above) require ultra-low power and dissipate little to no heat. Estimates predict that the LED array can be driven by two lithium ion batteries with protection circuitry. 4) The LED system operates in a range such that the maximal permissible temperature rise does not exceed 1 °C by using a detector array to adaptive ly adjust the power of LEDs and thereby controlling the temperature. 5) The array will be addressable and reeonfigurable to turn on/off different sections of the grid.

[0092] In embodiments herein described, the compound is selected to have chemical and/or biological reactions of interest relative to a biological environment of interest. In particular, chemical or biological reactions characterizing a compound of interest comprise chemical reactions resulting in imaging a targeted biological environment. In some of those embodiments, the caged compou d can be a label. In particular, selection of a suitable label can be performed to include label that when caged to the photoacid, are not able to emit the signal and regain the ability emit the signal upon decaging through excitation of the coupled photoacid. Detection of the uncaged label can be performed through suitable detector able to measure the labeling signal emitted by the label at issue that are identifiable by a skilled person.

[0093] For example, in embodiment the label is fluorescein, and when caged to the photoacid, the caged fluorescein compound does not fluoresce. Upon decaging through excitation of the light absorbi g moiety of the photoacid, fluorescein is expected to be decaged and then exhibit detectable fluorescence through approaches such as fluorescent spectroscopy or microscopy. Additional exemplary labels suitable to be administered as a caged compound herein described are expected to comprise as rhodamine, AiexaFluor dyes, Cy3, and Cy5.

[0094] in some embodiments, the compound to be cages is a drug that can be diagnostic or therapeutic, or a candidate drug. In some of these embodiments, the drug is a candidate drug. In an embodiment of a caged candidate drug, the candidate compound can be selected based on one or more of the following properties: 1) having minimal pharmacologic activity in the caged form, 2) following photonic decaging, the decaged form retaining pharmacological activity, 3) being chemically stable in conditions for transport and storage, 4) being be safe and not generate toxic metabolites upon treatment, 5) having acceptable pharmacokinetic and pharmacodynamic profile for entry into the desired site of activity, 6) not requiring special handling, 7) and requiring minimal formulation for administration (see Example 2), Exemplary drugs expected to be deliverable through the methods and systems herein described comprise In some embodiments, the compounds caged by a long wavelength absorber are neurotransmitters, anti-cancer agents, sedatives, antibodies, antibiotics, or protein therapeutics.

[0095] In some embodiments, a plurality of caged compounds can be administered simultaneously or sequentially. In this embodiment, separate compounds enter the biological environment, and upon decaging, the compounds are free to chemically or biologically react independently or together. In some embodiments, one compound is an imaging molecule and the separate compound is not an imaging compound. In a further embodiment, positive detectio of the imaging compound from decaging indicates decaging of the compound that is not an imaging compound. In other embodiments, the separate compounds released bind together to form a new complex. In other embodiments, one separate molecule enhances the effect of a second separate molecule. In further embodiments, the multiple caged compounds can be caged by different long wavelength absorbers that decage at different wavelengths allowing precise control of decaging.

[0096] In embodiments where the uncaged compound can have an established administering protocol possibly associated to specific fomiiilation (e.g. formulation for injection or parenteral administration). In some of those embodiments, administering the caged compound can be performed in accordance with the established protocol possibly following testing to verify specific conditions associated with, the caging of the compound at issue. For example, the skilled person can determine administering conditions by testing set doses, formulations, scheduled administration and/or other parameters identifying the administering and by irradiating the biological environment and different intervals for different ranges of time to detect an occurred uncaging. The skilled person can repeat these experiments for every dose investigated. Based on the data, the skilled person determines the timing between administering and irradiating through empirical analysis to determine the optimal l ength of time between admini stering the caged compound and the caged compound localizes to the desired site of decaging. These factors are dependent on absorption and diffusion rates of the compound in the specific biological environment and are different for each set of conditions. These factors directly connect to the efficacy and dosage which can be determined by the skilled person through a series of control and variable experiments (see Example 2 and Example 4).

[0097] In some embodiments, the administering of caged compounds can be performed to target a "leaky" biological environment that is characterized by an accumulation of fluids (e.g. blood) accompanied by a higher concentration of solutes and target molecules (e.g. cytokines and additional molecule produced in connection with an inflammatory response) (leaky environment). Exemplary leaky environments comprise tissues damaged as a result of an injury. In such embodiments, the site of injury may become "leaky" resulting blood vessel dilation and increased blood flow to the site. Additional exemplary leaky environments can comprise certain tumors and biological environments exhibiting an inflamed response. In those embodiments the administering can be performed taking into account that the difference in concentration, diffusion clearance rate and accumulation of the caged compound in the leaky environment with respect to competing surroundings.

[0098] In some embodiments, the administering is performed to target a biological environment exhibiting inflammation or and inflammatory response. The wording "inflammation" and inflammatory response as used herein indicate the complex biological response of vascular tissues of an individual to harmful stimuli, such as pathogens, damaged ceils, or irritants, and includes secretion of cytokines and more particularly of pro-inflammatory cytokines, which comprise cytokines which are produced predominantly by activated immune ceils such as microglia and are involved in the amplification of inflammatory reactions. Exemplary inflammations include acute inflammation and chronic inflammation. The wording "acute inflammation" as used herein indicates a short-term process characterized by the classic signs of inflammation (swel ling, redness, pain, heat, and loss of function) due to the infiltration of the tissues by plasma and leukocytes. An acute inflammation typically occurs as long as the injurious stimulus is present and ceases once the stimulus has been removed, broken down, or walled off by scarring (fibrosis). The wording "chronic inflammation" as used herein indicates a condition characterized by concurrent active inflammation, tissue destruction, and attempts at repair. Chronic inflammation is not characterized by the classic signs of acute inflammation listed above. Instead, chronically inflamed tissue is characterized by the infiltration of mononuclear immune cel ls (monocytes, macrophages, lymphocytes, and plasma cells), tissue destruction, and attempts at healing, which include angiogenesis and fibrosis. Chronic inflammation results from several known disease in the art that include asthma, an autoimmune disease, celiac disease, chronic prostatitis, inflammatory disease, pelvic inflammatory disease, arthritis, age related macular degeneration, sarcoidosis, vasculitis , diabetic retinopathy or nephropathy.

[0099] In embodiments, wherein the administering is performed to target a biological environment exhibiting inflammation, administered amounts of the caged compound, time of administration and irradiation as well as related wavelengths, can be determined based on the fact that cells exhibiting an inflammatory response can more easily absorb light compared to cells not in an inflammatory state. Exploiting this fact, a skilled person is expected to be able selectively target inflammation sites by exposing the biological environment to irradiation that is above the threshold of power to be absorbed by inflammatory cells, but below the threshold to be absorbed by non-inflammatory cells. The skilled person can determine a suitable range and/or power of irradiation necessary for a desired uncaging for example, through experiments in in vitro, in vivo, and ex vivo samples that are not exhibiting an inflammatory response and separate samples that are exhibiting an inflammatory response as described above by the methods described above,

[00100] In some embodiments, the administering can be performed on an inflamed biologic environment following a traumatic event. In some embodiments the administering can be performed in biologic environment exhibiting a chronic inflammation. An example of chronic milamrnation tha is associated with lowered pH is age related macular degeneration (AMD, FIG, 2), a condition of the eye. In embodiments wherein the biological environment is the eye, the compound is a neuromodulator. In this embodiment, the neuromodulator is a photoactivated cellular switch injected through intraocular injection. Further in this embodiment, an infrared stimulator in eyeglasses converts visible light to mfrared and transmits mfrared light into the eye. As a result, cell ion channels impart light sensitivity to these neurons and, by doing so, effectively bypass damaged photoreceptors. This embodiment is a retinal cellular prosthesis that enables light sensing by the retina in the blind. Thus, decaging of the neuromodulator is evident by light sensing.

