BIOMARKERS FOR H-NOX DELIVERY OF OXYGEN

The invention provides methods to monitor tumor oxygenation by H-NOX proteins. H-NOX proteins extravasate into and preferentially accumulate in tumor tissue for sustained delivery of oxygen. For example, the invention provides methods to monitor brain tumor oxygenation by H-NOX proteins for enhanced treatment of brain cancers.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation-in-part of International Application No. PCT/US2013/020602, filed Jan. 7, 2013. This application also claims benefit of U.S. provisional patent application No. 61/898,395, filed on Oct. 31, 2013 and U.S. provisional patent application No. 61/907,983, filed Nov. 22, 2013. The contents of each are incorporated herein by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

This work was supported by Grant Nos. 1 R43 CA138006 and 2 R44 CA138006. The U.S. government has rights in any patent issuing on this application.

SUBMISSION OF SEQUENCE LISTING ON ASCII TEXT FILE

The content of the following submission on ASCII text file is incorporated herein by reference in its entirety: a computer readable form (CRF) of the Sequence Listing (file name: 627042000420SeqList.txt, date recorded: Jun. 12, 2015, size: 49 KB).

TECHNICAL FIELD

This application pertains to H-NOX proteins and methods of using them to deliver oxygen to hypoxic tumors and methods to monitor tumor oxygenation to enhance anti-tumor therapies. H-NOX proteins provide a new therapeutic tool for delivering O2 to humans and, for veterinary purposes, to animals.

BACKGROUND OF THE INVENTION

H-NOX proteins (named for Heme-Nitric oxide and OXygen binding domain) are members of a highly-conserved, well-characterized family of hemoproteins (Iyer, L M et al. (2003) BMC Genomics 4(1):5; Karow, D S et al. (2004) Biochemistry 43(31):10203-10211; Boon, E M et al. (2005) Nature Chem. Biol. 1:53-59; Boon, E M et al. (2005) Curr. Opin. Chem. Biol. 9(5):441-446; Boon, E M et al. (2005) J. Inorg. Biochem. 99(4):892-902; Cary, S P et al. (2005) Proc Natl Acad Sci USA 102(37):13064-9; Karow D S et al. (2005) Biochemistry 44(49):16266-74; Cary, S P et al. (2006) Trends Biochem Sci 31(4):231-9; Boon, E M et al. (2006) J Biol Chem 281(31):21892-902; Winger, J A et al. (2007) J Biol Chem. 282(2):897-907). H-NOX proteins are nitric-oxide-neutral, unlike previous hemoglobin-based oxygen carriers, H-NOX do not scavenge circulating nitric oxide, and thus are not associated with hypertensive or renal side effects. The intrinsic low NO reactivity (and high NO stability) makes wild-type and mutant H-NOX proteins desirable blood substitutes because of the lower probability of inactivation of H-NOX proteins by endogenous NO and the lower probability of scavenging of endogenous NO by H-NOX proteins. Importantly, the presence of a distal pocket tyrosine in some H-NOX proteins (Pellicena, P. et al. (2004) Proc Natl. Acad Sci USA 101(35):12854-12859) is suggestive of undesirable, high NO reactivity, contraindicating use as a blood substitute. For example, by analogy, a Mycobacterium tuberculosis hemoglobin protein, with a structurally analogous distal pocket tyrosine, reacts extremely rapidly with NO, and is used by the Mycobacterium to effectively scavenge and avoid defensive NO produced by an infected host (Ouellet, H. et al. (2002) Proc. Natl. Acad. Sci. USA 99(9):5902-5907). However, it was surprisingly discovered that H-NOX proteins actually have a much lower NO reactivity than that of hemoglobin making their use as blood substitutes possible.

It was discovered that H-NOX proteins that bind NO but not O2 can be converted to H-NOX proteins that bind both NO and O2 by the introduction of a single amino acid mutation (see WO 2007/139791 and WO 2007/139767). Thus, the affinity of H-NOX proteins for O2 and NO and the ability of H-NOX proteins to discriminate between O2 and NO ligands can be altered by the introduction of one or more amino acid mutations, allowing H-NOX proteins to be tailored to bind O2 or NO with desired affinities. Additional mutations can be introduced to further alter the affinity for O2 and/or NO. The H-NOX protein family can therefore be manipulated to exhibit improved or optimal kinetic and thermodynamic properties for O2 delivery. For example, mutant H-NOX proteins have been generated with altered dissociation constants and/or off rates for O2 binding that improve the usefulness of H-NOX proteins for a variety of clinical and industrial applications. The ability to tune H-NOX proteins to bind and deliver O2 is a therapeutic avenue that addresses and overcomes the central shortcomings of current O2 carriers.

What is needed for certain therapeutic uses is an H-NOX with a long circulation half-life that can bind and deliver O2 to distal tissues for sufficient periods of time. Additionally, H-NOX proteins extravasate into tumors where they accumulate at different rates. For example, polymeric H-NOX proteins are tuned to transport oxygen through normoxic regions of tumors and release oxygen deep within hypoxic zones within tumors. For brain tumors, the H-NOX protein may cross the blood-brain bather to deliver O2 to hypoxic brain tumors. This combination of features represents a significant advance in the use of oxygen carriers as modifiers of the hypoxic niches of tumors to increase the efficacy of radiotherapy, chemotherapy and other anti-cancer treatments reliant on oxygenation of tumor cells. Provided herein are methods to detect tumor oxygenation (e.g. brain tumor oxygenation) to enhance therapeutic treatments for cancers.

H-NOX proteins for delivery of O2 or NO are provided by U.S. Pat. Nos. 8,404,631 and 8,404,632. Polymeric H-NOX proteins and H-NOX proteins for treating brain cancer are described in PCT/US2013/020602. The contents of each are incorporated herein by reference in its entirety.

All references cited herein, including patent applications and publications, are incorporated herein by reference in their entirety.

BRIEF SUMMARY OF THE INVENTION

The invention is directed towards methods of delivering, monitoring, and optimizing the delivery of O2 to hypoxic tumors. In some aspects, the invention provides methods of treating a hypoxic brain tumor in an individual comprising a) administering an effective amount of an H-NOX protein to the individual, b) determining the level of hypoxia in the brain tumor following administration of the H-NOX protein, and c) administering an effective amount of radiation to the individual wherein the tumor hypoxia measured in step b) is reduced compared to the level of hypoxia in the brain tumor prior to H-NOX administration. In some aspects, the invention provides methods of treating a hypoxic brain tumor in an individual comprising a) determining the level of hypoxia in the brain tumor, b) administering an effective amount of an H-NOX protein to the individual, c) determining the level of hypoxia in the brain tumor following administration of the H-NOX protein, and d) administering an effective amount of radiation to the individual wherein the tumor hypoxia measured in step c) is reduced compared to the level of hypoxia measured in step a).

In some aspects, the invention provides methods of optimizing therapeutic efficacy for treatment of a hypoxic brain tumor in an individual, the method comprising a) administering H-NOX to the individual, b) measuring the level of hypoxia of the tumor one or more times after administration of the H-NOX protein, c) administering radiation therapy when tumor hypoxia is reduced compared to the level of hypoxia prior to H-NOX administration. In some embodiments, the hypoxia is reduced by at least about 5%, 10%, 15%, 20%, 25% or 50%. In some embodiments, the level of tumor hypoxia is first determined prior to administration of H-NOX.

In some aspects, the invention provides methods of monitoring the efficacy of delivery of O2 to hypoxic brain tumor by an H-NOX protein in an individual, the method comprising a) administering an effective amount of H-NOX protein to the individual, b) measuring the level of hypoxia in the tumor at one or more time points after administration of the H-NOX protein, wherein a reduction of tumor hypoxia compared to the level of hypoxia in the tumor prior to administration of H-NOX indicates effective delivery of O2 to the brain tumor. In some embodiments, the reduction in tumor hypoxia indicates that the individual is suitable for administration of radiation therapy. In some embodiments, the hypoxia is reduced by at least about 5%, 10%, 15%, 20%, 25% or 50%. In some embodiments, the level of tumor hypoxia is first determined prior to administration of H-NOX.

In some embodiments of the above aspects, the level of hypoxia in the tumor is measured one or more of one hour, two hours, three hours, four hours, eight hours, twelve hours, 24 hours, 48 hours or 72 hours after administration of H-NOX.

In some aspects, the invention provides methods of monitoring responsiveness or lack of responsiveness to treatment with an H-NOX in an individual suffering from a brain tumor comprising measuring the hypoxic state of the tumor following H-NOX administration, wherein responsiveness is indicated by a reduction in tumor hypoxia. In some embodiments, responsiveness indicates that the individual is suitable for administration of radiation therapy.

In some aspects, the invention provides methods of identifying an individual with a brain tumor who is more likely to exhibit benefit from a therapy comprising an H-NOX protein, said method comprising a) determining the hypoxia level of the tumor, b) administering H-NOX to the individual, c) measuring the level of hypoxia of the tumor, wherein about a 5% decrease in hypoxia indicates the individual is more likely to exhibit benefit from radiation treatment in combination with H-NOX treatment. In some embodiments, the decrease in hypoxia of step c) is a at least a 10%, a 15%, a 20%, a 25%, a 50%, a 75% or a 100% decrease in hypoxia.

In some embodiments of any of the above aspects and embodiments, tumor hypoxia is measured by one or more of 18F-fluoromisonidazole (FMISO) tumor uptake, pimidazole uptake, 18F-fluoroazomycin arabinoside (FAZA) uptake, a nitroimidazole uptake, Copper(II)-diacetyl-bis(N4-methylthiosemicarbazone (Cu-ATSM) uptake, 19F magnetic resonance imaging of hexafluorobenzene (C6F6) uptake, 1H MRI of hexamethyldisiloxane uptake, tumor HIF-1α expression, tumor Glut-1 expression, tumor LDHA expression, tumor carbonic anhydrase IX (CA-9) expression, or lactate and/or pyruvate levels.

In some embodiments of any of the above aspects and embodiments, the determination of the level of hypoxia in the tumor is repeated. In some embodiments, the determination of tumor hypoxia is repeated after one or more of one week, two weeks, three weeks, or four weeks. In further embodiments, the administration of H-NOX to the individual is repeated if the tumor is hypoxic. In some embodiments, the methods further comprising administration of radiation following administration of H-NOX. In yet further embodiments, the administration or radiation is repeated if the tumor has reduced hypoxia compared to the tumor prior to administration of H-NOX.

In some embodiments of any of the above aspects and embodiments, the radiation is X-radiation. In some embodiments, the X-radiation is administered at about 0.5 gray to about 75 gray.

In some embodiments of any of the above aspects and embodiments, the brain cancer is glioblastoma. In some embodiments, the individual is a mammal. In some embodiments, the mammal is a human. In other embodiments, the mammal is a pet, a laboratory research animal, or a farm animal. In further embodiments, the pet, research animal or farm animal is a dog, a cat, a horse, a monkey, a rabbit, a rat, a mouse, a guinea pig, a hamster, a pig, or a cow.

In some embodiments of any of the above aspects and embodiments, the H-NOX protein is a T. tengcongensis H-NOX, a L. pneumophilia 2 H-NOX, a H. sapiens β1, a R. norvegicus β1, a C. lupus H-NOX domain, a D. melangaster β1, a D. melangaster CG14885-PA, a C. elegans GCY-35, a N. punctiforme H-NOX, C. crescentus H-NOX, a S. oneidensis H-NOX, or C. acetobutylicum H-NOX.

In some embodiments of any of the above aspects and embodiments, the H-NOX protein comprises an H-NOX domain corresponding to the H-NOX domain of T. tengcongensis set forth in SEQ ID NO:2. In some embodiments, the H-NOX comprises one or more distal pocket mutations. In some embodiments, the distal pocket mutation is an amino acid substitution at a site corresponding to L144 of T. tengcongensis H-NOX. In some embodiments, the H-NOX is a T. tengcongensis H-NOX comprising an amino acid substitution at position 144. In some embodiments, the amino acid substitution at position 144 is an L144F substitution. In some embodiments, the H-NOX comprises at least two distal pocket mutations. In further embodiments, the at least two distal pocket mutations are amino acid substitutions at sites corresponding to W9 and L144 of T. tengcongensis H-NOX. In other embodiments, the H-NOX is a T. tengcongensis H-NOX comprising amino acid substitutions at positions 9 and 144. In some embodiments, the amino acid substitution at position 9 is a W9F substitution and the amino acid substitution at position 144 is an L144F substitution.

In some embodiments of any of the above aspects and embodiments, the H-NOX protein is a polymeric H-NOX protein. In some embodiments, the polymeric H-NOX protein comprises monomers, wherein the monomers comprise an H-NOX domain and a polymerization domain. In some embodiments, the H-NOX domain is covalently linked to the polymerization domain. In some embodiments, the polymeric H-NOX protein is a trimeric H-NOX protein. In some embodiments, the trimeric H-NOX protein comprises one or more trimerization domains. In other embodiments, the trimeric H-NOX protein comprises three monomers, wherein the monomers comprise an H-NOX domain and a trimerization domain, wherein the trimerization domain is a bacteriophage T4 trimerization domain. In further embodiments, the trimerization domain is a foldon domain. In further embodiments, the foldon domain comprises the amino acid sequence of SEQ ID NO:4. In some embodiments, the trimeric H-NOX protein comprises three H-NOX monomers wherein each H-NOX monomer is fused to a foldon domain. In some embodiments, the trimeric H-NOX protein comprises three Tt L144F H-NOX monomers wherein each Tt L144F H-NOX monomer is fused to a foldon domain.

In some embodiments of any of the above aspects and embodiments, the H-NOX protein is fused to an Fc domain of an immunoglobulin. In other embodiments, the H-NOX protein is covalently bound to polyethylene glycol.

In some embodiments of any of the above aspects and embodiments, the H-NOX protein does not comprise a guanylyl cyclase domain.