[00101] In some embodiments, the administering can be performed to a target biologic environment presenting an acidic p in! f less than 7) which can occur in various biologic environments and in particular in a biologic environment presenting an inflammatory response. In particular, in those embodiments the administering can be performed taking into account the effect of the acidic pH on the uncaging of the compound by the photoacid, which is expected to be enhanced. As a result, the administering of a compound caged by a photoacid in a biologic environment having an acidic pH is expected to result in an enhanced deliver of the compound when uncaged by low power infrared light. In some embodiments, compounds also comprise an "armed" synthetic switch. In these embodiments, the "armed" synthetic switch responds to drops in pH of the biological environment.

[00102] In some embodiments, a preparation of molecules herein described can be administered to a tissue including neurons to result in a high density neural interface for neuromodulation as well as to be used for localized drug delivery and diagnosis for patients with neurological diseases (e.g., retina, brain, spinal chord, and additional environments identifiable by a skilled person.).

[001031 In some embodiments, the biological environment is a brain that has a traumatic injury. In further embodiments, the caged compound is a therapeutic drugs (such as Erythropoietin, Gabapentin (Neurontin©), pheno barbital, N-acetyicysteine, or progesterone) or an imaging molecule (such as fluorescein, rhodamme, Cy2, Cy3, Cy5, Cy7, AlexaFluor molecules). Caged compounds administered to the brain after a traumatic injury can gain access into brain parenchyma through compromised areas of the blood-brain barrier (BBB) during early stages where flow is not severely compromised. When the compound is caged by a photoacid, local pH drops from the traumatic brain injury (TBI) is expected enhance the photoacid effect. In one embodiment, caged drugs can be intravenously injected into the bloodstream of an injured patient. In this embodiment, the caged drug is in circulation and extravasates through the compromised BBB into injured brain tissues and the patient wears a garment that wraps around the skull like a modified balaclava or swimmer cap type hood that is embedded with an array of time-gated infrared (IR) LEDs as depicted FIG. 4B. Optionally, a cervical collar is placed to keep the patient's head secure. In the embodiment, the LEDs embedded in the garment is expected to expose light to trigger the decaging of the drug to treat the injury. This embodiment can be used to treat or triage a patient for subsequent medical procedures.

[00104] In accordance with the embodiment shown in FIG, 6, infrared light is applied across the skull to trigger the targeted release of TBI-combating drugs near the area of brain damage. The possibility of drug release in other brain regions is limited by using a synthetic switch that is selectively triggered in the presence of a tissue-specific p change in the area of early brain injury. FIG. 6 shows a body- worn, infrared-emitting device (600) placed on the injured patient. Device (600) comprises a plurality of modules (610), each module including infrared transmitters and infrared detectors, later shown.

[OOlOSj A skilled person in the art tests caged compounds for brain injury in a set of experiments as outlined in FIGS.JA and 5B. In FIG^SA, both the caged compounds and the infrared emitters can be optimized using bench top and cel lular assays (Activities I and II) as described earlier. Fresh cadaver studies are conducted to help provide a test structure that has similar size and stmctural and geometrical constraints (Activity I I I, see Example 3). Upon obtaining an optimized caged compound, pharmacokinetic studies are conducted in vitro (Activity IV, see Example 4). The long wavelength absorber, the caged compound, and the decaged compound after release from light absorption (Activity V) are tested for toxicity in animal models. In addition, the approach is validated in animal models of TBI (VI) as depicted in FIG, 5B.

[00106] In some embodiments, a preparation of caged compounds herein described can be used outside the central and peripheral nervous systems to image and treat other body parts including abnormal cellular growth such as that seen in cancer.

[00107] In some embodiments, a preparation of caged compounds herein described can be used in laboratory testing of biopsy tissue or parenchymal or hematopoietic cells

[001081 In some embodiments, the biological environment can be a cell culture. In these embodiments, administering of caged compounds can be performed by providing the caged compound to media of the cell culture. In these embodiments, irradiation is performed by a non-portable or portable device. Distance to the ceil culture from the non-portable or portable device is different for each device and power setting and is determined by empirical means as previously described. Decaging is detected by measuring the activity of decaged compound or mass spectroscopic analysis of samples from the cell culture.

[00109] In some embodiments, the biological environment is a tumor. In this embodiment, administering of caged compounds comprises direct injectio into the tumor. Here, the compound is an anti-cancer drug, a live/dead cell dye, or imaging molecule. In these embodiments, irradiation is performed by a non-portable or portable device. Distance to the tumor from the non-portable or portable device is different for each device and power setting and is determined by empirical means as previously described. Optionally, two caged molecules can be administered where one caged molecule when decaged, visual detection is possible resulting in a positive confirmation that a second caged molecule has been decaged as well. Dosage calculation is done by methods known to a skilled person (see Example 2).

[00110] In some embodiments, a preparation of caged compounds herein described which can deliver a payload of drugs, imaging molecules, or modulate neuronal activity, and can be activated upon exposure to light with longer wavelengths (800 nm - 1 .5 microns), can be administered by injection (e.g. intravenously or directly into tissue such as the vitreous cavity of the eye), ingestion or inhalation, or topically applied to an individual (e.g. as a cream or a drop).

[00111] In some embodiments, caged compounds herein described can be provided as a pari of systems to administer caged compounds based and decage caged compounds based on irradiation according to methods herein described. In some embodiments, the system can comprise one or more compound of interest caged within a photoacid, and a light emitting device adapted to irradiate light at a wavelength of from about 900 nm to about 1 100 nm for simultaneous combined or sequential use in a method of the disclosure.

[00112] In some embodiments, the systems herein described can be provided in the form of kits of parts. In a kit of parts, a long wavelength absorber, a compound, reagents necessary to cage the compound in the long wavelength absorber, devices suitable to administer the caged the compound to a biological environment, and a light source to emit a wavelength in a range from 900-1100 nm. In some embodiments, the systems to systems to perform administering and the decaging of caged compounds herein described can be provided in the form of kits of parts. In a kit of parts, one or more long wavelength absorbers, compounds, and other reagents to perform the reactions can be comprised in the kit independently. The long wavelength absorber or compound can be included in one or more compositions, and each compound can be in a composition together with a suitable vehicle.

[00113] In particular, in some embodiments, the caged compound herein described can be comprised in a composition together with a suitable vehicle. The term '"vehicle" as used herein indicates any of various media acting usually as solvents, carriers, binders or diluents for the caged compounds that are comprised in the composition as an active ingredient,

[00114| In particular, the composition including the caged compound can be used in one of the methods or systems herein described. In some embodimen ts, the composition is a pharmaceutical composition and the vehicle is a pharmaceutically acceptable vehicle. In those embodiments one ore more caged compounds herein described can be included in pharmaceutical compositions together with an excipient or diluent. In particular, in some embodiments, disclosed are pharmaceutical compositions which contain at least one caged compound as herein described, in combination with one or more compatible and pharmaceutically acceptable vehicles, and in particular with pharmaceutically acceptable diluents or excipienis. In those pharmaceutical compositions the caged compound can be administered as an active ingredient for treatment or prevention of a condition in an individual ,

[00115] The term "treatment" as used herein mdicates any activity that is part of a medical care for, or deals with, a condition, medically or surgically.

[00116] The term "prevention" as used herein indicates any activity which reduces the burden of mortality or morbidity from a condition in an individual. This takes place at primary, secondary and tertiary prevention levels, wherein: a) primary prevention avoids the development of a disease; b) secondary prevention activities are aimed at early disease treatment, thereby increasing opportunities for interventions to prevent progression of the disease and emergence of symptoms; and c) tertiary prevention reduces the negative impact of an already established disease by restoring function and reducing disease-related complications.

[001171 The term "condition" as used herein indicates a physical status of the bod)' of an individual (as a whole or as one or more of its parts), that does not conform to a standard physical status associated with a state of complete physical, mental and social wel l-being for the individual. Conditions herein described include but are not limited disorders and diseases wherein the term "disorder" indicates a condition of the living individual that is associated to a functional abnormality of the body or of any of its parts, and the term "disease" indicates a condition of the living individual that impairs norma ί functioning of the body or of any of its parts and is typically manifested by distinguishing signs and symptoms.

[00118] The term "excipient" as used herein indicates an inactive substance used as a carrier for the active ingredients of a medication. Suitable excipients for the pharmaceutical compositions herein described include any substance that enhances the ability of the body of an individual to absorb the multi-ligand capture agents or combinations thereof. Suitable excipients also include any substance that can be used to bulk up formulations with the peptides or combinations thereof, to allow for convenient and accurate dosage. In addition to their use in the single-dosage quantity, excipients can be used in the manufacturing process to aid in the handling of the peptides or combinations thereof concerned. Depending on the route of administration, and form of medication, different excipients can be used. Exemplary excipients include, but are not limited to, antiadherents, binders, coatings, disintegrants, fillers, flavors (such as sweeteners) and colors, glidants, lubricants, preservatives, sorbents.

[00119] The term "diluent" as used herein indicates a diluting agent which is issued to dilute or carry an active ingredient of a composition. Suitable diluents include any substance that can decrease the viscosity of a medicinal preparation.

[00120] Further characteristics of the present disclosure will become more apparent hereinafter from the following detailed disclosure by way or illustration only with reference to an experimental section.

EXAMPLES

[00121] The caged compounds, compositions methods and system herein described are further illustrated in the following examples, which are provided by way of illustration and are not intended to be limiting.

[00122] In particular, the following examples illustrate exemplary caged compound delivery methods and systems and related compounds and compositions. A person skil led in the art will appreciate the applicability and the necessary modifications to adapt the features described in detail in the present section, to additional caged compounds, methods and systems according to embodiments of the present disclosure.

Example 1: Photoaeids as Long Wavelength Absorbers

[001231 Exemplary photoaeids caging a compound of interest comprise photoaeids of Formula (I) wherein R 1 is

[00124| FIG. 1 exemplifies uncaging of a compound caged with a long wavelength absorber. The compound is labeled as a drug that is linked to a NIR absorber. Upon NIR irradiation, the linker is cleaved allowing release or dec-aging of the drug.

[00125] In particular, a photoacid compound is expected to be decaged in vitro using the following procedure. In a quartz cuvette, the photoacid compound (0.001 M) is dissolved in spectroscopic-grade acetonitrile and irradiated at 300 nm usi g a 1-kW xenon arc lamp at room temperature for 15 minutes. Aliquots are removed and analyzed by analytical LC-MS utilizing a gradient elution of 10-100% acetonitrile-water over 10 minutes. The data shown represents t :::: 0 minutes in the lower LC-MS trace, t ::: 15 minutes in the upper LC-MS trace.

[00126] Suitable adjustments for decaging in vivo and/or with specific compound are identifiable by a skilled person upon reading of the present disclosure.

[001271 Techniques used to identify lead candidate caged and photonicaliy decaged etomidate- and cyclosporine -based nanocaged compounds are described herein. The biological activity of caged and decaged etomidate- and cyclosporine-based nanocaged compounds is expected to be suitable in established ion channel and mterieukin-2 (IL-2) inhibition assays, respectively. Candidates with a 50% effective concentration (EC50) with near equivalent with the decaged compound following photonic liberation can be further evaluated. The effect of ambient pH on decaging can also be determined, since necrotic tissue are more acidic as compared to normal tissue. The pH of the medium is expected to be adjustable using lactic acid, so that the final pH will be 7.4, 6.0 and 5.0. The level and rate of decaging will be determined before and after photonic decaging over time, where the active moiety and its metabolites will be measured using LC-MS assay.

[00128] Determining the Level of Decaging: the level and time required to liberate the active moiety can be determined using a time course study. The time of IR activation can be initially performed in biological assays performed in in vitro. Caged compounds can be incubated in medium and undergo photonic decaging. The time require to and extend of active drug liberation can be determined using ion channel and IL-2 inhibition assay to evaluate etomidate- and cycSosporine-caged compounds, respectively. The intensity of infrared excitation can be varied to determine the optimal wavelength and timing required to maximally liberate the active moiety. In addition, the impact of microenvironment pH on decaging of the active moiety can be determined by adjusting the medium pH at 7,4, 6.0 and 5.0. To affirm that biological activity is a direct consequence of decaging the active moiety, medium is collected to determine the levels of caged and decaged compounds via LC-MS. An increasing ratio of decaged versus nanocaged is expected to guide in the identification of the hit compounds.

[00129] Ion Channel Inhibition Assay: HEK293 cells with heterologous expression of a4 and β3 subunits of GABA(A receptor, using transfeeiion with DEAE-dextran, are used in inhibition assays. Whole-ceil currents can be recorded at a holding potential of -60 mV (EPC-9 amplifier, HEKA Elektronik, controlled by Pulse software) elicited by drug candidates at a range of concentrations, that has or has not undergone photonic decaging, and applied via a home-built, multibarrel, gravity-fed, solenoid-gated application system, that has an exchange time of ~10 ms. The external solution can contain (in mM): 142 Nad, 1 CaCI2, 6 MgC12, 8 KG, 10 glucose, and 10 HEPES, H 7.4 (-300 mOsm). The pipette internal solution can contain in mM: 140 CsCl, 4 NaCl, 0.5 ( aC ; .?.. 10 HEPES, 5 EGTA, 2 Mg2+, ATP, and 0.2 GTP. Pipettes can be pulled using a standard Narishige puller. Pipette resistance can be -4 MOhms.

[00130] CSA Biological Activity: Human mammary epithelial cells (HMEC) can be incubated for 2 hours with cyclosporine (CSA) or the caged-CSA. that has or has not undergone photonic decaging. IL-2 biological activity can be determined using NO elaboration and e OS expression. Medium can be collected and the level of NO can be determined using Griess reaction. In addition the cells can be collected and total RNA can be isolated using TRIzol reagent (Invitrogen) to determine the level of eNOS expression.

[00131| Metabolic Fate Analysis: Lead eandidate(s) can be assessed for metabolic fate using hepatic microsomes derived from male Sprague-Dawley (In Vitro Technologies, Baltimore, MD). Five μΤ 100 μ.Μ lead compound can be added into glass vials and heated at 37°C, where 395 μ] microsomes (20 mg/mL) suspended in 0.1 M PBS (pH 7.4) can be added and incubated for 3 min. The reaction is initiated by adding 100 p.L 50 mM NADPH in 0. 1 M PBS, pH 7.4. At 8 time points, from 3 to 90 min, 50 μΕ can be removed, and reaction quenched using 300 ,uL MeOH that contains an appropriate IS control. Samples can then he centrifuged at 300 X g for 20 min at 4°C, where the supernatant can be scanned for metabolites using LCQ Deca LC-MS. The structure of the metabolites can be reconstructed using deconvolution of the parent compound. The kinetics of metabolite formation and clearance can be analyzed using a linear fit of the natural logarithm of the ratio of the compound peak area to the internal standard peak area against time. A control reaction (without NADPH co-factor) can be used in order to assess the amount of thermal breakdown, insolubility and non-specific binding that contributes to the overall loss of the test compound. Using the assumption that the substrate concentration of I μΜ, Km, CLint values can be calculated from the negative slope of the linear fit divided by the microsomal concentration.

[00132] Pharmacokinetics and Toxicokinetics Study in Mice: Lead candidate can be assessed for pharmacokinetic and toxicokinetics. C57BL can be administered a dosage and randomly assigned to 1 of 7 time points for blood collection. At time of sacrifice, blood and vital organs can be harvested for histological evaluation for organ toxicity. Blood concentrations of the lead candidates can be collected after a IV dose, 100 t uL blood can be taken from the saphenous vein of two mice per time point at end of dosage; 0, 0.5, 1, 2, 4, 8, and 24 after the end of IV dosage. The drug levels ca be measured in the plasma and the specified organs as aforementioned above using LC-MS. Organs can be histologically graded for toxicity.