In some embodiments of any of the above aspects and embodiments, the O2 dissociation constant of the H-NOX protein is within 2 orders of magnitude of that of hemoglobin, and wherein the NO reactivity of the H-NOX protein is at least 10-fold lower than that of hemoglobin. In some embodiments, the O2 dissociation constant of the polymeric H-NOX protein is between about 1 nM and about 1000 nM at 20° C. In some embodiments, the O2 dissociation constant of the H-NOX protein is between about 1 μM and about 10 μM at 20° C. In some embodiments, the O2 dissociation constant of the H-NOX protein is between about 10 μM and about 50 μM at 20° C. In some embodiments, the NO reactivity of the H-NOX protein is less than about 700 s−1 at 20° C. In some embodiments, the NO reactivity of the H-NOX protein is at least 100-fold lower than that of hemoglobin. In other embodiments, the NO reactivity of the H-NOX protein is at least 1,000-fold lower than that of hemoglobin. In some embodiments, the koff for oxygen of the H-NOX protein is less than or equal to about 0.65 s−1 at 20° C. In some embodiments, the koff for oxygen of the H-NOX protein is between about 0.21 s−1 and about 0.65 s−1 at 20° C. In some embodiments, the koff for oxygen of the H-NOX protein is between about 1.35 s−1 and about 2.9 s−1 at 20° C. In some embodiments, the rate of heme autoxidation of the H-NOX protein is less than about 1 h−1 at 37° C.

BRIEF DESCRIPTION OF THE DRAWINGS

FIGS. 1A and 1B show that H-NOX monomer and H-NOX trimer is distributed and retained in mice bearing HCT-116 colon-derived tumors. FIG. 1A shows immunohistochemistry staining of tumors with H-NOX protein antibody showed persistence of H-NOX trimer in tumors for 60 minutes as compared to H-NOX monomer which was partially cleared at 60 minutes. FIG. 1B shows quantification of H-NOX protein staining intensity in HCT-116 tumor sections. N=6, all groups. Mean values+/−SEM.

FIGS. 2A and 2B show that H-NOX monomer and H-NOX trimer reduced tumor hypoxia in mice bearing HCT-116 colon-derived tumors. FIG. 2A shows representative tumor section of a 125 mm3 tumor isolated from mice treated with vehicle, H-NOX monomer, or H-NOX trimer. FIG. 2B shows quantification of an anti-pimonidazole antibody (Hypoxyprobe-1) intensity in tumor sections. N=6, all groups. Mean values+/−SEM. * indicates hypoxia throughout tumor, ** indicates no hypoxia in tumor.

FIGS. 3A-3C show tumor penetration and oxygenation by H-NOX monomer in mice bearing HCT-116 colon-derived tumors. FIG. 3A shows tumor sections stained with an anti-H-NOX protein antibody. FIG. 3B shows tumor sections stained with Hypoxyprobe-1. FIG. 3C shows quantification of the Hypoxyprobe-1 as a function of distance from the vasculature in the tumors from six mice per group. * indicates hypoxia throughout tumor, indicates no hypoxia in tumor.

FIGS. 4A-4G show that H-NOX trimer was distributed and retained in mice bearing a RIF-1 syngeneic sarcoma tumor Immunofluorescence images of a representative section from a 400 mm3 tumor isolated from a mouse 120 minutes after administration of 750 mg/kg H-NOX trimer (FIG. 4A) or buffer (FIG. 4C), and of a 800 mm3 tumor isolated from a mouse 120 minutes after administration of 750 mg/kg H-NOX trimer (FIG. 4B) or buffer (FIG. 4D). H-NOX protein staining was done with anti-H-NOX antibody. FIGS. 4E and 4F shows tumor oxygenation by H-NOX trimer in mice bearing a RIF-1 syngeneic sarcoma tumor. FIG. 4E shows tumor sections stained with an anti-pimonidazole antibody two hours after H-NOX or buffer control administration. Whole tumor picture is shown. FIG. 4F shows tumor sections stained with anti-pimonidazole antibody (Hypoxyprobe-1) and anti-CD31 antibody (BD Bioscience) two hours after H-NOX or buffer control administration. High magnification picture are shown. FIG. 4G shows biodistribution of H-NOX in RIF1 syngeneic sarcoma tumors. Two hours after intravenous injection, H-NOX trimer diffuses from the vasculature into the tumor tissue Immunohistochemistry staining of tumor sections with H-NOX antibody and CD31 antibody (vasculature marker, BD Bioscience). No fluorescent staining is detected in mice injected with buffer.

FIGS. 5A-5C show H-NOX trimer penetrated tumor in mice bearing a sarcoma derived tumor and reduced tumor hypoxia. FIG. 5A shows a western blot membrane was probed with an anti-H-NOX antibody for detection of H-NOX trimer, with Hypoxyprobe-1 for detection of hypoxia-associated proteins, or with an anti-actin antibody for assessment of total protein levels. FIG. 5B shows quantification of pimonidazole staining intensity in tumor sections. FIG. 5C shows quantification of anti-HIF-1α staining intensity in tumor sections.

FIGS. 6A and 6B are panels of immunohistochemistry images showing tumor penetration by H-NOX trimer and reduced brain tumor hypoxia in mice bearing U251 orthotopic brain tumors. FIG. 6A shows H-NOX trimer staining with an anti-H-NOX antibody in a U251 tumor two hours after administration with H-NOX trimer or saline (control). FIG. 6B shows hypoxyprobe-1 staining in U251 tumors two hours after administration with H-NOX trimer or saline (control). Enlarged images from a portion of the tumors are shown.

FIGS. 7A-7D show tumor penetration by H-NOX trimer and reduced brain tumor hypoxia in mice bearing U251 orthotopic brain tumors. FIG. 7A shoes immunofluorescence images of Hypoxyprobe-1 staining in U251 tumors two hours after administration with H-NOX trimer (right panels) or saline (buffer, left panels). FIG. 7B shows quantification of Hypoxyprobe-1 staining from the immunofluorescence images (H-NOX trimer-right panels or saline-left panels). FIG. 7C shows immunofluorescence images of HIF-1α staining in U251 tumor two hours after administration with H-NOX trimer or saline (buffer). FIG. 7D shows quantification of HIF-1α staining from the immunofluorescence images.

FIGS. 8A-8E show the biodistribution of H-NOX trimer in U251 orthotopic brain tumor and healthy brain. FIG. 8A shows H-NOX trimer staining with an anti-H-NOX antibody in a U251 tumor two hours after administration with H-NOX trimer. FIG. 8B shows nuclear DAPI staining in U251 tumors showing tumor localization in the brain. FIGS. 8C and 8D show enlarged images from a portion of the tumors from FIG. 8A show a diffused pattern of H-NOX inside the tumor and vascular-restricted pattern outside the tumor. FIG. 8E shows H-NOX trimer staining with an anti-H-NOX antibody and vasculature staining with anti-CD31 antibody (BD Bioscience) in healthy mouse brain.

FIGS. 9A and 9B show real-time fluorescent images of H-NOX monomer or H-NOX trimer in mouse U251 orthotopic glioblastoma tumors. FIG. 9A shows H-NOX monomer was cleared by two hours. FIG. 9B shows H-NOX trimer persisted in tumors, peaking at 1-4 hours. Images acquired by IVIS; arrows indicate areas of fluorescence above a specific threshold; asterisks indicate peak level of fluorescence intensity.

FIGS. 10A-10D show ex vivo fluorescence images of H-NOX monomer or H-NOX trimer in mouse BT-12 orthotopic glioblastoma tumors. Brains bearing BT-12 tumors were resected 30 minutes after 750 mg/kg H-NOX monomer administration (FIG. 10A), 60 minutes after 750 mg/kg H-NOX monomer administration (FIG. 10B), 60 minutes after 750 mg/kg H-NOX trimer administration (FIG. 10C), or 60 minutes after vehicle administration (FIG. 10D).

FIGS. 11A-11D show real-time fluorescence images of H-NOX monomer in mouse U251 orthotopic glioblastoma tumors. Imaging was acquired at 30 minutes (FIG. 11A), 60 minutes (FIG. 11B), 120 minutes (FIG. 11C), and 240 minutes (FIG. 11D) after H-NOX monomer administration.

FIGS. 12A-12D show real-time fluorescence images of H-NOX trimer in mouse U251 orthotopic glioblastoma tumors. Imaging was acquired at 30 minutes (FIG. 12A), 60 minutes (FIG. 12B), 120 minutes (FIG. 12C), and 240 minutes (FIG. 12D) after H-NOX trimer administration. Arrows indicate areas of fluorescence; asterisks indicate peak level of fluorescence intensity.

FIGS. 13A and 13B show real-time fluorescence images of H-NOX monomer in mouse U251 orthotopic glioblastoma tumors. Accumulation of H-NOX monomer in the kidney at 30 minutes (FIG. 13A) and 60 minutes (FIG. 13B) after H-NOX monomer administration.

FIGS. 14A-14F show real-time fluorescence images of H-NOX trimer in mouse GBM-43 orthotopic glioblastoma intracranial and spinal tumors. Distribution of H-NOX trimer in the spinal column prior to H-NOX trimer administration (FIG. 14A) and 0.5 hour (FIG. 14B), 1 hour (FIG. 14C), 2 hours (FIG. 14D), 4 hours (FIG. 14E), and 6 hours (FIG. 14F) after H-NOX trimer administration.

FIG. 15 shows real-time fluorescence images of H-NOX trimer in mouse U251 orthotopic glioblastoma intracranial tumors. Top panel shows the distribution of H-NOX trimer in the brain prior to H-NOX trimer administration (0 minutes) and at 30 min, 1 hour, 2 hours, 4 hours, 6 hours, and 72 hours after H-NOX trimer administration. Bottom panel shows the distribution of H-NOX monomer.

FIGS. 16A-16F show real-time bioluminescence images of H-NOX trimer in mouse U251 orthotopic glioblastoma intracranial and spinal tumors. H-NOX trimer distribution prior to H-NOX trimer administration (FIG. 16A) and at 30 min (FIG. 16B), 1 hour (FIG. 16C), 2 hours (FIG. 16D), 4 hours (FIG. 16E), and 6 hours (FIG. 16F) after H-NOX trimer administration at a dose of 295 mg/kg.

FIGS. 17A-17F show real-time fluorescence images of H-NOX trimer in mouse U251 orthotopic glioblastoma tumors. H-NOX trimer distribution prior to H-NOX trimer administration (FIG. 17A) and at 30 min (FIG. 17B), 1 hour (FIG. 17C), 2 hours (FIG. 17D), 4 hours (FIG. 17E), and 6 hours (FIG. 17F) after H-NOX trimer administration at a dose of 30 mg/kg.

FIGS. 17A-17F show real-time fluorescence images of H-NOX trimer L144F variant distribution in a U251 orthotopic glioblastoma mouse model containing small intracranial tumors. H-NOX trimer L144F variant distribution FIG. 18A) prior to H-NOX trimer administration and at 30 min (FIG. 18B), 1 hour (FIG. 18C), 2 hours (FIG. 18D), 4 hours (FIG. 18E), and 6 hours (FIG. 18F) after H-NOX trimer L144F variant administration at a dose of 30 mg/kg. Small tumors were 1000× fold smaller than large tumors as determined by bioluminescence (BLI) score.

FIGS. 19A-19D show fluorescence images of H-NOX trimer distribution. Ex vivo fluorescence images of a GBM43 orthotopic glioblastoma mouse model administered 30 mg/kg H-NOX trimer (FIG. 19A) or 750 mg/kg H-NOX trimer (FIG. 19B). Real-time bioluminescence imaging in a U251 orthotopic glioblastoma mouse model containing large intracranial tumors (FIG. 19C) or small intracranial tumors (FIG. 19D) after administration of 295 mg/kg H-NOX trimer.

FIG. 20 shows real-time fluorescence images of H-NOX trimer distribution in two mouse models of orthotopic glioblastoma tumors (U251 and GBM-43) and one model of an atypical teratoid/rhabdoid tumor (AT/RT). Images were taken 60 minutes after H-NOX trimer administration and the color scale for each image was optimized

FIGS. 21A-21E show ex vivo fluorescence images of H-NOX protein distribution in the tumor-bearing hemisphere of three mouse models of orthotopic glioblastoma tumors. FIG. 21A shows H-NOX trimer distribution 60 minutes after administration in a GBM43 orthotopic glioblastoma mouse model, FIG. 21B shows H-NOX trimer distribution 6 days after administration in a U251 orthotopic glioblastoma mouse model, FIG. 21C shows H-NOX monomer distribution 30 minutes after administration in a BT-12 an atypical teratoid/rhabdoid tumor (AT/RT) mouse model, FIG. 21D shows H-NOX trimer distribution 60 minutes after administration in a BT-12 orthotopic AT/RT mouse model, and FIG. 21E shows lack of H-NOX protein signal 30 minutes after vehicle administration in a BT-12 orthotopic AT/RT mouse model.

FIG. 22 is an immunofluorescence image showing escape of H-NOX trimer from the vasculature and diffusion throughout a U251 brain tumor in an orthotopic glioblastoma tumor mouse model. Tumor sections were stained with an anti-H-NOX antibody (top panel) and an anti-CD31 antibody (vasculature) (bottom panel).

FIGS. 23A-23D show that a trimeric H-NOX protein accumulates in and penetrates deep into the tumor tissue in the orthotopic glioblastoma mouse model. Mice bearing orthotopic glioblastoma BT-12 (FIG. 23A) or U251 tumors were injected with either 80 kD H-NOX trimer, OMX-4.80 (FIGS. 23A-D), or 23 kD H-NOX monomer, OMX-4 (FIG. 23A, top panel), via tail vein and monitored by live imaging of fluorescently labeled protein (FIGS. 23A, 23B) or immunohistochemical analysis using anti-H-NOX antibody (FIGS. 23C, 23D).

FIGS. 24A-24D show results of sandwich ELISA assays of H-NOX trimer in the brain of healthy mice. FIG. 24A shows an ELISA assay on brain after intravenous injection of H-NOX trimer (750 mg/kg). FIG. 24B shows an ELISA assay on brain after intravenous injection of H-NOX trimer (200 mg/kg). FIG. 24C shows the brain/plasma ratio of H-NOX trimer (750 mg/kg). FIG. 24D shows the brain/plasma ratio of H-NOX trimer (200 mg/kg). Plasma and brain were collected at 30, 60, 90 and 120 min after H-NOX trimer administration. N=3, all groups. Mean values+/−SEM.