[00133] LC-MS Assays: A validated LC-MS assay is used to determine the intact caged compound, free decaged active moiety, and the uncoupled caged carrier molecule. Multiple reaction monitoring (MRM) signatures can be specifically determined for the cage and decaged compounds, permitting the development of multiplex assay where all of the a aiytes can be determined simultaneously. The assays are validated for culture medium a d plasma samples. Samples are extracted using a using liquid-liquid extraction to remove protein, where the insoluble proteins can be separated by ceiitrifugation. The supernatant ca be evaporated to dryness, and reconstituted with 50 μΐ, of running buffer. Separation of the anaiytes can be performed using C18 reverse phase columns, and the anaiytes can be quantified using the specific MRMs for each of the analytes. -x¾rnp¾e 3: Human Cachu er Tests To Demonstrate Feasibility of Local Release Oi

[00134] Human cadavers are the biological environment used to identify the release of caged fluorescent and drug compounds. In this particular example, human heads cadavers are used. The results of these experiments are analogous to live patient treatment from traumatic brain energy. FIG. 3 depicts a cascade of events upon primary injury, which includes neuroinflammation. Long wavelength visible and NIR radiation can penetrate deeper into tissue than light of shorter wavelength, and by treating at the site of neuroinflammation, secondary brain injury is diminished, FIG.j4A shows that the penetration is significant (left) and highly impactful in the case of TBI (right). In particular, near-infrared radiation NIR at 50μ ¥/οπι ;! penetrates 8cm into the brain. Sunlight has IQmW/crr in the NIR range. The quantum energy available at this wavelength within a safe thermal budget is nominal.

[00135] NIR emitters and detectors are placed non-invasively across the human skull to detect the transmission of the selected wavelengths and adjust the energy of the LED emitters appropriately as depicted in FIG. 4B. Transmission can be tested across at least 16 separate points using either moveable single LED transmitters or a grid of programmable and reconfigurable 16 LED transmitters (LED portals can be placed in a ''balaclava" like head piece). This is expected to provide a map over the thinner temporal and thicker occipital and frontal parts of the skull. In another test, standard neurosurgical procedures are used to implant deep brain stimulating electrodes to introduce a custom optical-fluidic probes. After a straight-line coronal incision is created at Kocher's point measuring approximately 2 cm anterior to the coronal suture and at the midpupillary line a single burr hole can be created. The aperture can then be augmented using a saw blade to create a craniotomy approximately 3 cm in diameter. The dura can be fenestrated in a cruxiate fashion and the blunt-tipped cannuiated introducer can be advanced certain fixed distances as confirmed by centimeter markings on the periphery of the cannula and both distance and general location confirmed by intraoperative ultrasound guidance. The stylette can be removed from the cannula and the optica 1-fiuidic brain probe introduced. The probe can be placed at varying depths and al low us to generate a penetration-depth map for the extracranial LED source (see details under methodology of LED systems). Both cadaver tests are performed with caged compounds of choice, but in particular caged-fluorescein and caged-drug (etomidate and eyelosporme) are described here. After a craniotomy made in the fashion described above, cryoprobe can be applied to the brain tissue to create a localized area of blood-brain damage.

[001361 To evaluate the level of diffusion and accumulation in the brain tissues, both caged or decaged compounds can be given as a parenteral infusion into warmed heparinized blood at a rate of 75 cc/hr. The level of accumulation and decagirsg can be determined using two methods, 1) photonic decaging of fluorescein from the cage compound and 2) quantification of caged drugs and photonically decaged compounds.

[00137] Specifically, the caged-eompounds (e.g. fluorescein or drug coupling) can be injected in warmed heparinized blood (98.6 C F) via the carotids (infusion can be maintained at 75 cc/hour). The infused cage-compound system is then activated at site of injur} ' using an extracranial LED system. The following treatment groups and controls can be employed; 1) cryotherapy with NIR illumination (treatment group) and the three controls 2) cryotherapy without N IR illumination, 2) no cryotherapy and IR illumination, and 3) no cryotherapy and no NIR illumination. Ail four of these can be conducted in same cadaver head so as to reduce the number of cadavers needed.

[00138] The level of fluorescein liberated is determined by measuring the amount of fluorescence found in the tissue. Fluorescent images (with zooming features turned off) can be taken under a fluorescent lamp. The level of fluorescence can be quantified in the images using ImageJ software using both intensity and area of fluorescence detection. To confirm the level of fluorescence, the fluorescein marker can be collected from injured brain tissue, and extracted using methanol. Following centrifugation, the level of fluorescence in the supernatant can be measured. The various treatments can be compared to determine the level of decaging.

[00139] In the caged drug studies, the concentration can be determined using a multiple LCMS. The level of liberated active drug is expected to quantify the amount of nanocage and decaged compounds simultaneously. To determine whether the impact of therapy through data collected for cadaver experiments, cadavers can be histologically evaluated and compared: 1) normal brain, 2) cryotherapy -†- illuminated, 3) cryotherapy - illuminated, and 4) normal area + illumination.

Example 4: Efficacy and Dosage Optimization Study Using TBI Animal Models

[00140] In this example, two animals models are described (CCI in rats and CCI plus hemorrhage in mice) to determine the efficacy and dosage of caged compounds for traumatic brain injur}'. Dosimetry can be determined for the rodent models to achieve similar activation parameters at the level of injury in pilot studies. The drug and NIR dosimetry can then be optimized, again in pilot studies in mice. For the in vivo work, the CCI mouse model can be used and the groups for testing can include 1) sham, 2) TBI plus vehicle, 3) TBI plus drug, 4) TBI plus drug plus IR light, and 5) TBI plus IR light alone. Rats and mice can be followed over a one-month period after TBI undergo both comprehensive motor and cognitive outcome evaluations along with neuropathology. The primary outcome parameter can be cognitive outcome (latency to find the hidden platform in the Morris water maze paradigm). Selected dmgs can be tested in the rat CCI model. An overview is shown in the FIG, SB. For each study, the groups for testing can include all of the appropriate controls, namely, 1) sham, 2) TBI plus vehicle, 3) TBI plus drug, 4) TBI plus drug plus IR light, and 5) TBI plus IR light alone. In the long-term outcome studies, rats and mice can be followed for 1 -mo after TBI and can undergo comprehensive motor and cognitive testing along with neuropathology. The primary outcome parameter is expected to be cognitive outcome (latency to find the hidden platform in the Morris water maze paradigm).

[00141] Beam Balance: Vestibulomotor function can be assessed in mice using a beam- balance task, wire grip, and a Morris water maze (MWM: The mouse MWM consists of a white pool (83 cm diameter, 60 cm deep) filled with water to 29 cm and goal platform (clear plastic, 10 cm diameter) is positioned 1 cm below the water's surface -15 cm from the southwest wall. The rat MWM employs a 180 cm in diameter and 60 cm high pool filled with water (28 cm depth), A platform 10 cm in diameter and 26 cm high (2 cm befow the water's surface) can he used as the hidden goal. A video camera above the pool records swim movement (PC-based tracking). Motor performance during cognitive tests can also be assessed by measuring swim speed in MWM testing to ensure thai cognitive performance is not confounded by motor deficits.

[00142] Spatial Learning 'Test: The hidden platform task assesses ability to learn spatial relations between cues and the escape platform (4 daily trials over 7d). The last 2d consist of a visible platform task- to control for non-specific visual or motor deficits. A probe trial is also measured. Ail testing can be conducted under blinded conditions. Data analysis for functional testing is expected to be carried out using two-way ANOVA for repeated measures.

[00143] Neuropathology (CCI and CCI+HS): At the designated time, mice can be anesthetized with isoflurane and perfused (10% buffered formalin). The brain can be post-fixed and cryoprotected. Coronal sections can be cut (1.0 mm). Brain sections can be stained with H&E. The lesion areas and non-injured hemispheres are determined in each section by a blinded observer by image analysis. Lesion and hemispheric volumes are calculated. Contusion volume is assessed and expressed in mm3 in the injured hemisphere, and as a % of non-injured hemisphere. Cel ls in CA1 and CA3 regions is expected to be quantified by stereology in an unbiased manner (Stereolnvestigator software 7.50.1 , MicroBrightField).

Skis II

[00144] A method to fabricate the above described wearable array can be divided into three steps:

[00145] Step 1 : Initial prototype: the layout of the grid and properties of the optical devices (LEDs, lenses and detectors) are determined using 3D electromagnetic (EM) simulations and results from the testing procedure described in Example 3. The constraints are expected to be the maximum and minimum optical intensity to achieve close-tQ-uniform and safe illumination. The electronics and integrated circuits are expected be specified for a low-power addressable array. As a first solution the array can be divided into a number of sub-arrays. These small sub-arrays can have fiat surfaces, but are expected to be connected over a flexible membrane to create a curved shape approximating the curvature of human and/or animal head.