FIGS. 25A-25C are a series of graphs showing that H-NOX trimer sensitized intracranial xenografts to fractionated radiation therapy in a U251 mouse model of human glioblastoma. FIG. 25A shows mean bioluminescence imaging (BLI) scores+/−SEM from mice in both treatment groups, as well as an untreated control group (no H-NOX, no RT). N=9, all groups. FIG. 25B shows individual BLI scores for the RT and RT+H-NOX trimer groups on Day 29 (box in A). Line shows group mean, +\−SEM. The BLI scores of the RT+H-NOX trimer mice were significantly lower than those from mice treated with RT alone (p=0.039, Student's t-test). FIG. 25C shows H-NOX trimer group showed significantly enhanced survival, as compared to mice that received only radiotherapy (p=0.025, logrank test).

FIGS. 26A and 26B are a series of graphs showing that H-NOX trimer sensitized intracranial xenografts to fractionated radiation therapy in two mouse models of human glioblastoma. FIG. 26A shows percent survival in a U251 orthotopic glioblastoma mouse model administered 2 Gy radiation therapy (2 Gy), H-NOX trimer L144F variant (L144F Trimer), 2 Gy radiation therapy in combination with H-NOX trimer L144F variant (2 Gy+L144F Trimer), or treatment buffer (TB). Logrank p-values: 2 Gy versus 2 Gy+L144F Trimer (p=0.158), 2 Gy versus TB (p=0.0612), and L144F Trimer versus TB (p=0.326). FIG. 26B shows percent survival in a GBM43 orthotopic glioblastoma mouse model administered 2 Gy radiation therapy (2 Gy), 4 Gy radiation therapy (4 Gy), 8 Gy radiation therapy (8 Gy), 2 cycles of 4 Gy radiation therapy (4 Gy×2), 4 Gy radiation therapy in combination with H-NOX trimer (4 Gy+H-NOX), or treatment buffer (untreated). Logrank p-values: 4 Gy versus 4 Gy+H-NOX (p=0.597), 4 Gy versus 4 Gy×2 (p=0.038), and 4 Gy×2 versus 4 Gy+H-NOX (p=0.111).

FIGS. 27A-27D show that a single dose of a trimeric H-NOX protein reduces tumor hypoxia. Mice bearing orthotopic glioblastoma U251 tumors were treated with either trimeric Tt L144F H-NOX, OMX-4.80, or buffer alone (top panels of FIGS. 27A and 27D) via tail vein bolus injection. Tumors were harvested 2 hr-30 hr after OMX-4.80 administration, and assayed by immunohistochemistry for pimonidazole (Hypoxyprobe-1 mAb) and total cell nuclear (DAPI) staining in FIGS. 27A and 27B, or for HIF1α and tumor cell marker (HLA) in FIGS. 27C and 27D. FIG. 27A shows representative tumor sections from mice treated with buffer or trimeric Tt L144F H-NOX. Hypoxia staining (pimonidazole) is shown in green (left column of FIG. 27A) and total cell nuclear staining (DAPI) is shown in dark blue (right column of FIG. 27A). FIG. 27B shows quantification of pimonidazole staining intensity in tumor sections. N=5-6 per group. Mean values+/−SEM, p<0.05. FIG. 27C shows quantification of HIF1α staining intensity in tumor sections. N=5-6 per group. Mean values+/−SEM, p<0.05. FIG. 27D shows representative tumor sections from mice treated with buffer or OMX-4.80. Hypoxia staining (HIF1α) is shown in green (left column of FIG. 27D) and staining of human tumor cells in red (HLA; right column of FIG. 27D). Only 2 hr and 24 hr time points are shown.

FIGS. 28A-28F show treatment with trimeric Tt L144F H-NOX reduces tumor hypoxia at the invasive edges of the tumor. Mice bearing orthotopic glioblastoma U251 tumors were treated with either buffer control (FIG. 28A and FIG. 28C) or trimeric Tt L144F H-NOX, OMX-4.80, (FIGS. 28B, 28D, 28E, and 28F), via tail vein injection and received an injection of hypoxia marker, pimonidazole an hour prior to sacrifice. Brains containing tumors were extracted and subjected to immunohistochemical analysis using anti-pimonidazole (FIGS. 28A and 28B), HIF-1α (FIGS. 28C and 28D), or trimeric Tt L144F H-NOX antibodies (FIGS. 28E and 28F). Slides were counterstained with DNA marker (DAPI, blue labeled nuclei) and images merged (FIGS. 28A-28D, and 28F).

FIGS. 29A-29D show treatment with a trimeric Tt L144F H-NOX protein enhances efficacy of a single dose of radiation in an orthothopic glioblastoma model and in a RIF-1 syngeneic tumor model. FIG. 29A shows mean bioluminescence imaging (BLI) scores±SEM from mice receiving H-NOX and 10 gray radiation (triangles) and 10 gray radiation in buffer control (no H-NOX, circles), as well as an untreated (buffer, no RT, inverted triangles) control group. N≧8, all groups. FIG. 29B shows that trimeric Tt L144F H-NOX group shows significantly enhanced survival, as compared to mice that received only radiotherapy (p<0.05, logrank test). FIG. 29C shows mean bioluminescence imaging (BLI) scores±SEM from mice receiving H-NOX and 10 gray radiation (triangles), 10 gray radiation in buffer control (circles), 15 gray radiation in buffer control (blue inverted triangles), as well as an untreated (buffer, no RT, black inverted triangles) control group. N≧8, all groups. FIG. 29D shows tumor volume measurements in RIF1 tumors: trimeric Tt L144F H-NOX+15 Gy group (red triangles), buffer+15 Gy alone (black dotted), buffer+25 Gy RT (gray dotted). Untreated control (solid black line). Inactive H-NOX, OMX-1.80+15 Gray (green line). Mean±SEM, N=7-9 per group, p<0.01, Student's t-test.

FIGS. 30A-30G show the nucleic acid and amino acid sequences of H-NOX proteins. FIG. 30A shows wild-type Thermoanaerobacter tengcongensis H-NOX (SEQ ID NOs:1 and 2). FIG. 30B shows wildtype Legionella pneumophilia Orf2 H-NOX (SEQ ID NOs:13 and 14). FIG. 30C shows wildtype Legionella pneumophilia Orf1 H-NOX (SEQ ID NOs:15 and 16). FIG. 30D shows Homo sapiens β1 (1-385) H-NOX (SEQ ID NOs:17 and 18). FIG. 30E shows Homo sapiens β2 (1-217) H-NOX (SEQ ID NOs:19 and 20). FIG. 30F shows Rattus norvegicus β1 H-NOX (SEQ ID NOs:21 and 22). FIG. 30G shows Rattus norvegicus β2 H-NOX (SEQ ID NOs:23 and 24).

FIG. 31A shows immunohistochemical staining for pimonidazole in GL261, U251, GBM43 and GBM6 intracranial tumor models. FIG. 31B shows immunohistochemical staining for HIF-1α in GL261, U251, GBM43 and GBM6 intracranial tumor models.

FIG. 32A shows a schematic representation of quantitative oxygen dependencies for OMX-4.80, OMX-1.80, bioreductive activation of imaging agents (pimonidazole), and biological responses to hypoxia (HIF-1α). Three commonly used units for oxygen concentration are shown on the x axis. This schematic is theoretical and adapted from the review “Targeting hypoxia in cancer therapy” by William R. Wilson & Michael P. Hay (Nature Reviews Cancer 11, 393-410, 2011). FIG. 32B shows a scatterplot comparing HIF-1α levels with pimonidazole levels obtained by immunohistochemical analysis in U251 intracranial tumors from mice treated with either buffer control or OMX-4.80.

DETAILED DESCRIPTION OF THE INVENTION

The present invention is based in part on the surprising discovery that H-NOX proteins preferentially extravasate and accumulate in tissues thereby providing a long oxygenation window and circulation half-life to deliver oxygen where needed to treat disease conditions. For example, H-NOX proteins can cross the blood-brain bather and accumulate in brain tumors such as gliomas. As such, H-NOX proteins can be used to deliver oxygen to sensitize hypoxic tumors to anticancer therapies such as radiation therapy. The invention includes methods of monitoring the oxygenation of tumors (e.g. brain tumors) which allows optimization of therapeutic efficacy for the treatment of hypoxic tumors.

In some aspects, the invention provides method of treating a hypoxic brain tumor in an individual where an effective amount of an H-NOX protein is administered to the individual to deliver oxygen to the tumor, determining the level of hypoxia in the brain tumor following administration of the H-NOX protein, and then administering an effective amount of radiation to the individual where the tumor hypoxia has decreased as a result of tumor oxygenation by the H-NOX protein.

In some aspects, the invention provides methods of optimizing therapeutic efficacy for treatment of a hypoxic brain cancer in an individual where H-NOX is administered to the individual to deliver O2 to the tumor, the level of hypoxia of the tumor is measured one or more times after administration of the H-NOX proteins such that radiation therapy may be administered to the individual when tumor hypoxia is reduced compared to the level of hypoxia prior to H-NOX administration.

In some aspects, the invention provides methods of monitoring the efficacy of delivery of O2 to hypoxic brain tumor by an H-NOX protein in an individual wherein an effective amount of H-NOX protein is administered to the individual, the level of hypoxia in the tumor is measured at one or more time points after administration of the H-NOX protein, wherein a reduction of tumor hypoxia compared to the level of hypoxia in the tumor prior to administration of H-NOX indicates effective delivery of O2 to the brain tumor.

In some aspects, the invention provides methods of monitoring responsiveness or lack of responsiveness to treatment with an H-NOX in an individual suffering from a brain tumor comprising measuring the hypoxic state of the tumor following H-NOX administration, wherein responsiveness is indicated by a reduction in tumor hypoxia.

In some aspects, the invention provides methods of identifying an individual with a brain tumor who is more likely to exhibit benefit from a therapy. The method includes determining the hypoxia level of the tumor, administering H-NOX to the individual to deliver oxygen to the tumor, and measuring the level of hypoxia of the tumor. A 5% decrease in hypoxia indicates the individual is more likely to exhibit benefit from radiation treatment.

DEFINITIONS

Unless defined otherwise, the meanings of all technical and scientific terms used herein are those commonly understood by one of skill in the art to which this invention belongs. One of skill in the art will also appreciate that any methods and materials similar or equivalent to those described herein can also be used to practice or test the invention.

For use herein, unless clearly indicated otherwise, use of the terms “a”, “an,” and the like refers to one or more.

In this application, the use of “or” means “and/or” unless expressly stated or understood by one skilled in the art. In the context of a multiple dependent claim, the use of “or” refers back to more than one preceding independent or dependent claim.

Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X.”

It is understood that aspect and embodiments of the invention described herein include “comprising,” “consisting,” and “consisting essentially of” aspects and embodiments.

The terms “polypeptide” and “protein” are used interchangeably to refer to a polymer of amino acid residues, and are not limited to a minimum length. Such polymers of amino acid residues may contain natural or non-natural amino acid residues, and include, but are not limited to, peptides, oligopeptides, dimers, trimers, and polymers of amino acid residues. Both full-length proteins and fragments thereof are encompassed by the definition. The terms also include post-expression modifications of the polypeptide, for example, glycosylation, sialylation, acetylation, phosphorylation, and the like. Furthermore, for purposes of the present invention, a “polypeptide” refers to a protein which includes modifications, such as deletions, additions, and substitutions (generally conservative in nature), to the native sequence, as long as the protein maintains the desired activity. These modifications may be deliberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the proteins or errors due to PCR amplification. As used herein, a protein may include two or more subunits, covalently or non-covalently associated; for example, a protein may include two or more associated monomers.

The terms “nucleic acid molecule”, “nucleic acid” and “polynucleotide” may be used interchangeably, and refer to a polymer of nucleotides. Such polymers of nucleotides may contain natural and/or non-natural nucleotides, and include, but are not limited to, DNA, RNA, and PNA. “Nucleic acid sequence” refers to the linear sequence of nucleotides that comprise the nucleic acid molecule or polynucleotide.

As used herein, an “H-NOX protein” means a protein that has an H-NOX domain (named for Heme-Nitric oxide and OXygen binding domain). An H-NOX protein may or may not contain one or more other domains in addition to the H-NOX domain. In some examples, an H-NOX protein does not comprise a guanylyl cyclase domain. An H-NOX protein may or may not comprise a polymerization domain.

As used herein, a “polymeric H-NOX protein” is an H-NOX protein comprising two or more H-NOX domains. The H-NOX domains may be covalently or non-covalently associated.

As used herein, an “H-NOX domain” is all or a portion of a protein that binds nitric oxide and/or oxygen by way of heme. The H-NOX domain may comprise heme or may be found as an apoproprotein that is capable of binding heme. In some examples, an H-NOX domain includes six alpha-helices, followed by two beta-strands, followed by one alpha-helix, followed by two beta strands. In some examples, an H-NOX domain corresponds to the H-NOX domain of Thermoanaerobacter tengcongensis H-NOX set forth in SEQ ID NO:2. For example, the H-NOX domain may be at least about 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% identical to the H-NOX domain of Thermoanaerobacter tengcongensis H-NOX set forth in SEQ ID NO:2. In some embodiments, the H-NOX domain may be 10%-20%, 20%-30%, 30%-40%, 40%-50%, 50%-60%, 60%-70%, 70%-80%, 80%-90%, 90%-95%, 95%-99% or 100% identical to the H-NOX domain of Thermoanaerobacter tengcongensis H-NOX set forth in SEQ ID NO:2.

As used herein, a “polymerization domain” is a domain (e.g. a polypeptide domain) that promotes the association of monomeric moieties to form a polymeric structure. For example, a polymerization domain may promote the association of monomeric H-NOX domains to generate a polymeric H-NOX protein. An exemplary polymerization domain is the foldon domain of T4 bacteriophage, which promotes the formation of trimeric polypeptides. Other examples of polymerization domains include, but are not limited to, Arc, POZ, coiled coil domains (including GCN4, leucine zippers, Velcro), uteroglobin, collagen, 3-stranded coiled colis (matrilin-1), thrombosporins, TRPV1-C, P53, Mnt, avadin, streptavidin, Bcr-Abl, COMP, verotoxin subunit B, CamKII, RCK, and domains from N ethylmaleimide-sensitive fusion protein, STM3548, KaiC, TyrR, Hcp1, CcmK4, GP41, anthrax protective antigen, aerolysin, a-hemolysin, C4b-binding protein, Mi-CK, arylsurfatase A, and viral capsid proteins.