[00146] Step 2: Revised prototype aiming to: a) insure reliability, b) reduce power and weight, c) optimize the devices based on measurements of initial prototype.

[00147] Step 3: Final product design: based on steps 1 and 2, design an ergonomic, curve fitting, wearable system using technologies such as fabric of optical fibers, integrated with photonic and electronic devices. This step can be performed according to some embodiments, to also use input from the end users in the field in order to make it portable and rugged.

[00148] As mentioned in step 1 above, EM simulations can be used to determine the layout of the grid and properties of optical devices. FIG._10 and FIG. 11 show preliminary results from 2D EM simulations. As mentioned above, the parameters that can be changed and/or optimized are: number of LEDs, power per LED, the distance of the array from the skul l, separation between LEDs and LED/lens viewing half angle. There are trade-offs between these parameters and level of uniformity (inside the brain), power consumption and weight of the system. As shown in FIG, 1 J , in the absence of a lensing system, the array should be placed about 3 cm far from the skull to achieve good uniformity inside the brain. This distance can be used if a lensing system is added to the design.

[00149] In summary, in embodiments described herein compounds are described that are capable of being "caged", chemically and/or physically connected to a long wavelength absorber, a moiety capable of absorbing a wavelength greater than or equal to 750 am. Caged compounds are then capable of being decaged, where the decaged molecules are capable of participating in biological and/or chemical reaction independent of the long wavelength absorber. Decaging is accomplished by excitation of the long wavelength absorber [00150] The entire disclosure of each document cited (including patents, patent applications, journal articles including related supplemental and/or supporting information sections, abstracts, laboratory manuals, books, or other disclosures) in the Background, Summary, Detailed Description, and Examples is hereby incorporated herein by reference. All references cited in this disclosure are incorporated by reference to the same extent as if each reference had been incorporated by reference in its entirety individually. However, if any inconsistency arises between a cited reference and the present disclosure, the present disclosure takes precedence.

[00151] The terms and expressions which have been employed herein are used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions {hereof but it is recognized that various modifications are possible within the scope of the disclosure claimed. Thus, it should be understood that although the disclosure has been specifically disclosed by preferred embodiments, exemplary embodiments and optional features, modification and variation of the concepts herein disclosed can be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this disclosure as defined by the appended claims.

[00152| It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. As used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the content clearly dictates otherwise. The term "plurality" includes two or more referents unless the content clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosure pertains.

[00153] The term "alky!" as used herein refers to a linear, branched, or cyclic saturated hydrocarbon group typically although not necessarily containing 1 to about 15 carbon atoms, or 1 to about 6 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, oct l, decyl, and the like, as well as cycloalkyl groups such as cyclopeiityi, cycloliexyl and the like. Generally, although again not necessarily, alky- groups herein contain 1 to about 15 carbon atoms. The term "cycloalkyl" intends a cyclic alkyl group, typically having 4 to 8, or 5 to 7, carbon atoms. The term "substituted alkyl" refers to alkyl substituted with one or more substituent groups, and the terms "heteroatom-containing alkyl" and "heteroalkyl" refer to alkyl in which at least one carbon atom is replaced with a heteroatom. If not otherwise indicated, the terms "alkyl" and "lower alkyl" include linear, branched, cyclic, unsubstituted, substituted, and/or heteroatom-containing alkyl and lower alkyl, respectively.

[00154] The term "hydrocarbyl" as used herein refers to any univalent radical, derived from a hydrocarbon, such as, for example, methyl or phenyl. The term "hydrocarbylene" refers to divalent groups formed by removing two hydrogen atoms from a hydrocarbon, the tree valencies of which .may or may not be engaged in a double bond, typically but not necessarily containing 1 to 20 carbon atoms, in particular 1 to 12 carbon atoms and more particularly I to 6 carbon atoms which includes but is not limited to linear cyclic, branched, saturated and unsaturated species, such as alkylene, alkenyiene alkynylene and divalent aryi groups, e.g., 1,3-phenylene, -CH 2 CH 2 CH?-propane-l,3-diyi, -C¾- methylene, -CH-CH-CH-CH-. The term "hydrocarbyl" as used herein refers to univalent groups formed by removing a hydrogen atom from a hydrocarbon, typically but not necessarily containing I to 20 carbon atoms, in particular 1 to 12 carbon atoms and more particularly 1 to 6 carbon atoms, including but not limited to linear cyclic, branched, saturated and unsaturated species, such as univalent alkyl, alkenyl, alkynyl and aryl groups e.g. ethyl and phenyl groups.

[001551 The term "heteroatom-containing" as in a "heteroatom-containing alky group" refers to a alkyl group in which one or more carbon atoms is replaced with an atom other than carbon, e.g., nitrogen, oxygen, sulfur, phosphorus or silicon, typically nitrogen, oxygen or sulfur. Similarly, the term "heteroalkyl" refers to an alkyl substituent that is heteroatom-containing, the term "heterocyclic" refers to a cyclic substituent that is heteroatom-containing, the terms "heteroaryl" and "heteroaromatic" respectively refer to "aryl" and "aromatic" substituents that are heteroatom-containing, and the like. It should be noted that a "heterocyclic" group or compound may or may not be aromatic, and further that "heterocycles" may be monocyclic, bicyclic, or polycyclic as described above with respect to the term "aryi." Examples of heteroalkyl. groups include aikoxyaryl, alk lsulfanyl-substituted alkyl, N-alkylated amino alkyl, and the like. Examples of heteroaryl substitiients include pyrrolyl, pyrrolidmyl, pyridinyl, quinolinyl, mdolyl, pyrimidinyl, imidazolyl, 1 ,2,4-triazolyl, tetrazolyl, and additional groups identifiable by a skilled person., and examples of heteroatom-contaming alicyclic groups are pyrrolidmo, morphoiino, piperazino, piperidino, and additional substitiients identifiable by a skilled person.

[00156] The term "alkoxy" as used herein intends an alkyl group bound through a single, terminal ether linkage; that is, an "alkoxy" group may be represented as -O-aikyl where alkyl is as defined above. A "lower alkoxy" group intends an alkoxy group containing 1 to 6 carbon atoms. Analogously, "alkenyloxy" and "lower alkenyloxy" respectively refer to an alkenyl and lower alkenyl group bound through a single, terminal ether linkage, and "alkynyloxy" and "lower alkynyloxy" respectively refer to an aikynyl and lower aikynyl group bound through a single, terminal ether linkage.

[00157] The term "alkylamino" as used herein intends an alkyl group bound through a single terminal amine linkage; that is, an "alkylamino" may be represented as -NH-alkyl where alkyl is as defined above. A "lower alkylamino" intends a alkylamino group containing 1 to 6 carbon atoms. The term "dialkylamino" as used herein intends two identical or different bound through a common amine linkage; that is, a "dialkylamino" may be represented as -N(alkyl)2 where alkyl is as defined above. A "lower dialkylamino" intends a alkylamino wherein each alkyl group contains 1 to 6 carbon atoms. Analogously, "alkenylamino", "lower aikenyiamino", "aikynylamino", and "lower aikynylamino" respectively refer to an alkenyl, lower alkenyl, aikynyl and lower aikynyl bound through a single terminal amine linkage; and "diafkenylamino", "lower dialkenylamino" , "dialkyiiyiamino", "lower diaikynylamino" respectively refer to two identical alkenyl, lower alkenyl, uikynyl and lower aikynyl bound through a common amine linkage. Similarly, "alkenylalkyiiyiamino", "alkenylalkylamino", and "aikynyl alkylamino" respectively refer to alkenyl and aikynyl, alkenyl and alkyl, and aikynyl and alkyl groups bound through a common amine linkage. [00158] The term "aryl" as used herein, and unless otherwise specified, refers to an aromatic substituent containing a single aromatic ring or multiple aromatic rings that are fused together, directly linked, or indirectly linked (such that the different aromatic rings are bound to a common group such as a methylene or ethylene moiety), Aryl groups can contain 5 to 24 carbon atoms, or aryl groups contain 5 to 14 carbon atoms. Exemplary aryl groups contain one aromatic ring or two fused or linked aromatic rings, e.g., phenyl, naphthyl, hi henyl, dipheny {ether, diphenylamine, benzophenone, and the like. "Substituted aryl" refers to an aryl moiety substituted with one or more substituent groups, and the terms "beteroatom-contaming aryl" and "heteroaryl." refer to aryl substituents in which at least one carbon atom is replaced with a heteroatom, as will be described in further detail infra,

[00159] The term "arene", as used herein, refers to an aromatic ring or multiple aromatic rings that are fused together. Exemplary arenes include, for example, benzene, naphthalene, anthracene, and the like. The term "heteroarene", as used herein, refers to an arene in which one or more of the carbon atoms has been replaced by a heteroatom (e.g. O, N, or S). Exemplary heteroarenes include, for example, indole, benzimidazoie, thiophene, benzthiazole, and the like. The terms "substituted arene" and "substituted heteroarene", as used herein, refer to arene and heteroarene molecules in which one or more of the carbons and/or heteroatoms are substituted with substituent groups.