As used herein, an “amino acid linker sequence” or an “amino acid spacer sequence” is a short polypeptide sequence that may be used to link two domains of a protein. In some embodiments, the amino acid linker sequence is one, two, three, four, five, six, seven, eight, nine, ten or more than ten amino acids in length. Exemplary amino acid linker sequences include but are not limited to a Gly-Ser-Gly sequence and an Arg-Gly-Ser sequence.

As used herein, a “His6 tag” refers to a peptide comprising six His residues attached to a polypeptide. A His6 tag may be used to facilitate protein purification; for example, using chromatography specific for the His6 tag. Following purification, the His6 tag may be cleaved using an exopeptidase.

The term “substantially similar” or “substantially the same,” as used herein, denotes a sufficiently high degree of similarity between two or more numeric values such that one of skill in the art would consider the difference between the two or more values to be of little or no biological and/or statistical significance within the context of the biological characteristic measured by said value. In some embodiments the two or more substantially similar values differ by no more than about any one of 5%, 10%, 15%, 20%, 25%, or 50%.

The phrase “substantially reduced,” or “substantially different,” as used herein, denotes a sufficiently high degree of difference between two numeric values such that one of skill in the art would consider the difference between the two values to be of statistical significance within the context of the biological characteristic measured by said values. In some embodiments, the two substantially different numeric values differ by greater than about any one of 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, or 90%. In some embodiment, the two substantially different numeric values differ by about any one of 10%-20%, 20%-30%, 30%-40%, 40%-50%, 50%-60%, 60%-70%, 70%-80%, 80%-90%, 90%-95%, 95%-99% or 100%.

A “native sequence” polypeptide comprises a polypeptide having the same amino acid sequence as a polypeptide found in nature. Thus, a native sequence polypeptide can have the amino acid sequence of naturally occurring polypeptide from any organism. Such native sequence polypeptide can be isolated from nature or can be produced by recombinant or synthetic means. The term “native sequence” polypeptide specifically encompasses naturally occurring truncated or secreted forms of the polypeptide (e.g., an extracellular domain sequence), naturally occurring variant forms (e.g., alternatively spliced forms) and naturally occurring allelic variants of the polypeptide.

A polypeptide “variant” means a biologically active polypeptide having at least about 80% amino acid sequence identity with the native sequence polypeptide after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Such variants include, for instance, polypeptides wherein one or more amino acid residues are added, or deleted, at the N- or C-terminus of the polypeptide. In some embodiments, a variant will have at least about any one of 80%, 90% or 95% amino acid sequence identity with the native sequence polypeptide. In some embodiments, a variant will have about any one of 80%-90%, 90%-95% or 95%-99% amino acid sequence identity with the native sequence polypeptide.

As used herein, a “mutant protein” means a protein with one or more mutations compared to a protein occurring in nature. In one embodiment, the mutant protein has a sequence that differs from that of all proteins occurring in nature. In various embodiments, the amino acid sequence of the mutant protein is at least about any of 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 95, 97, 98, 99, or 99.5% identical to that of the corresponding region of a protein occurring in nature. In some embodiments, the amino acid sequence of the mutant protein is at least about any of 10%-20%, 20%-30%, 30%-40%, 40%-50%, 50%-60%, 60%-70%, 70%-80%, 80%-90%, 90%-95%, 95%-99% or 100% identical to that of the corresponding region of a protein occurring in nature. In some embodiments, the mutant protein is a protein fragment that contains at least about any of 25, 50, 75, 100, 150, 200, 300, or 400 contiguous amino acids from a full-length protein. In some embodiments, the mutant protein is a protein fragment that contains about any of 25-50, 50-75, 75-100, 100-150, 150-200, 200-300, or 300-400 contiguous amino acids from a full-length protein. Sequence identity can be measured, for example, using sequence analysis software with the default parameters specified therein (e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705). This software program matches similar sequences by assigning degrees of homology to various amino acids replacements, deletions, and other modifications.

As used herein, a “mutation” means an alteration in a reference nucleic acid or amino acid sequence occurring in nature. Exemplary nucleic acid mutations include an insertion, deletion, frameshift mutation, silent mutation, nonsense mutation, or missense mutation. In some embodiments, the nucleic acid mutation is not a silent mutation. Exemplary protein mutations include the insertion of one or more amino acids (e.g., the insertion of 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids), the deletion of one or more amino acids (e.g., a deletion of N-terminal, C-terminal, and/or internal residues, such as the deletion of at least about any of 5, 10, 15, 25, 50, 75, 100, 150, 200, 300, or more amino acids or a deletion of about any of 5-10, 10-15, 15-25, 25-50, 50-75, 75-100, 100-150, 150-200, 200-300, or 300-400 amino acids), the replacement of one or more amino acids (e.g., the replacement of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids), or combinations of two or more of the foregoing. The nomenclature used in referring to a particular amino acid mutation first identifies the wild-type amino acid, followed by the residue number and finally the substitute amino acid. For example, Y140L means that tyrosine has been replaced by a leucine at residue number 140. Likewise, a variant H-NOX protein may be referred to by the amino acid variations of the H-NOX protein. For example, a T. tengcongensis Y140L H-NOX protein refers to a T. tengcongensis H-NOX protein in which the tyrosine residue at position number 140 has been replaced by a leucine residue and a T. tengcongensis W9F/Y140L H-NOX protein refers to a T. tengcongensis H-NOX protein in which the tryptophan residue at position 9 has been replaced by a phenylalanine residue and the tyrosine residue at position number 140 has been replaced by a leucine residue.

An “evolutionary conserved mutation” is the replacement of an amino acid in one protein by an amino acid in the corresponding position of another protein in the same protein family.

As used herein, “derived from” refers to the source of the protein into which one or more mutations is introduced. For example, a protein that is “derived from a mammalian protein” refers to protein of interest that results from introducing one or more mutations into the sequence of a wild-type (i.e., a sequence occurring in nature) mammalian protein.

As used herein, “Percent (%) amino acid sequence identity” and “homology” with respect to a peptide, polypeptide or antibody sequence are defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the specific peptide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or MEGALIGN™ (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.

As used herein, a “koff” refers to a dissociation rate, such as the rate of release of O2 or NO from a protein. A lower numerical lower koff indicates a slower rate of dissociation.

As used herein, “kon” refers to an association rate, such as the rate of binding of O2 or NO to a protein. A lower numerical lower kon indicates a slower rate of association.

As used herein, “dissociation constant” refers to a “kinetic dissociation constant” or a “calculated dissociation constant.” A “kinetic dissociation constant” or “KD” is a ratio of kinetic off-rate (koff) to kinetic on-rate (kon), such as a KD value determined as an absolute value using standard methods (e.g., standard spectroscopic, stopped-flow, or flash-photolysis methods) including methods known to the skilled artisan and/or described herein. “Calculated dissociation constant” or “calculated KD” refers to an approximation of the kinetic dissociation constant based on a measured koff. A value for the kon is derived via the correlation between kinetic KD and koff as described herein.

As used herein, “oxygen affinity” is a qualitative term that refers to the strength of oxygen binding to the heme moiety of a protein. This affinity is affected by both the koff and kon for oxygen. A numerically lower oxygen KD value means a higher affinity.

As used herein, “NO affinity” is a qualitative term that refers to the strength of NO binding to a protein (such as binding to a heme group or to an oxygen bound to a heme group associated with a protein). This affinity is affected by both the koff and kon for NO. A numerically lower NO KD value means a higher affinity.

As used herein, “NO stability” refers to the stability or resistance of a protein to oxidation by NO in the presence of oxygen. For example, the ability of the protein to not be oxidized when bound to NO in the presence of oxygen is indicative of the protein's NO stability. In some embodiments, less than about any of 50, 40, 30, 10, or 5% of an H-NOX protein is oxidized after incubation for about any of 1, 2, 4, 6, 8, 10, 15, or 20 hours at 20° C.

As used herein, “NO reactivity” refers to the rate at which iron in the heme of a heme-binding protein is oxidized by NO in the presence of oxygen. A lower numerical value for NO reactivity in units of s−1 indicates a lower NO reactivity

As used herein, an “autoxidation rate” refers to the rate at which iron in the heme of a heme-binding protein is autoxidized. A lower numerical autoxidation rate in units of s−1 indicates a lower autoxidation rate.

The term “vector” is used to describe a polynucleotide that may be engineered to contain a cloned polynucleotide or polynucleotides that may be propagated in a host cell. A vector may include one or more of the following elements: an origin of replication, one or more regulatory sequences (such as, for example, promoters and/or enhancers) that regulate the expression of the polypeptide of interest, and/or one or more selectable marker genes (such as, for example, antibiotic resistance genes and genes that may be used in colorimetric assays, e.g., β-galactosidase). The term “expression vector” refers to a vector that is used to express a polypeptide of interest in a host cell.

A “host cell” refers to a cell that may be or has been a recipient of a vector or isolated polynucleotide. Host cells may be prokaryotic cells or eukaryotic cells. Exemplary eukaryotic cells include mammalian cells, such as primate or non-primate animal cells; fungal cells, such as yeast; plant cells; and insect cells. Exemplary prokaryotic cells include bacterial cells; for example, E. coli cells.

The term “isolated” as used herein refers to a molecule that has been separated from at least some of the components with which it is typically found in nature or produced. For example, a polypeptide is referred to as “isolated” when it is separated from at least some of the components of the cell in which it was produced. Where a polypeptide is secreted by a cell after expression, physically separating the supernatant containing the polypeptide from the cell that produced it is considered to be “isolating” the polypeptide. Similarly, a polynucleotide is referred to as “isolated” when it is not part of the larger polynucleotide (such as, for example, genomic DNA or mitochondrial DNA, in the case of a DNA polynucleotide) in which it is typically found in nature, or is separated from at least some of the components of the cell in which it was produced, e.g., in the case of an RNA polynucleotide. Thus, a DNA polynucleotide that is contained in a vector inside a host cell may be referred to as “isolated”.

The terms “individual” or “subject” are used interchangeably herein to refer to an animal; for example a mammal. In some embodiments, methods of treating mammals, including, but not limited to, humans, rodents, simians, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets, are provided. In some examples, an “individual” or “subject” refers to an individual or subject in need of treatment for a disease or disorder.

A “disease” or “disorder” as used herein refers to a condition where treatment is needed.

The term “cancer” refers to a malignant proliferative disorder associated with uncontrolled cell proliferation, unrestrained cell growth, and decreased cell death via apoptosis.

The term “tumor” is used herein to refer to a group of cells that exhibit abnormally high levels of proliferation and growth. A tumor may be benign, pre-malignant, or malignant; malignant tumor cells are cancerous. Tumor cells may be solid tumor cells or leukemic tumor cells. The term “tumor growth” is used herein to refer to proliferation or growth by a cell or cells that comprise a tumor that leads to a corresponding increase in the size of the tumor.

As used herein, “treatment” is an approach for obtaining beneficial or desired clinical results. “Treatment” as used herein, covers any administration or application of a therapeutic for disease in a mammal, including a human. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, any one or more of: alleviation of one or more symptoms, diminishment of extent of disease, preventing or delaying spread (e.g., metastasis, for example metastasis to the lung or to the lymph node) of disease, preventing or delaying recurrence of disease, delay or slowing of disease progression, amelioration of the disease state, inhibiting the disease or progression of the disease, inhibiting or slowing the disease or its progression, arresting its development, and remission (whether partial or total). Also encompassed by “treatment” is a reduction of pathological consequence of a proliferative disease. The methods of the invention contemplate any one or more of these aspects of treatment.

In the context of cancer, the term “treating” includes any or all of: inhibiting growth of tumor cells or cancer cells, inhibiting replication of tumor cells or cancer cells, lessening of overall tumor burden and ameliorating one or more symptoms associated with the disease.

The terms “inhibition” or “inhibit” refer to a decrease or cessation of any phenotypic characteristic or to the decrease or cessation in the incidence, degree, or likelihood of that characteristic. To “reduce” or “inhibit” is to decrease, reduce or arrest an activity, function, and/or amount as compared to a reference. In certain embodiments, by “reduce” or “inhibit” is meant the ability to cause an overall decrease of 20% or greater. In another embodiment, by “reduce” or “inhibit” is meant the ability to cause an overall decrease of 50% or greater. In yet another embodiment, by “reduce” or “inhibit” is meant the ability to cause an overall decrease of 75%, 85%, 90%, 95%, or 99%.

As used herein, “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, suppress and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. For example, a late stage cancer, such as development of metastasis, may be delayed.

A “reference” as used herein, refers to any sample, standard, or level that is used for comparison purposes. A reference may be obtained from a healthy and/or non-diseased sample. In some examples, a reference may be obtained from an untreated sample. In some examples, a reference is obtained from a non-diseased on non-treated sample of a subject individual. In some examples, a reference is obtained from one or more healthy individuals who are not the subject or patient.

“Preventing,” as used herein, includes providing prophylaxis with respect to the occurrence or recurrence of a disease in a subject that may be predisposed to the disease but has not yet been diagnosed with the disease.

An “effective amount” of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.

A “therapeutically effective amount” of a substance/molecule of the invention, agonist or antagonist may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the substance/molecule, agonist or antagonist to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the substance/molecule, agonist or antagonist are outweighed by the therapeutically beneficial effects. A therapeutically effective amount may be delivered in one or more administrations. A therapeutically effective amount also encompasses an amount sufficient to confer benefit, e.g., clinical benefit.

A “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.

Responsiveness of a patient can be assessed using any endpoint indicating a benefit to the patient, including, without limitation, (1) inhibition, to some extent, of disease progression, including slowing down and complete arrest; (2) reduction in lesion size; (3) inhibition (i.e., reduction, slowing down or complete stopping) of disease cell infiltration into adjacent peripheral organs and/or tissues; (4) inhibition (i.e. reduction, slowing down or complete stopping) of disease spread; (5) relief, to some extent, of one or more symptoms associated with the disorder; (6) increase in the length of disease-free presentation following treatment; and/or (8) decreased mortality at a given point of time following treatment. In the case of H-NOX mediated oxygenation of hypoxic tissue (e.g. a hypoxic tumor), responsiveness can be assessed by oxygenation of the tissue or by a reduction in the hypoxia of the tissue. In some embodiments, any reduction in tissue hypoxia indicates responsiveness to H-NOX treatment. In some embodiments, a reduction in tissue hypoxia of about 5% or greater indicates responsiveness to H-NOX treatment.

The term “benefit” is used in the broadest sense and refers to any desirable effect and specifically includes clinical benefit as defined herein. In some embodiments, tumor oxygenation or a reduction in tumor hypoxia indicates benefit.

Clinical benefit can be measured by assessing various endpoints, e.g., inhibition, to some extent, of disease progression, including slowing down and complete arrest; reduction in the number of disease episodes and/or symptoms; reduction in lesion size; inhibition (i.e., reduction, slowing down or complete stopping) of disease cell infiltration into adjacent peripheral organs and/or tissues; inhibition (i.e. reduction, slowing down or complete stopping) of disease spread; relief, to some extent, of one or more symptoms associated with the disorder; increase in the length of disease-free presentation following treatment, e.g., progression-free survival; increased overall survival; higher response rate; and/or decreased mortality at a given point of time following treatment. In the case of H-NOX mediated oxygenation of hypoxic tissue (e.g. a hypoxic tumor), clinical benefit can be assessed by oxygenation of the tissue or by a reduction in the hypoxia of the tissue. In some embodiments, any reduction in tissue hypoxia indicates benefit of H-NOX treatment. In some embodiments, a reduction in tissue hypoxia of about 5% or greater indicates benefit of H-NOX treatment.

The terms “pharmaceutical formulation” and “pharmaceutical composition” refer to a preparation which is in such form as to permit the biological activity of the active ingredient(s) to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered. Such formulations may be sterile and essentially free of endotoxins.

A “pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid, or liquid filler, diluent, encapsulating material, formulation auxiliary, or carrier conventional in the art for use with a therapeutic agent that together comprise a “pharmaceutical composition” for administration to a subject. A pharmaceutically acceptable carrier is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation. The pharmaceutically acceptable carrier is appropriate for the formulation employed.

A “sterile” formulation is aseptic or essentially free from living microorganisms and their spores.

Administration “in combination with” one or more further therapeutic agents includes simultaneous (concurrent) and consecutive or sequential administration in any order.

The term “concurrently” is used herein to refer to administration of two or more therapeutic agents, where at least part of the administration overlaps in time or where the administration of one therapeutic agent falls within a short period of time relative to administration of the other therapeutic agent. For example, the two or more therapeutic agents are administered with a time separation of no more than about 60 minutes, such as no more than about any of 30, 15, 10, 5, or 1 minutes.

The term “sequentially” is used herein to refer to administration of two or more therapeutic agents where the administration of one or more agent(s) continues after discontinuing the administration of one or more other agent(s). For example, administration of the two or more therapeutic agents are administered with a time separation of more than about 15 minutes, such as about any of 20, 30, 40, 50, or 60 minutes, 1 day, 2 days, 3 days, 1 week, 2 weeks, or 1 month.

As used herein, “in conjunction with” refers to administration of one treatment modality in addition to another treatment modality. As such, “in conjunction with” refers to administration of one treatment modality before, during or after administration of the other treatment modality to the individual.

The term “package insert” is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.

An “article of manufacture” is any manufacture (e.g., a package or container) or kit comprising at least one reagent, e.g., a medicament for treatment of a disease or disorder (e.g., cancer), or a probe for specifically detecting a biomarker described herein. In certain embodiments, the manufacture or kit is promoted, distributed, or sold as a unit for performing the methods described herein.

As used herein, “HIF-1α” refers to the alpha subunit of hypoxia inducible factor1. Examples of HIF-1α include but are not limited to human HIF-1α (NCBI RefSeq NP001521, NP851397, NM181054, and NM001530), murine HIF-1α (NCBI RefSeq NP034561 and NM010431), rat HIF-1α (NCBI RefSeq NP077335 and NM024359), canine HIF-1α (NCBI RefSeq XP865494, XP865463, XM860350, and XM860370), chimpanzee HIF-1α (NCBI RefSeq XP001168834, XP001168628, XM001168972, and XM001168811), monkey HIF-1α (NCBI RefSeq XP002805105, XP002805107, XM002805059, and XM002805060), and the like.

As used herein, “HIF-2α” or “endothelial PAS domain protein 1” refers to the alpha subunit of hypoxia inducible factor 2. Examples of HIF-2α include but are not limited to human HIF-2α (NCBI RefSeq NP001421 and NM001430), murine HIF-2α (NCBI RefSeq NP034267 and NM010137), and rat HIF-2α (NCBI RefSeq NP075578 and NM023090), and the like.

As used herein, “HIF-3α” refers to the alpha subunit of hypoxia inducible factor 3. Examples of HIF-3α include but are not limited to human HIF-3α (NCBI RefSeq NP690009, NP690008, NM152796, NM152795), murine HIF-3α (NCBI RefSeq NP001156422, NP058564, NM001162950, NM016868), rat HIF-3α (NCBI RefSeq NP071973 and NM022528), chimpanzee HIF-3α (NCBI RefSeq XP512767, XP001167388, XM001167499, and XM001167448), canine HIF-3α (NCBI RefSeq XP533636 and XM533636), and the like.

As used herein, “glut1” or “solute carrier family 2 (facilitated glucose transporter), member 1” refers to the glucose transporter type 1. Examples of glut1 include but are not limited to human glut1 (NCBI RefSeq NP006507 and NM006516), murine glut1 (NCBI RefSeq NP035530 and NM011400), rat glut1 (NCBI RefSeq NP620182 and NM138827), and the like.

As used herein, “LDHA” refers to Lactose DeHydrogenase A. Examples of LDHA include but are not limited to human LDHA (NCBI RefSeq NP001158888, NP001158887, NM001135239, NM001165416), murine LDHA (NCBI RefSeq NP001129541, NP034829 and NM001136069, NM010699), rat LDHA (NCBI RefSeq NP058721 and NM017025), chimpanzee LDHA (NCBI RefSeq NP001029268 and NM001034096), canine LDHA (NCBI RefSeq XP534084, XP865353, XM534084, and XM860260), and the like.

As used herein, “CA-9”, “caix” or “carbonic anhydrase IX” refers to an isoform of carbonic anhydrase. Examples of carbonic anhydrase IX include but are not limited to human carbonic anhydrase IX (NCBI RefSeq NP001207 and NM001216), murine carbonic anhydrase IX (NCBI RefSeq NP647466 and NM139305), rat carbonic anhydrase IX (NCBI RefSeq NP001101426 and NM001107956), chimpanzee carbonic anhydrase IX (NCBI RefSeq XP001167245, XP528593, XM528593, and XM001167245), canine carbonic anhydrase IX (NCBI RefSeq XP854749, NP001138646, NM001145174, and XM849656), and the like.

The term “biomarker” as used herein refers to an indicator, e.g., predictive, diagnostic, and/or prognostic, which can be detected in a sample. The biomarker may serve as an indicator of a particular subtype of a disease or disorder (e.g., cancer) characterized by certain, molecular, pathological, histological, and/or clinical features. For example, biomarkers for tumor hypoxia include but are not limited to 18F-fluoromisonidazole (FMISO) tumor uptake, pimidazole uptake, 18F-fluoroazomycin arabinoside (FAZA) uptake, a nitroimidazole uptake, Copper(II)-diacetyl-bis(N4-methylthiosemicarbazone (Cu-ATSM) uptake, 19F magnetic resonance imaging of hexafluorobenzene (C6F6) uptake, 1H MRI of hexamethyldisiloxane uptake, tumor HIF-1α expression, tumor Glut-1 expression, tumor LDHA expression, tumor carbonic anhydrase IX (CA-9) expression, or lactate and/or pyruvate levels.

The “amount” or “level” of a biomarker associated with an increased clinical benefit to an individual is a detectable level in a biological sample. These can be measured by methods known to one skilled in the art and also disclosed herein. The expression level or amount of biomarker assessed can be used to determine the response to the treatment.

The terms “level of expression” or “expression level” in general are used interchangeably and generally refer to the amount of a biomarker in a biological sample. “Expression” generally refers to the process by which information (e.g., gene-encoded and/or epigenetic) is converted into the structures present and operating in the cell. Therefore, as used herein, “expression” may refer to transcription into a polynucleotide, translation into a polypeptide, or even polynucleotide and/or polypeptide modifications (e.g., posttranslational modification of a polypeptide). Fragments of the transcribed polynucleotide, the translated polypeptide, or polynucleotide and/or polypeptide modifications (e.g., posttranslational modification of a polypeptide) shall also be regarded as expressed whether they originate from a transcript generated by alternative splicing or a degraded transcript, or from a post-translational processing of the polypeptide, e.g., by proteolysis. “Expressed genes” include those that are transcribed into a polynucleotide as mRNA and then translated into a polypeptide, and also those that are transcribed into RNA but not translated into a polypeptide (for example, transfer and ribosomal RNAs).

“Elevated expression,” “elevated expression levels,” “elevated levels” and “overexpressed” refers to an increased expression or increased levels of a biomarker in an individual relative to a control, such as an individual or individuals who are not suffering from the disease or disorder (e.g., cancer) or an internal control (e.g., housekeeping biomarker). In some examples, elevated expression or overexpression is the result of gene amplification.

“Reduced expression,” “reduced expression levels,” or “reduced levels” refers to a decrease expression or decreased levels of a biomarker in an individual relative to a control, such as an individual or individuals who are not suffering from the disease or disorder (e.g., cancer) or an internal control (e.g., housekeeping biomarker).

The term “housekeeping biomarker” refers to a biomarker or group of biomarkers (e.g., polynucleotides and/or polypeptides) which are typically similarly present in all cell types. In some embodiments, the housekeeping biomarker is a “housekeeping gene.” A “housekeeping gene” refers herein to a gene or group of genes which encode proteins whose activities are essential for the maintenance of cell function and which are typically similarly present in all cell types.

H-NOX Proteins Overview of H-NOX Protein Family

Unless otherwise indicated, any wild-type or mutant H-NOX protein can be used in the compositions, kits, and methods as described herein. As used herein, an “H-NOX protein” means a protein that has an H-NOX domain (named for Heme-Nitric oxide and OXygen binding domain). An H-NOX protein may or may not contain one or more other domains in addition to the H-NOX domain. H-NOX proteins are members of a highly-conserved, well-characterized family of hemoproteins (Iyer, L. M. et al. (Feb. 3, 2003). BMC Genomics 4(1):5; Karow, D. S. et al. (Aug. 10, 2004). Biochemistry 43(31):10203-10211; Boon, E. M. et al. (2005). Nature Chem. Biol. 1:53-59; Boon, E. M. et al. (October 2005). Curr. Opin. Chem. Biol. 9(5):441-446; Boon, E. M. et al. (2005). J. Inorg. Biochem. 99(4):892-902). H-NOX proteins are also referred to as Pfam 07700 proteins or HNOB proteins (Pfam—A database of protein domain family alignments and Hidden Markov Models, Copyright (C) 1996-2006 The Pfam Consortium; GNU LGPL Free Software Foundation, Inc., 59 Temple Place—Suite 330, Boston, Mass. 02111-1307, USA). In some embodiments, an H-NOX protein has, or is predicted to have, a secondary structure that includes six alpha-helices, followed by two beta-strands, followed by one alpha-helix, followed by two beta-strands. An H-NOX protein can be an apoprotein that is capable of binding heme or a holoprotein with heme bound. An H-NOX protein can covalently or non-covalently bind a heme group. Some H-NOX proteins bind NO but not O2, and others bind both NO and O2. H-NOX domains from facultative aerobes that have been isolated bind NO but not O2. H-NOX proteins from obligate aerobic prokaryotes, C. elegans, and D. melanogaster bind NO and O2. Mammals have two H-NOX proteins: β1 and β2. An alignment of mouse, rat, cow, and human H-NOX sequences shows that these species share >99% identity. In some embodiments, the H-NOX domain of an H-NOX protein or the entire H-NOX protein is at least about any of 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 95, 97, 98, 99, or 99.5% identical to that of the corresponding region of a naturally-occurring Thermoanaerobacter tengcongensis H-NOX protein (e.g. SEQ ID NO:2) or a naturally-occurring sGC protein (e.g., a naturally-occurring sGC β1 protein). In some embodiments, the H-NOX domain of an H-NOX protein or the entire H-NOX protein is at least about any of 10-20%, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, 80-90%, 90-95%, 95-99, or 99-99.9% identical to that of the corresponding region of a naturally-occurring Thermoanaerobacter tengcongensis H-NOX protein (e.g. SEQ ID NO:2) or a naturally-occurring sGC protein (e.g., a naturally-occurring sGC β1 protein). As discussed further herein, an H-NOX protein may optionally contain one or more mutations relative to the corresponding naturally-occurring H-NOX protein. In some embodiments, the H-NOX protein includes one or more domains in addition to the H-NOX domain. In particular embodiments, the H-NOX protein includes one or more domains or the entire sequence from another protein. For example, the H-NOX protein may be a fusion protein that includes an H-NOX domain and part or all of another protein, such as albumin (e.g., human serum albumin). In some embodiments, only the H-NOX domain is present. In some embodiments, the H-NOX protein does not comprise a guanylyl cyclase domain. In some embodiments, the H-NOX protein comprises a tag; for example, a His6 tag.