[00160] The terms "cyclic", "cyclo-", and "ring" refer to alicyclic or aromatic groups that may or may not be substituted and/or heteroatom containing, and that may be monocyclic, bicyciic, or polycyclic. The term "alicyclic" is used in the conventional sense to refer to an aliphatic cyclic moiety, as opposed to an aromatic cyclic moiety, and may be monocyclic, bicyciic or polycyclic.

[00161] The terms "halo", "halogen", and "haiide" are used in the conventional sense to refer to a chloro, bromo, fluoro or iodo substituent or Sigand.

[00162] The term "substituted" as in "substituted alkyl," "substituted aryl," and the like, is meant that in the, alkyl, aryl, or other moiety, at least one hydrogen atom bound to a carbon (or other) atom is replaced with one or more non-hydrogen substituents. [00163] Examples of such substituents include, without limitation: functional groups such as halo, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5-C24 aryloxy, C6-C24 aralkyloxy, C6-C24 alkaryloxy, acyl (including C2-C24 alkylcarbonyl (-CO-alkyl) and C6-C24 arylearbonyl (-CO-aryl)), acyloxy (-O-acyl, including C2-C24 alkylcarbonyloxy (-O-CO-alkyI) and C6-C24 arylcarbonyloxy (-0- CO-aryl)), C2-C24 alkoxy carbonyl (-(CO)-O-alkyl), C6-C24 aryloxycarbonyl (-(CO)-O- aryl), halocarbonyl (-CO)-X where X is halo), C2-C24 alkylcarbonato (-O-(CO)-O- alkyl), C6-C24 arylcarbonato (-O-(CO)-O-aryl), carboxy (-COOH), carboxylato ( COO " ), carbamoyl (-(CO)-NH2), mono-(Cl-C24 alkyl)-substituted carbamoyl i-(CO)-NH(C! ~ C24 alkyl)), di-(Cl-C24 alkyl)-substituted carbamoyl (-(C0)-N(C1-C24 alky 1)2), mono- (C5-C24 aryi)-substituted carbamoyl (-(CO)-NH-aryf), di-(C5-C24 aryl)-substituted carbamoyl (-(CO)-N(C5-C24 aryl)2), di-\--{( 1 -< " 24 aikyi),N~(C5-C24 aryl)-substituted carbamoyl, thiocarbamoyl (-(CS)-NH2), mono-(C3 -C24 alkyl)-substituted thiocarbamoyl (-(C0)-NH(C1-C24 alkyl)), di-(Cl -C24 alkyl Ksubstituted thiocarbamoyl {-ί <Ό)-\ί< ! - C24 alkyl)2), mono-(C5-C24 aryl)-substituted thiocarbamoyl (-(CO)-NH-aryl), di-(C5- C24 aryl)-substituted thiocarbamoyl (-(CO)-N(C5-C24 aryl)2), di-N-(Cl -C24 alkyl),N- (C5-C24 aryl)-substituted thiocarbamoyl, carbamido (-NH-(CO)-NH2), cyano(-C≡N), cyanato ί~ϋ-ί X ). thiocyanato (-S-i X ). formyl (-(CO)-H), thioformyl ( (( S -i i h amino (-NH2), mono-(Cl-C24 alkyl)-substituted amino, di-(Cl-C24 alkyl)-substituted amino, mono-(C5-C24 aryl)-substituted amino, di-(C5-C24 aryl)-s ' ubstituted amino, C2- C24 alkylamido (-NH-(CO)-alkyl), C6-C24 arylamido (-NH-(CO)-aryl), imino (-CR=NH where R - hydrogen, C1-C24 alkyl, C5-C24 aryi, C6-C24 alkaryl, C6-C24 aralkyl, and additional imino identifiable by a skilled person.), C2-C20 alkylimino ( CR= (alkyl), where R hydrogen, C1-C24 alkyl, C5-C24 and, C6-C24 alkaryl, C6-C24 aralkyl, and additional alkylimino identifiable by a skilled person.), aryl imino (-CR : =N(aryl), where R = hydrogen, C1-C20 alkyl, C5-C24 aryl, C6-C24 alkaryl, C6-C24 aralkyl, and additional arylimino identifiable by a skil led person.), nitro (-N02), nitroso (-NO), sulfo (-S02- OH), sulfonato (-S02-0 " ), C1-C24 alkylsulfanyl (-S-alkyl; also termed "alkylthio"), C5- C24 arylsulfanyl (-S-aryl; also termed "arylthio"), C1-C24 alkylsulfinyi (-(SO)-alkyl), C5-C24 arylsuifmyl (-(SO)-aryl), C1-C24 aikyisulfonyl (-S02-alkyl), C5-C24 arylsulfonyl (-S02-aryl), boryl (-BH2), borono (-B(OH)2), boronato (~B(OR)2 where R is alkyl or other hydrocarbyl), phosphono (-P(0)(OH)2). phosphonato (-P(0)(0 ~ )2), phosphinato (~P(0)(Q ~ )), phospho (-P02), phosphino (-FH2), silyl (-SiR3 wherein R is hydrogen or hydrocarbyl), and silyloxy (-O-silyl); and the hydrocarbyl moieties Cl- C24 alkyl (e.g. C1 -C12 alkyl and C1 -C6 alkyl), C2-C24 alkenyl (e.g. C2-C12 alkenyl and C2-C6 alkenyl), C2-C24 alkynyi (e.g. C2-C12 aikynyl and C2-C6 aikynyl), C5-C24 aryl (e.g. C5-C14 aryl), C6-C24 alkaryi (e.g. C6-C16 alkaryi), and C6-C24 aralkyl (e.g. C6- C16 aralkyl).

[00164| The term "acyi" refers to substituents having the formula -(CO)-alkyl, -(CO)- aryl, or -(CO)-aralkyl, and the term "acyloxy" refers to substituents having the formula - 0(C())-aikyi, ~0(CO)~aryl, or -0(CO)-aralkyl, wherein "alkyl," "aryl, and "aralkyl" are as defined above.

[001 5] The term "alkaryi" refers to an aryl group with an alkyl substituent, and the term "aralkyl" refers to an alkyl group with an aryl substituent, wherein "aryl" and "alkyl" are as defined above. In some embodiments, alkaryi and aralkyl groups contain 6 to 24 carbon atoms, and particularly alkaryi and aralkyl groups contain 6 to 16 carbo atoms. Alkaryi groups include, for example, p-methylphenyl, 2,4-dimethy!phenyl, p- cyclohexylphenyl, 2,7-dimethylnaphthyl, 7-eyclooctylnaphthyl, 3-ethyl-cyclopenta- 1 ,4- diene, and the like. Examples of aralkyl groups include, without limitation, benzyl, 2- phenyl-ethyl, 3 -phenyl-propyl, 4-phenyl-butyl, 5-phenyl-pentyl, 4-phenylcyclohexyl, 4- benzylcyclohexyl, 4-phenylcyc!ohexylmethyl, 44ienzylcyclohex)4methyi, and the like. The terms "alkaryloxy" and "aralkyloxy" refer to substituents of the formula -OR wherein R is alkaryi or aralkyl, respectively, as just defined.