Polymeric H-NOX Proteins

In some aspects, the invention provides polymeric H-NOX proteins comprising two or more H-NOX domains. The two or more H-NOX domains may be covalently linked or noncovalently linked. In some embodiments, the polymeric H-NOX protein is in the form of a dimer, a trimer, a tetramer, a pentamer, a hexamer, a heptamer, an octomer, a nanomer, or a decamer. In some embodiments, the polymeric H-NOX protein comprises homologous H-NOX domains. In some embodiments, the polymeric H-NOX protein comprises heterologous H-NOX domains; for example, the H-NOX domains may comprises amino acid variants of a particular species of H-NOX domain or may comprise H-NOX domains from different species. In some embodiments, at least one of the H-NOX domains of a polymeric H-NOX protein comprises a mutation corresponding to an L144F mutation of T. tengcongensis H-NOX. In some embodiments, at least one of the H-NOX domains of a polymeric H-NOX protein comprises a mutation corresponding to a W9F/L144F mutation of T. tengcongensis H-NOX. In some embodiments, the polymeric H-NOX proteins comprise one or more polymerization domains. In some embodiments, the polymeric H-NOX protein is a trimeric H-NOX protein. In some embodiments, the polymeric H-NOX protein comprises at least one trimerization domain. In some embodiments, the trimeric H-NOX protein comprises three T. tengcongensis H-NOX domains. In some embodiments the trimeric H-NOX domain comprises three T. tengcongensis L144F H-NOX domains. In some embodiments the trimeric H-NOX domain comprises three T. tengcongensis W9F/L144F H-NOX domains

In some aspects of the invention, the polymeric H-NOX protein comprises two or more associated monomers. The monomers may be covalently linked or noncovalently linked. In some embodiments, monomeric subunits of a polymeric H-NOX protein are produced where the monomeric subunits associate in vitro or in vivo to form the polymeric H-NOX protein. In some embodiments, the monomers comprise an H-NOX domain and a polymerization domain. In some embodiments, the polymerization domain is covalently linked to the H-NOX domain; for example, the C-terminus of the H-NOX domain is covalently linked to the N-terminus or the C-terminus of the polymerization domain. In other embodiments, the N-terminus of the H-NOX domain is covalently linked to the N-terminus or the C-terminus of the polymerization domain. In some embodiments, an amino acid spacer is covalently linked between the H-NOX domain and the polymerization domain. An “amino acid spacer” and an “amino acid linker” are used interchangeably herein. In some embodiments, at least one of the monomeric subunits of a polymeric H-NOX protein comprises a mutation corresponding to an L144F mutation of T. tengcongensis H-NOX. In some embodiments, at least one of the monomeric subunits of a polymeric H-NOX protein comprises a mutation corresponding to a W9F/L144F mutation of T. tengcongensis H-NOX. In some embodiments the polymeric H-NOX protein is a trimeric H-NOX protein. In some embodiments, the monomer of a trimeric H-NOX protein comprises an H-NOX domain and a foldon domain of T4 bacteriophage. In some embodiments, the monomer of a trimeric H-NOX protein comprises a T. tengcongensis H-NOX domain and a foldon domain. In some embodiments, the monomer of a trimeric H-NOX protein comprises a T. tengcongensis L144F H-NOX domain and a foldon domain. In some embodiments, the monomer of a trimeric H-NOX protein comprises a T. tengcongensis W9F/L144F H-NOX domain and a foldon domain. In some embodiments, the trimer H-NOX protein comprises three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain. In some embodiments, the H-NOX domain is linked to the foldon domain with an amino acid linker; for example a Gly-Ser-Gly linker. In some embodiments, at least one H-NOX domain comprises a tag. In some embodiments, at least one H-NOX domain comprises a His6 tag. In some embodiments, the His6 tag is linked to the foldon domain with an amino acid linker; for example an Arg-Gly-Ser linker. In some embodiments, all of the H-NOX domains comprise a His6 tag. In some embodiments, the trimeric H-NOX protein comprises the amino acid sequence set forth in SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:26 or SEQ ID NO:28.

The exemplary H-NOX domain from T. tengcongensis is approximately 26.7 kDal. In some embodiments, the polymeric H-NOX protein has an atomic mass greater than any of about 50 kDal, 75 kDal, 100 kDal, 125 kDal, to about 150 kDal.

The invention provides polymeric H-NOX proteins that show greater accumulation in one or more tissues in an individual compared to a corresponding monomeric H-NOX protein comprising a single H-NOX domain following administration of the H-NOX protein to the individual. A corresponding H-NOX protein refers to a monomeric form of the H-NOX protein comprising at least one of the H-NOX domains of the polymeric H-NOX protein. Tissues of preferential polymeric H-NOX accumulation include, but are not limited to tumors and tissue with damaged vasculature. In some embodiments the polymeric H-NOX protein persists in a mammal for at least about 1, 2, 3, 4, 6, 12 or 24 hours following administration of the H-NOX protein to the individual. In some embodiments the polymeric H-NOX protein persists in a mammal for about 1-2, 2-3, 3-4, 4-6, 6-12 or 12-24 hours following administration of the H-NOX protein to the individual In some embodiments, less than about 10% of the polymeric H-NOX is cleared from mammal by the kidneys within less than any of about 1 hour, 2 hours or 3 hours following administration of the H-NOX protein to the individual.

Sources of H-NOX Proteins and H-NOX Domains

H-NOX proteins and H-NOX domains from any genus or species can be used in the compositions, kits, and methods described herein. In various embodiments, the H-NOX protein or the H-NOX domains of a polymeric H-NOX protein is a protein or domain from a mammal (e.g., a primate (e.g., human, monkey, gorilla, ape, lemur, etc), a bovine, an equine, a porcine, a canine, or a feline), an insect, a yeast, or a bacteria or is derived from such a protein. Exemplary mammalian H-NOX proteins include wild-type human and rat soluble guanylate cyclase (such as the β1 subunit). Examples of H-NOX proteins include wild-type mammalian H-NOX proteins, e.g. H. sapiens, M. musculus, C. familiaris, B. Taurus, C. lupus and R. norvegicus; and wild-type non-mammalian vertebrate H-NOX proteins, e.g., X. laevis, O. latipes, O. curivatus, and F. rubripes. Examples of non-mammalian wild-type NO-binding H-NOX proteins include wild-type H-NOX proteins of D. melanogaster, A. gambiae, and M. sexta; examples of non-mammalian wild-type O2-binding H-NOX proteins include wild-type H-NOX proteins of C. elegans gcy-31, gcy-32, gcy-33, gcy-34, gcy-35, gcy-36, and gcy-37; D. melanogaster CG14885, CG14886, and CG4154; and M. sexta beta-3; examples of prokaryotic wild-type H-NOX proteins include T. tengcongensis, V. cholera, V. fischerii, N. punctiforme, D. desulfuricans, L. pneumophila 1, L. pneumophila 2, and C. acetobutylicum.

NCBI Accession numbers for exemplary H-NOX proteins include the following: Homo sapiens β1 [gi:2746083], Rattus norvegicus β1 [gi:27127318], Drosophila melangaster β1 [gi:861203], Drosophila melangaster CG14885-PA [gi:23171476], Caenorhabditis elegans GCY-35 [gi:52782806], Nostoc punctiforme [gi:23129606], Caulobacter crescentus [gi:16127222], Shewanella oneidensis [gi:24373702], Legionella pneumophila (ORF 2) [CUCGC272624], Clostridium acetobutylicum [gi:15896488], and Thermoanaerobacter tengcongensis [gi:20807169]. Canis lupus H-NOX is provided by GenBank accession DQ008576. Nucleic acid and amino acid sequences of exemplary H-NOX proteins and domains are provided in FIG. 30.

Exemplary H-NOX protein also include the following H-NOX proteins that are listed by their gene name, followed by their species abbreviation and Genbank Identifiers (such as the following protein sequences available as of May 21, 2006; May 22, 2006; May 21, 2007; or May 22, 2007, which are each hereby incorporated by reference in their entireties): Npun5905_Npu23129606, alr2278_Ana17229770, SO2144_Sone24373702, Mdeg1343_Mde23027521, VCA0720_Vch15601476, CC2992_Ccr16127222, Rsph2043_Rhsp22958463 (gi:46192757), Mmc10739_Mcsp22999020, Tar4_Tte20807169, Ddes2822_Dde23475919, CAC3243_Cac15896488, gcy-31_Ce17568389, CG14885_Dm24647455, GUCY1B3_Hs4504215, HpGCS-beta1_Hpul14245738, Gycbeta100B_Dm24651577, CG4154_Dm24646993 (gi:NP650424.2, gi:62484298), gcy-32_Ce13539160, gcy-36_Ce17568391 (gi:32566352, gi:86564713), gcy-35_Ce-17507861 (gi:71990146), gcy-37_Ce17540904 (gi:71985505), GCY1a3_Hs20535603, GCY1a2-Hs899477, or GYCa-99B_Dm729270 (gi:68067738) (Lakshminarayan et al. (2003). BMG Genomics 4:5-13). The species abbreviations used in these names include Ana—Anabaena Sp; Ccr—Caulobacter crescentus; Cac—Clostridium acetobutylicum; Dde—Desulfovibrio desulfuricans; Mcsp—Magnetococcus sp.; Mde—Microbulbifer degradans; Npu—Nostoc punctiforme; Rhsp—Rhodobacter sphaeroides; Sone—Shewanella oneidensis; Tte—Thermoanaerobacter tengcongensis; Vch—Vibrio cholerae; Ce—Caenorhabditis elegans; Dm—Drosophila melanogaster; Hpul—Hemicentrotus pulcherrimus; Hs—Homo sapiens.

Other exemplary H-NOX proteins include the following H-NOX proteins that are listed by their organism name and Pfam database accession number (such as the following protein sequences available as of May 21, 2006; May 22, 2006; May 17, 2007; May 21, 2007; or May 22, 2007, which are each hereby incorporated by reference in their entireties): Caenorhabditis briggsae Q622M5_CAEBR, Caenorhabditis briggsae Q61P44_CAEBR, Caenorhabditis briggsae Q61R54_CAEBR, Caenorhabditis briggsae Q61V90_CAEBR, Caenorhabditis briggsae Q61A94_CAEBR, Caenorhabditis briggsae Q60TP4_CAEBR, Caenorhabditis briggsae Q60M10_CAEBR, Caenorhabditis elegans GCY37_CAEEL, Caenorhabditis elegans GCY31_CAEEL, Caenorhabditis elegans GCY36_CAEEL, Caenorhabditis elegans GCY32_CAEEL, Caenorhabditis elegans GCY35_CAEEL, Caenorhabditis elegans GCY34_CAEEL, Caenorhabditis elegans GCY33_CAEEL, Oryzias curvinotus Q7T040_ORYCU, Oryzias curvinotus Q75WFO_ORYCU, Oryzias latipes P79998_ORYLA, Oryzias latipes Q7ZSZ5_ORYLA, Tetraodon nigroviridis Q4SW38_TETNG, Tetraodon nigroviridis Q4RZ94_TETNG, Tetraodon nigroviridis Q4S6K5_TETNG, Fugu rubripes Q90VY5_FUGRU, Xenopus laevis Q6INK9_XENLA, Homo sapiens Q5T8J7_HUMAN, Homo sapiens GCYA2_HUMAN, Homo sapiens GCYB2_HUMAN, Homo sapiens GCYB1_HUMAN, Gorilla gorilla Q9N1939 PRIM, Pongo pygmaeus Q5RAN8_PONPY, Pan troglodytes Q9N192_PANTR, Macaca mulatta Q9N194_MACMU, Hylobates lar Q9N191_HYLLA, Mus musculus Q8BXH3_MOUSE, Mus musculus GCYB1_MOUSE, Mus musculus Q3UTI4_MOUSE, Mus musculus Q3UH83_MOUSE, Mus musculus Q6XE41_MOUSE, Mus musculus Q80YP4_MOUSE, Rattus norvegicus Q80WX7_RAT, Rattus norvegicus Q80WX8_RAT, Rattus norvegicus Q920Q1_RAT, Rattus norvegicus Q54A43_RAT, Rattus norvegicus Q80WY0_RAT, Rattus norvegicus Q80WY4_RAT, Rattus norvegicus Q8CH85_RAT, Rattus norvegicus Q80WY5_RAT, Rattus norvegicus GCYB1_RAT, Rattus norvegicus Q8CH90_RAT, Rattus norvegicus Q91XJ7_RAT, Rattus norvegicus Q80WX9_RAT, Rattus norvegicus GCYB2_RAT, Rattus norvegicus GCYA2_RAT, Canis familiaris Q4ZHR9_CANFA, Bos taurus GCYB1_BOVIN, Sus scrofa Q4ZHR7_PIG, Gryllus bimaculatus Q59HN5_GRYBI, Manduca sexta 077106_MANSE, Manduca sexta 076340_MANSE, Apis mellifera Q5UAFO_APIME, Apis mellifera Q5FANO_APIME, Apis mellifera Q6L5L6_APIME, Anopheles gambiae str PEST Q7PYK9_ANOGA, Anopheles gambiae str PEST Q7Q9W6_ANOGA, Anopheles gambiae str PEST Q7QF31_ANOGA, Anopheles gambiae str PEST Q7PS01_ANOGA, Anopheles gambiae str PEST Q7PFY2_ANOGA, Anopheles gambiae Q7KQ93_ANOGA, Drosophila melanogaster Q24086_DROME, Drosophila melanogaster GCYH_DROME, Drosophila melanogaster GCYH_DROME, Drosophila melanogaster GCYDA_DROME, Drosophila melanogaster GCYDB_DROME, Drosophila melanogaster Q9VA09_DROME, Drosophila pseudoobscura Q29CE1_DROPS, Drosophila pseudoobscura Q296C7_DROPS, Drosophila pseudoobscura Q296C8_DROPS, Drosophila pseudoobscura Q29BU7_DROPS, Aplysia californica Q7YWK7_APLCA, Hemicentrotus pulcherrimus Q95NK5_HEMPU, Chlamydomonas reinhardtii, Q5YLC2_CHLRE, Anabaena sp Q8YUQ7_ANASP, Flavobacteria bacterium BBFL7 Q26GR89 BACT, Psychroflexus torquis ATCC 700755 Q1VQE59 FLAO, marine gamma proteobacterium HTCC2207 Q1YPJ59 GAMM, marine gamma proteobacterium HTCC2207 Q1YTK49 GAMM, Caulobacter crescentus Q9A451_CAUCR, Acidiphilium cryptum JF-5 Q2DG60_ACICY, Rhodobacter sphaeroides Q3JOU9_RHOS4, Silicibacter pomeroyi Q5LPV1_SILPO, Paracoccus denitrificans PD1222, Q3PC67_PARDE, Silicibacter sp TM1040 Q3QNY29 RHOB, Jannaschia sp Q28ML8_JANSC, Magnetococcus sp MC-1 Q3XT279 PROT, Legionella pneumophila Q5WXPO_LEGPL, Legionella pneumophila Q5WTZ5_LEGPL, Legionella pneumophila Q5X268_LEGPA, Legionella pneumophila Q5X2R2_LEGPA, Legionella pneumophila subsp pneumophila Q5ZWM9_LEGPH, Legionella pneumophila subsp pneumophila Q5ZSQ8_LEGPH, Colwellia psychrerythraea Q47Y43_COLP3, Pseudoalteromonas atlantica T6c Q3CSZ5_ALTAT, Shewanella oneidensis Q8EF49_SHEON, Saccharophagus degradans Q21E20_SACD2, Saccharophagus degradans Q21ER7_SACD2, Vibrio angustum S14 Q1ZWE59 VIBR, Vibrio vulnificus Q8DAE2_VIBVU, Vibrio alginolyticus 12G01 Q1VCP6_VIBAL, Vibrio sp DAT722 Q2FA229 VIBR, Vibrio parahaemolyticus Q87NJ1_VIBPA, Vibrio fischeri Q5E1F5_VIBF1, Vibrio vulnificus Q7MJS8_VIBVY, Photobacterium sp SKA34 Q2C6Z59 GAMM, Hahella chejuensis Q2SFY7_HAHCH, Oceanospirillum sp MED92 Q2BKV09 GAMM, Oceanobacter sp RED65 Q1NO359 GAMM, Desulfovibrio desulfuricans Q310U7_DESDG, Halothermothrix orenii H 168 Q2AIW59 FIRM, Thermoanaerobacter tengcongensis Q8RBX6_THETN, Caldicellulosiruptor saccharolyticus DSM 8903 Q2ZH17_CALSA, Clostridium acetobutylicum Q97E73_CLOAB, Alkaliphilus metalliredigenes QYMF Q3C7639 CLOT, Clostridium tetani Q899J9_CLOTE, and Clostridium beijerincki NCIMB 8052 Q2WVN0_CLOBE. These sequences are predicted to encode H-NOX proteins based on the identification of these proteins as belonging to the H-NOX protein family using the Pfam database as described herein.