[00166] When a Markush group or other grouping is used herein, all individual members of the group and all combinations and possible subcombinations of the group are intended to be individually included in the disclosure. Every combination of components or materials described or exemplified herein can be used to practice the disclosure, unless otherwise stated. One of ordinary skill in the art will appreciate that methods, device elements, and materials other than those specifically exemplified can be employed in the practice of the disclosure without resort to undue experimentation. All art-know r n functional equivalents, of any such methods, device elements, and materials are intended to be included in this disclosure. Whenever a range is given in the specification, for example, a temperature range, a frequency range, a time range, or a composition range, all intermediate ranges and all subranges, as well as, all individual values included in the ranges given are intended to he included in the disclosure. Any one or more individual members of a range or group disclosed herein can be excluded from a claim of this disclosure. The disclosure illustratively described herein suitably can be practiced in the absence of any element or elements, limitation or limitations, which is not specifically disclosed herein.

[00167] "Optional" or "optionally" means that the subsequently described circumstance may or may not occur, so that the description includes instances where the circumstance occurs and instances where it does not according to the guidance provided in the present disclosure. For example, the phrase "optionally substituted" means that a non-hydrogen substituent may or may not be present on a given atom, and, thus, the description includes structures wherein a non-hydrogen substituent is present and structures wherein a non- hydrogen substituent is not present. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or un substituted." Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned can be identified in view of the desired features of the compound in view of the present disclosure, and in view of the features that result in the formation of stable or chemically feasible compounds. The term "stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recover} ' , purification, and use for one or more of the puiposes disclosed herein.

[00168] A number of embodiments of the disclosure have been described. The specific embodiments provided herein are examples of useful embodiments of the disclosure and it will be apparent to one skilled in the art that the disclosure can be carried out using a large number of variations of the devices, device components, methods steps set forth in the present description. As will be obvious to one of skill in the art, methods and devices useful for the present methods can include a large number of optional composition and processing elements and steps.

[00169] In particular, it will be understood that various modifications may be made without departing from the spirit and scope of the present disclosure. Accordingly, other embodiments are within the scope of the following claims.

REFERENCES

1. Corrigan JD, Selassie AW, Orman JA. The epidemiology of traumatic brain injury. J Head Trauma Rehabi!. 201 Q;25(2): 72-80.

2. Sauaia A, Moore FA, Moore EE, Moser S, Brennan R, Read RA, Pons PT. Epidemiology of trauma deaths: a reassessment. J Trauma. 1995; 38(2): 185-93.

3. Narayan R , Michel ME, Ansel! B, et al. Clinical trials in head injury. J Neurotrauma. 2002 ; 19(5) : 503 -57.

4. Granacher RP, Traumatic Brain Injury: Methods for Clinical & Forensic Neuropsychiatric Assessment, 2nd Ed. Boca Raton: CRC. pp. 26-32. ISBN 0-8493-8138- X

5. Giza CC, Hovda DA. The neurometabolic cascade of concussion. J of Athletic Training 2001 ;36(3):228-235

6. Martland HS. Punch Drunk, JAMA 1928:91 : 1103-7

7. Plassman BL, Havlik RJ; Steffens DC, et al. Documented head injury in early- adulthood and risk of Alzheimer's disease and other dementias. Neurology 2000;55(8): 1158-1166

8. Lye TC and Shores EA. Traumatic brain injury as a risk factor for Alzheimer's disease: A review. Neuropsych Rev 2000;10: 115-129

9. Garga , Lowen stein DH. Posttraumatic epilepsy: a major problem in desperate need of major advances. Epilepsy Curr. 2006;6(l): l-5.

10. Bower JH, Maraganore DM, Peterson BJ, et al. Head trauma preceding PD; a case-control study. Neurology. 2003;6Q(10): 161G-5.

11. Kochanek PM, Carney N, Adelson PD, et al. Guidelines for the acute medical management of severe traumatic brain injury in infants, children, and adolescents— second edition. Pediatr Crit Care Med. 2012; ! 3 Suppl 1 :S 1~82.

12. Colin BF, Rejger V, Hagenouw-Taal JC, Voormolen JH. Results of a feasibility trial to achieve total immobilization of patients in a neurosurgical intensive care unit wi th ctomidate. Anaesthesia. 1983;38 Suppi:47-50.

13. Bergen JM, Smith DC. A review of etomidate for rapid sequence intubation in the emergency department. J Emerg Med 15 :221 -30, 1997.

14. Adnet F, Minadeo JP, Finot MA, Borron SW, Fauconnier V, Lapandry C, Baud FJ. A survey of sedation protocols used for emergency endotracheal intubation in poisoned patients in the French prehospital medical system. Eur J Emerg Med 5:415-9, 1988

15. Murugaiah KD, Hemmings HC Jr. Effects of intravenous general anesthetics on [3HJGABA. release from rat cortical synaptosomes. Anesthesiology 89:919-28, 1998

16. Watson JC, Drummond JC, Patei PM, Sano T, Akrawi W, U HS. An assessment of the cerebral protective effects of etomidate in a model of incomplete forebrain ischemia in the rat. Neurosurgery 30:540-4, 1992

17. Patei PM, Goskowicz RL, Drummond JC, Cole DJ. Etomidate reduces ischemia- induced gmtamate release in the hippocampus in rats subjected to incomplete forebrain ischemia. Anesth Anaig 80:933-9, 1995

18. Albensi BC, Sullivan PC, Thompson MB, Scheff SW, Mattsors MP: Cyclosporin ameliorates traumatic brain-injury induced alterations of hippoeampal synaptic plasticity. Exp Neurol 162:385-389, 2000

19. Hatton J et al. Dosing and safety of cyclosporin in patients with severe brain injury J Neurosurgery 2008; 109: 699-707.

20. G. Grasso, " Neuroprotective effect of recombinant human erythropoietin in experimental subarachnoid hemorrhage," Journal of Neurosurgical Sciences, vol. 45, no. l , pp. 7-14, 2001 .

21. E. Morishita, S. Masuda, M, Nagao, Y. Yasuda, and R. Sasaki, "Erythropoetin receptor is expressed in rat hippoeampal and cerebral cortical neurons, and erythropoietin prevents in vitro giutamate-induced neuronal death," Neuroscience, vol. 76, no. 1 , pp. 105-1 16, 1996. 22. M. Bernaudin, H. H. Marti, S, Roussei, et al., "A potential role for erythropoietin in focal permanent cerebral ischemia in mice," Journal of Cerebral. Blood Flow and Metabolism, vol. 19, no. 6, pp. 643-651, 1999.

23. C. Alafaci, F. Salpiet.ro, G. Grasso, et al., "Effect of recombinant human erythropoietin on cerebral ischemia following experimental subarachnoid hemorrhage," European Journal of Pharmacology, vol. 406, no. 2, pp. 2.19—225, 2000.

24. H. H. Marti, M. Gassmann, R. H. Wenger, et al., "Detection of erythropoietin in human liquor: intri sic erythropoietin production in the brain," Kidney International, vol. 51, no. 2, pp. 4.16-418, 1997.

25. S. E. Juul, S. A. Stallings, and R. D. Christenseii, "Erythropoietin in the cerebrospinal fluid of neonates who sustained C S injur}?," Pediatric Research, vol. 46, no. 5, pp. 543-547, 1999.

26. DeWitt DS, Prough DS. Blast-induced brain injury and posttraumatic hypotension and hypoxemia. Neurotrauma 26:877-87, 2009.

27. Hovda DA, Lee SM, Smith ML, Von Stuck S, Bergsneider M, Kelly D, Shalmon E, Martin N, Caron M, Mazziotta J, et al. The neurochemical and metabolic cascade following brain injury: moving from animal models to man, J Neurotrauma 1995;12:903-

6.