Additional H-NOX proteins, H-NOX domains of polymeric H-NOX proteins, and nucleic acids, which may be suitable for use in the pharmaceutical compositions and methods described herein, can be identified using standard methods. For example, standard sequence alignment and/or structure prediction programs can be used to identify additional H-NOX proteins and nucleic acids based on the similarity of their primary and/or predicted protein secondary structure with that of known H-NOX proteins and nucleic acids. For example, the Pfam database uses defined alignment algorithms and Hidden Markov Models (such as Pfam 21.0) to categorize proteins into families, such as the H-NOX protein family (Pfam—A database of protein domain family alignments and Hidden Markov Models, Copyright (C) 1996-2006 The Pfam Consortium; GNU LGPL Free Software Foundation, Inc., 59 Temple Place—Suite 330, Boston, Mass. 02111-1307, USA). Standard databases such as the swissprot-trembl database (world-wide web at “expasy.org”, Swiss Institute of Bioinformatics Swiss-Prot group CMU—1 rue Michel Servet CH-1211 Geneva 4, Switzerland) can also be used to identify members of the H-NOX protein family. The secondary and/or tertiary structure of an H-NOX protein can be predicted using the default settings of standard structure prediction programs, such as PredictProtein (630 West, 168 Street, BB217, New York, N.Y. 10032, USA). Alternatively, the actual secondary and/or tertiary structure of an H-NOX protein can be determined using standard methods.

In some embodiments, the H-NOX domain has the same amino acid in the corresponding position as any of following distal pocket residues in T. tengcongensis H-NOX: Thr4, Ile5, Thr8, Trp9, Trp67, Asn74, Ile75, Phe78, Phe82, Tyr140, Leu144, or any combination of two or more of the foregoing. In some embodiments, the H-NOX domain has a proline or an arginine in a position corresponding to that of Pro115 or Arg135 of T. tengcongensis H-NOX, respectively, based on sequence alignment of their amino acid sequences. In some embodiments, the H-NOX domain has a histidine that corresponds to His105 of R. norvegicus β1 H-NOX. In some embodiments, the H-NOX domain has or is predicted to have a secondary structure that includes six alpha-helices, followed by two beta-strands, followed by one alpha-helix, followed by two beta-strands. This secondary structure has been reported for H-NOX proteins.

If desired, a newly identified H-NOX protein or H-NOX domain can be tested to determine whether it binds heme using standard methods. The ability of an H-NOX domain to function as an O2 carrier can be tested by determining whether the H-NOX domain binds O2 using standard methods, such as those described herein. If desired, one or more of the mutations described herein can be introduced into the H-NOX domain to optimize its characteristics as an O2 carrier. For example, one or more mutations can be introduced to alter its O2 dissociation constant, koff for oxygen, rate of heme autoxidation, NO reactivity, NO stability or any combination of two or more of the foregoing. Standard techniques such as those described herein can be used to measure these parameters.

Mutant H-NOX Proteins

As discussed further herein, an H-NOX protein or an H-NOX domain of a polymeric H-NOX protein may contain one or more mutations, such as a mutation that alters the O2 dissociation constant, the koff for oxygen, the rate of heme autoxidation, the NO reactivity, the NO stability, or any combination of two or more of the foregoing compared to that of the corresponding wild-type protein. In some embodiments, the invention provides a polymeric H-NOX protein comprising one or more H-NOX domains that may contain one or more mutations, such as a mutation that alters the O2 dissociation constant, the koff for oxygen, the rate of heme autoxidation, the NO reactivity, the NO stability, or any combination of two or more of the foregoing compared to that of the corresponding wild-type protein. Panels of engineered H-NOX domains may be generated by random mutagenesis followed by empirical screening for requisite or desired dissociation constants, dissociation rates, NO-reactivity, stability, physio-compatibility, or any combination of two or more of the foregoing in view of the teaching provided herein using techniques as described herein and, additionally, as known by the skilled artisan. Alternatively, mutagenesis can be selectively targeted to particular regions or residues such as distal pocket residues apparent from the experimentally determined or predicted three-dimensional structure of an H-NOX protein (see, for example, Boon, E. M. et al. (2005). Nature Chemical Biology 1:53-59, which is hereby incorporated by reference in its entirety, particularly with respect to the sequences of wild-type and mutant H-NOX proteins) or evolutionarily conserved residues identified from sequence alignments (see, for example, Boon E. M. et al. (2005). Nature Chemical Biology 1:53-59, which is hereby incorporated by reference in its entirety, particularly with respect to the sequences of wild-type and mutant H-NOX proteins).

In some embodiments of the invention, the mutant H-NOX protein or mutant H-NOX domain of a polymeric H-NOX protein has a sequence that differs from that of all H-NOX proteins or domains occurring in nature. In various embodiments, the amino acid sequence of the mutant protein is at least about any of 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, 95, 97, 98, 99, or 99.5% identical to that of the corresponding region of an H-NOX protein occurring in nature. In various embodiments, the amino acid sequence of the mutant protein is about 10-20%, 20-30%, 30-40%, 40-50%, 50-60%, 60-70%, 70-80%, 80-90%, 90-95%, 95-99%, or 99.5% identical to that of the corresponding region of an H-NOX protein occurring in nature. In some embodiments, the mutant protein is a protein fragment that contains at least about any of 25, 50, 75, 100, 150, 200, 300, or 400 contiguous amino acids from a full-length protein. In some embodiments, the mutant protein is a protein fragment that contains 25-50, 50-75, 75-100, 100-150, 150-200, 200-300, or 300-400 contiguous amino acids from a full-length protein. Sequence identity can be measured, for example, using sequence analysis software with the default parameters specified therein (e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705). This software program matches similar sequences by assigning degrees of homology to various amino acids replacements, deletions, and other modifications.

In some embodiments of the invention, the mutant H-NOX protein or mutant H-NOX domain of a polymeric H-NOX protein comprises the insertion of one or more amino acids (e.g., the insertion of 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids). In some embodiments of the invention, the mutant H-NOX protein or mutant H-NOX domain comprises the deletion of one or more amino acids (e.g., a deletion of N-terminal, C-terminal, and/or internal residues, such as the deletion of at least about any of 5, 10, 15, 25, 50, 75, 100, 150, 200, 300, or more amino acids or a deletion of 5-10, 10-15, 15-25, 25-50, 50-75, 75-100, 100-150, 150-200, 200-300, or 300-400 amino acids). In some embodiments of the invention, the mutant H-NOX protein or mutant H-NOX domain comprises the replacement of one or more amino acids (e.g., the replacement of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids), or combinations of two or more of the foregoing. In some embodiments, a mutant protein has at least one amino acid alteration compared to a protein occurring in nature. In some embodiments, a mutant nucleic acid sequence encodes a protein that has at least one amino acid alteration compared to a protein occurring in nature. In some embodiments, the nucleic acid is not a degenerate version of a nucleic acid occurring in nature that encodes a protein with an amino acid sequence identical to a protein occurring in nature.

In some embodiments the mutation in the H-NOX protein or H-NOX domain of a polymeric H-NOX protein is an evolutionary conserved mutations (also denoted class I mutations). Examples of class I mutations are listed in Table 1A. In Table 1A, mutations are numbered/annotated according to the sequence of human β1 H-NOX, but are analogous for all H-NOX sequences. Thus, the corresponding position in any other H-NOX protein can be mutated to the indicated residue. For example, Phe4 of human β1 H-NOX can be mutated to a tyrosine since other H-NOX proteins have a tyrosine in this position. The corresponding phenylalanine residue can be mutated to a tyrosine in any other H-NOX protein. In particular embodiments, the one or more mutations are confined to evolutionarily conserved residues. In some embodiments, the one or more mutations may include at least one evolutionarily conserved mutation and at least one non-evolutionarily conserved mutation. If desired, these mutant H-NOX proteins are subjected to empirical screening for NO/O2 dissociation constants, NO-reactivity, stability, and physio-compatibility in view of the teaching provided herein.

TABLE 1A Exemplary Class I H-NOX mutations targeting evolutionary conserved residues F4Y F4L H7G A8E L9W Q30G E33P N61G C78H A109F I145Y I145H K151E I157F E183F

In some embodiments, the mutation is a distal pocket mutation, such as mutation of a residue in alpha-helix A, D, E, or G (Pellicena, P. et al. (Aug. 31, 2004). Proc Natl. Acad Sci USA 101(35):12854-12859). Exemplary distal pocket mutations (also denoted class II mutations) are listed in Table 1B. In Table 1B, mutations are numbered/annotated according to the sequence of human β1 H-NOX, but are analogous for all H-NOX sequences. Because several substitutions provide viable mutations at each recited residue, the residue at each indicated position can be changed to any other naturally or non-naturally-occurring amino acid (denoted “X”). Such mutations can produce H-NOX proteins with a variety of desired affinity, stability, and reactivity characteristics.

TABLE 1B Exemplary Class II H-NOX mutations targeting distal pocket residues V8X L9X F70X M73X F77X C78X I145X I149X

In particular embodiments, the mutation is a heme distal pocket mutation. As described herein, a crucial molecular determinant that prevents O2 binding in NO-binding members of the H-NOX family is the lack of an H-bond donor in the distal pocket of the heme. Accordingly, in some embodiments, the mutation alters H-bonding between the H-NOX domain and the ligand within the distal pocket. In some embodiments, the mutation disrupts an H-bond donor of the distal pocket and/or imparts reduced O2 ligand-binding relative to the corresponding wild-type H-NOX domain. Exemplary distal pocket residues include Thr4, Ile5, Thr8, Trp9, Trp67, Asn74, Ile75, Phe78, Phe82, Tyr140, and Leu144 of T. tengcongensis H-NOX and the corresponding residues in any other H-NOX protein. In some embodiments, the H-NOX protein or H-NOX domain of a polymeric H-NOX protein comprises one or more distal pocket mutations. In some embodiments, the H-NOX protein or H-NOX domain of a polymeric H-NOX protein comprises one, two, three, four, five, six, seven, eight, nine, ten or more than ten distal pocket mutations. In some embodiments, the distal pocket mutation corresponds to a L144F mutation of T. tengcongensis H-NOX. In some embodiments, the distal pocket mutation is a L144F mutation of T. tengcongensis H-NOX. In some embodiments, H-NOX protein or the H-NOX domain of a polymeric H-NOX protein comprises two distal pocket mutations. In some embodiments, the H-NOX protein or H-NOX domain of a polymeric H-NOX protein corresponds to a W9F/L144F mutation of T. tengcongensis H-NOX. In some embodiments, the H-NOX protein or H-NOX domain of a polymeric H-NOX protein is a W9F/L144F mutation of T. tengcongensis H-NOX.

Residues that are not in the distal pocket can also affect the three-dimensional structure of the heme group; this structure in turn affects the binding of O2 and NO to iron in the heme group. Accordingly, in some embodiments, the H-NOX protein or H-NOX domain of a polymeric H-NOX protein has one or more mutations outside of the distal pocket. Examples of residues that can be mutated but are not in the distal pocket include Pro115 and Arg135 of T. tengcongensis H-NOX. In some embodiments, the mutation is in the proximal pocket which includes His105 as a residue that ligates to the heme iron.

In some embodiments when two or more mutations are present; at least one mutation is in the distal pocket, and at least one mutation is outside of the distal pocket (e.g., a mutation in the proximal pocket). In some embodiments, all the mutations are in the distal pocket.