28. Readnower RD, Chavko M, Adeeb S, et al. I crease in blood-brain barrier permeability, oxidative stress, and activated microglia in a rat model of blast-induced traumatic brain injury. J Neurosci Res. 2010;88:3530-9

29. Garman RH, Jenkins LW, Switzer RC, et al: Blast exposure injury in rats with body protection is characterized primarily by diffuse axonal injury. J Neurotrauma 201 1 ;28:947-959

30. Shear DA, Lu XC, Pedersen R, Wei G, Chen Z, Davis A, Yao C, Dave J, Torteila FC.Severitv'' profile of penetrating ballistic-like brain injury on neurofunctional outcome, blood-brain barrier permeability, and brain edema formation. J Neurotrauma. 2011; 28:2185-95.

31. Marion DW, Curley KC, Schwab K, Hicks RR. mTB Diagnostics Workgroup. Proceedings of the military mTBI Diagnostics Workshop, St. Pete Beach, J N eurotrauma. 2011 A.pr;28{4):517-26 32. Management of Concussion mTBI Working Group. VA'DoD Clinical Practice Guideline for Management of Concussion/Mild Traumatic Brain Injury. J Rebabil Res Dev 2009;46(6):CPl-68

33. Finkelsiein E, Corso P, Miller T, et al: The Incidence and Economic Burden of Injuries in the United States. New York (NY): Oxford University Press; 2006

34. Coronado, McGuire, Paul, Sugerman, Pearson. The Epidemiology and Prevention of TB I 2012

35. Taylor BC, Hagel EM, Carlson KF, Cifu DX, Cutting A, Bideispach DE, Sayer NA. Prevalence and costs of co-occurring traumatic brain injury with and without psychiatric disturbance and pain among Afghanistan and Iraq War Veteran V.A, users. Med Care. 2012; 50(4):342-6.

36. National Institute of Neurological Disorders and Stroke (1989, Feb) Interagency Head injury Task Force Report. Bethesda, MD

37. Anslyn EV & Dougherty DA (2006) Modern physical organic chemistry (University Science, Sausalito, CA.) pp xxviii, 1095 p

38. Turro NJ, Ramamurihy V, & Scaiano JC (2010) Modern Molecular Photochemistry of Organic Molecules (University Science Books, Sausalito, CA) p 1084

39. The Molecular Probes® Handbook A Guide to Fluorescent Probes and Labeling

Technologies

40. Jobsis, Frans F., Noninvasive, Infrared Monitoring of Cerebral and Myocardial Oxygen Sufficiency and Circulatory, Science , Vol. 198, No. 4323 (Dec. 23, 1977), pp. 1264-1267

41. IEC 60601-2-33 Requirements for the Safety of MR Equipment for Medical Diagnostics

42. Sinz EH, ochanek PM, Dixon CE, et al: Inducible nitric oxide synthase is an endogenous neuroprotectant after traumatic brain injury in rats and mice. J Clin Invest 104:647-656, 1999

43. Tehranian R, Rose ME, Vagni V, et al: Transgenic mice that overexpress the anti-apoptotie Bcl-2 protein have improved histological outcome but unchanged behavioral outcome after traumatic brain injury. Brain Res 26: 1 101 : 126-135, 2006

44. Shellington DK, Wu X, Exo J, et al; Acute volume-limited fluid resuscitation with polynitroxylated pegyiated hemoglobin attenuates neuronal death after combined traumatic brain injury and hemorrhagic hypotension in mice. Crit Care Med 39:494-505, 2011

45. Williams C, Mehrian Shai R, Wu Y, Hsu YH, Sitzer T, Spann B, McCieary C, Mo Y, Miller CA. Transcriptome analysis of synaptoneurosom.es identifies neuroplasticity genes overexpressed in incipient Alzheimer's disease. PLoS One. 2009;4(3):e4936

46. Pratap M, Oisen RW, Otis TS, Waihier M. 2009. Etomidate, propofoi and the neurosteroid THDOC increase the GABA efficacy of recombinant α4β3δ and α4β3 GABAA receptors expressed in HE cells. Neuropharmacology. 56:155-160

47. Meera P, Wallner M, Song M, Toro L. 1997. Large conductance voltage- and calcium-dependent K+ channel, a distinct member of voltage-dependent ion channels with seven N-teraiinal transmembrane segments (S0-S6), an extracellular N terminus, and an intracellular (S9-S 10) C terminus. Proc Natl Acad Sci U S A 94: 14066- 14071

48. Mordwinkin NM, Meeks CJ, Jadhav SS, Espinoza T, Roda N, Dizerega GS, Louie SG, Rodgers KE. Angiotensin-(l-7) administration reduces oxidative stress in diabetic bone marrow. Endocrinology. 2012 May; 153(5):2189-97.

49. Bayir H, Kagan VE, Borisenko GG, Tyurina YY, Janesko L, Vagni VA, Billiar TR, Williams DL, Kochanek PM. Enhanced oxidative stress in iNOS-deficient mice after traumatic brain injury: support for a neuroprotective role of iNOS. J Cereb Blood Flow Metab 25:673-684, 2005

50. Whalen MJ, Carlos TM, Dixon CE, Wisniewski SR. Schiding JK, Clark RSB, Baum E, Marion DW, Kochanek PM: Effect of Traumatic Brain Injury in Mice Deficient in Intercellular Adhesion Molecule- 1 : Assessment of Histopathologic and Functional Outcome. J eurotrauma .16:299-309, .1999

51. Tehranian R, Rose ME, Vagni V, Pickreil AM, Griffith RP, Liu H, Clark RSB, Dixon CE, Kochanek PM, Graham SH, Disruption of bax protein prevents neuronal cell death but produces cognitive impairment in mice following traumatic brain injury. J Neurotrauma 25:755-767, 2008

52. Haseikorn ML, Shellington D, Jackson EK, Vagni V, Janesko- Feldman K, Dubey RK, Gillespie DG, Cheng D, Bell MJ, Jenkins LW, Romanics G, Schnermann J, Koclianek PM: Adenosine Al receptor activation as a brake on microglial proliferation after experimental traumatic brain injur}? in mice. J Neurotrauma 27:901-10, 2010. PMC2943944

53. Foley LM, Kitchens TK, Melick J, Bayir H, Ho C, ochanek PM: Effect of inducible nitric oxide synthase on cerebral blood flow after experimental traumatic brain injury in mice. J Neurotrauma 25:299-310, 2008

54. Hendrich KS, Koclianek PM, Melick JA, Schiding JK, Statler KD, Williams DS, Marion DW, Ho C: Cerebral perfusion during anesthesia with fentanyl isofiurane or pentobarbital in normal rats studied by arterial spin-labeled M RL Magn Reson Med 46:202-206, 2001

55. Kochanek PM, Hendrich KS, Dixon CE, Schiding JK, Williams DS, Ho C: Cerebral blood flow at one year after controlled cortical impact in rats: assessment by magnetic resonance imaging. J Neurotrauma. 2002 Sep; 19(9): 1029-37

56. Blasiole B, Bayir H, Vagni V, Janesko-Feldman K, Wisniewski SR, Chiekhi A, Long J, Atkins J, Kochanek PM. 100% Oxygen is beneficial during resuscitation of experimental combined traumatic brain injury and hemorrhagic shock in mice. ATACCC Annual Meeting August 15-19, 2011

57. Manole M, Kochanek P, Foley L, Hitchens T, Bayir H, Alexander H, Garman R, Ma L, Hsia C, Clark R: Polynitroxil albumin and albumin therapy after pediatric asphyxia] cardiac arrest: Effects on cerebral blood flow and neurological outcome. J Cereb Blood Flow Metab 201 1 Nov 30. [Epub ahead of print]

58. Coronado VG, Thurman DJ, Greenspan Al, Weissman BM. Epidemiology. In: Jallo J, Loftus CM, cds. Neurotrauma and critical care: brain. New York, NY: Thieme; 2009:3-19.

59. Paul M, Xu L, Wald MM, Coronado V. Traumatic brain injur}' in the United States: emergency department visits, hospitalizations, and deaths, 2002—2006. Atlanta, GA: CDC 1 , National Center for Injur}' Prevention and Control; 2010

60. Turro NJ, Ramamurthy V and Scaiiio JC (2010) Modem Molecular Photochemistry of Organic Moleculse. Unveristy Science Books, Sausalito, CA.