To reduce the immunogenicity of H-NOX protein or H-NOX domains derived from sources other than humans, amino acids in an H-NOX protein or H-NOX domain can be mutated to the corresponding amino acids in a human H-NOX. For example, one or more amino acids on the surface of the tertiary structure of a non-human H-NOX protein or H-NOX domain can be mutated to the corresponding amino acid in a human H-NOX protein or H-NOX domain. In some variations, mutation of one or more surface amino acids may be combined with mutation of two or more distal pocket residues, mutation of one or more residues outside of the distal pocket (e.g., a mutation in the proximal pocket), or combinations of two or more of the foregoing.

The invention also relates to any combination of mutation described herein, such as double, triple, or higher multiple mutations. For example, combinations of any of the mutations described herein can be made in the same H-NOX protein. Note that mutations in equivalent positions in other mammalian or non-mammalian H-NOX proteins are also encompassed by this invention. Exemplary mutant H-NOX proteins or mutant H-NOX domains comprise one or more mutations that impart altered O2 or NO ligand-binding relative to the corresponding wild-type H-NOX domain and are operative as a physiologically compatible mammalian O2 blood gas carrier.

The residue number for a mutation indicates the position in the sequence of the particular H-NOX protein being described. For example, T. tengcongensis ISA refers to the replacement of isoleucine by alanine at the fifth position in T. tengcongensis H-NOX. The same isoleucine to alanine mutation can be made in the corresponding residue in any other H-NOX protein or H-NOX domain (this residue may or may not be the fifth residue in the sequence of other H-NOX proteins). Since the amino acid sequences of mammalian β1 H-NOX domains differ by at most two amino acids, mutations that produce desirable mutant H-NOX proteins or H-NOX domains when introduced into wild-type rat β1 H-NOX proteins are also expected to produce desirable mutant H-NOX proteins or H-NOX domains when introduced into wild-type β1 H-NOX proteins or H-NOX domains from other mammals, such as humans.

In some embodiments, the H-NOX protein is a trimer comprising three T. tengcongensis L144F H-NOX domains and three foldon domains. In some embodiments, the H-NOX protein is a trimer comprising three T. tengcongensis W9F/L144F H-NOX domains and three foldon domains. In some embodiments, the H-NOX protein is a trimer comprising three T. tengcongensis wildtype H-NOX domains and three foldon domains.

Modifications to H-NOX Proteins

Any of the wild-type or mutant H-NOX proteins, including polymeric H-NOX proteins, can be modified and/or formulated using standard methods to enhance therapeutic or industrial applications. For example, and particularly as applied to heterologous engineered H-NOX proteins, a variety of methods are known in the art for insulating such agents from immune surveillance, including crosslinking, PEGylation, carbohydrate decoration, etc. (e.g., Rohlfs, R. J. et al. (May 15, 1998). J. Biol. Chem. 273(20):12128-12134; Migita, R. et al. (June 1997). J. Appl. Physiol. 82(6):1995-2002; Vandegriff, K. D. et al. (Aug. 15, 2004). Biochem J. 382(Pt 1):183-189, which are each hereby incorporated by reference in their entireties, particularly with respect to the modification of proteins) as well as other techniques known to the skilled artisan. Fusing an H-NOX protein, including a polymeric H-NOX protein, with a human protein such as human serum albumin can increase the serum half-life, viscosity, and colloidal oncotic pressure. In some embodiments, an H-NOX protein is modified during or after its synthesis to decrease its immunogenicity and/or to increase its plasma retention time. H-NOX proteins can also be encapsulated (such as encapsulation within liposomes or nanoparticles).

In some embodiments, the H-NOX protein comprises one of more tags; e.g. to assist in purification of the H-NOX protein. Examples of tags include, but are not limited to His6, FLAG, GST, and MBP. In some embodiments, the H-NOX protein comprises one of more His6 tags. The one or more His6 tags may be removed prior to use of the polymeric H-NOX protein; e.g. by treatment with an exopeptidase. In some embodiments, the H-NOX protein is a trimer comprising three T. tengcongensis L144F H-NOX domains, three foldon domains, and three His6 tags. In some embodiments, the H-NOX protein is a trimer comprising three T. tengcongensis W9F/L144F H-NOX domains, three foldon domains, and three His6 tags. In some embodiments, the H-NOX protein is a trimer comprising three T. tengcongensis wildtype H-NOX domains, three foldon domains, and three His6 tags.

Polymerization Domains

In some aspects, the invention provides polymeric H-NOX proteins comprising two or more H-NOX domains and one or more polymerization domains. Polymerization domains are used to link two or more H-NOX domains to form a polymeric H-NOX protein. One or more polymerization domains may be used to produce dimers, trimers, tetramers, pentamers, etc. of H-NOX proteins. Polymerization domains are known in the art, such as: the foldon of T4 bacteriophage fibritin, Arc, POZ, coiled coil domains (including GCN4, leucine zippers, Velcro), uteroglobin, collagen, 3-stranded coiled colis (matrilin-1), thrombosporins, TRPV1-C, P53, Mnt, avadin, streptavidin, Bcr-Abl, COMP, verotoxin subunit B, CamKII, RCK, and domains from N ethylmaleimide-sensitive fusion protein, STM3548, KaiC, TyrR, Hcp1, CcmK4, GP41, anthrax protective antigen, aerolysin, a-hemolysin, C4b-binding protein, Mi-CK, arylsurfatase A, and viral capsid proteins. The polymerization domains may be covalently or non-covalently linked to the H-NOX domains. In some embodiments, a polymerization domain is linked to an H-NOX domain to form a monomer subunit such that the polymerization domains from a plurality of monomer subunits associate to form a polymeric H-NOX domain. In some embodiments, the C-terminus of an H-NOX domain is linked to the N-terminus of a polymerization domain. In other embodiments, the N-terminus of an H-NOX domain is linked to the N-terminus of a polymerization domain. In yet other embodiments, the C-terminus of an H-NOX domain is linked to the C-terminus of a polymerization domain. In some embodiments, the N-terminus of an H-NOX domain is linked to the C-terminus of a polymerization domain.

Linkers may be used to join a polymerization domain to an H-NOX domain; for example, for example, amino acid linkers. In some embodiments, a linker comprising any one of one, two, three, four, five, six, seven, eight, nine, ten or more than ten amino acids may be placed between the polymerization domain and the H-NOX domain. Exemplary linkers include but are not limited to Gly-Ser-Gly and Arg-Gly-Ser linkers.

Bacteriophage T4 Fibritin Trimerization Domain

An exemplary polymerization domain is the foldon domain of bacteriophage T4. The wac gene from the bacteriophage T4 encodes the fibritin protein, a 486 amino acid protein with a C-terminal trimerization domain (residues 457-483) (Efimov, V. P. et al. (1994) J Mol Biol 242:470-486). The domain is able to trimerize fibritin both in vitro and in vivo (Boudko, S. P. et al. (2002) Eur J Biochem 269:833-841; Letarov, A. V., et al., (1999) Biochemistry (Mosc)64:817-823; Tao, Y., et al., (1997) Structure 5:789-798). The isolated 27 residue trimerization domain, often referred to as the “foldon domain,” has been used to construct chimeric trimers in a number of different proteins (including HIV envelope glycoproteins (Yang, X. et al., (2002) J Virol 76:4634-4642), adenoviral adhesins (Papanikolopoulou, K., et al., (2004) J Biol Chem 279:8991-8998; Papanikolopoulou, K. et al. (2004) J Mol Biol 342:219-227), collagen (Zhang, C., et al. (2009) Biotechnol Prog 25:1660-1668), phage P22 gp26 (Bhardwaj, A., et al. (2008) Protein Sci 17:1475-1485), and rabies virus glycoprotein (Sissoeff, L., et al. (2005) J Gen Virol 86:2543-2552). An exemplary sequence of the foldon domain is provided by SEQ ID NO:4.

The isolated foldon domain folds into a single β-hairpin structure and trimerizes into a β-propeller structure involving three hairpins (Guthe, S. et al. (2004) J Mol Biol 337:905-915). The structure of the foldon domain alone has been determined by NMR (Guthe, S. et al. (2004) J Mol Biol 337:905-915) and the structures of several proteins trimerized with the foldon domain have been solved by X-ray crystallography (Papanikolopoulou, K., et al., (2004) J Biol Chem 279:8991-8998; Stetefeld, J. et al. (2003) Structure 11:339-346; Yokoi, N. et al. (2010) Small 6:1873-1879). The domain folds and trimerizes rapidly reducing the opportunity for misfolding intermediates or off-pathway oligomerization products (Guthe, S. et al. (2004) J Mol Biol 337:905-915). The foldon domain is very stable, able to maintain tertiary structure and oligomerization in >10% SDS, 6.0M guanidine hydrochloride, or 80° C. (Bhardwaj, A., et al. (2008) Protein Sci 17:1475-1485; Bhardwaj, A., et al. (2007) J Mol Biol 371:374-387) and can improve the stability of sequences fused to the foldon domain (Du, C. et al. (2008) Appl Microbiol Biotechnol 79:195-202.

In some embodiments, the C-terminus of an H-NOX domain is linked to the N-terminus of a foldon domain. In other embodiments, the N-terminus of an H-NOX domain is linked to the N-terminus of a foldon domain. In yet other embodiments, the C-terminus of an H-NOX domain is linked to the C-terminus of a foldon domain. In some embodiments, the N-terminus of an H-NOX domain is linked to the C-terminus of a foldon domain.

In some embodiments, linkers are be used to join a foldon domain to an H-NOX domain. In some embodiments, a linker comprising any one of one, two, three, four, five, six, seven, eight, nine, ten or more than ten amino acids may be placed between the polymerization domain and the H-NOX domain. Exemplary linkers include but are not limited to Gly-Ser-Gly and Arg-Gly-Ser linkers. In some embodiments, the invention provides a trimeric H-NOX protein comprising from N-terminus to C-terminus: a T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker, and a foldon domain. In some embodiments, the invention provides a trimeric H-NOX protein comprising from N-terminus to C-terminus: a T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker, a foldon domain, an Arg-Gly-Ser amino acid linker, and a His6 tag. In some embodiments, the T. tengcongensis H-NOX domain comprises an L144F mutation. In some embodiments, the T. tengcongensis H-NOX domain comprises a W9F mutation and a L144F mutation. In some embodiments, the T. tengcongensis H-NOX domain is a wild-type H-NOX domain.

Monomeric H-NOX Domain Subunits

In one aspect, the invention provides recombinant monomeric H-NOX proteins (i.e. monomeric H-NOX subunits of polymeric H-NOX proteins) that can associate to form polymeric H-NOX proteins. In some embodiments, the invention provides recombinant H-NOX proteins comprising an H-NOX domain as described herein and a polymerization domain. The H-NOX domain and the polymerization domain may be covalently linked or noncovalently linked. In some embodiments, the C-terminus of an H-NOX domain of the recombinant monomeric H-NOX protein is linked to the N-terminus of a polymerization domain. In other embodiments, the N-terminus of an H-NOX domain of the recombinant monomeric H-NOX protein is linked to the N-terminus of a polymerization domain. In yet other embodiments, the C-terminus of an H-NOX domain of the recombinant monomeric H-NOX protein is linked to the C-terminus of a polymerization domain. In some embodiments, the N-terminus of an H-NOX domain of the recombinant monomeric H-NOX protein is linked to the C-terminus of a polymerization domain. In some embodiments, the recombinant monomeric H-NOX protein does not comprise a guanylyl cyclase domain.

In some embodiments, the monomeric H-NOX protein comprises a wild-type H-NOX domain. In some embodiments of the invention, the monomeric H-NOX protein comprises one of more mutations in the H-NOX domain. In some embodiments, the one or more mutations alter the O2 dissociation constant, the koff for oxygen, the rate of heme autooxidation, the NO reactivity, the NO stability or any combination of two or more of the foregoing compared to that of the corresponding wild-type H-NOX domain. In some embodiments, the mutation is a distal pocket mutation. In some embodiments, the mutation comprises a mutation that is not in the distal pocket. In some embodiments, the distal pocket mutation corresponds to a L144 mutation of T. tengcongensis (e.g. a L144F mutation). In some embodiments, the recombinant monomeric H-NOX protein comprises two distal pocket mutations corresponding to a W9 and a L144 mutation of T. tengcongensis (e.g. a W9F/L144F mutation).

In some aspects, the invention provides recombinant monomeric H-NOX proteins that associate to form trimeric H-NOX proteins. In some embodiments, the recombinant H-NOX protein comprises an H-NOX domain and a trimerization domain. In some embodiments, the trimerization domain is a foldon domain as discussed herein. In some embodiments, the H-NOX domain is a T. tengcongensis H-NOX domain. In some embodiments the C-terminus of the T. tengcongensis H-NOX domain is covalently linked to the N-terminus of the foldon domain. In some embodiments the C-terminus of the T. tengcongensis H-NOX domain is covalently linked to the C-terminus of the foldon domain. In some embodiments, the T. tengcongensis domain is an L144F H-NOX domain. In some embodiments, the T. tengcongensis domain is a W9F/L144F H-NOX domain. In some embodiments, the T. tengcongensis domain is a wild-type H-NOX domain.

In some embodiments, the H-NOX domain is covalently linked to the polymerization domain using an amino acid linker sequence. In some embodiments, the amino acid linker sequence is one, two, three, four, five, six, seven, eight, nine, ten or more than ten amino acids in length. Exemplary amino acid linker sequences include but are not limited to a Gly-Ser-Gly sequence and an Arg-Gly-Ser sequence. In some embodiments, the polymeric H-NOX protein is a trimeric H-NOX protein comprising three H-NOX domains and three trimerization sequences wherein the H-NOX domain is covalently linked to the trimerization domain via an amino acid linker sequence. In some embodiments, the monomeric H-NOX protein comprises the following from the N-terminus to the C-terminus: an L144F T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, and a foldon domain. In some embodiments, the monomeric H-NOX protein comprises the following from the N-terminus to the C-terminus: a W9F/L144F T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, and a foldon domain. In some embodiments, the monomeric H-NOX protein comprises the following from the N-terminus to the C-terminus: a wild-type T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, and a foldon domain.

In some embodiments, the recombinant monomeric H-NOX protein comprises a tag; e.g., a His6, a FLAG, a GST, or an MBP tag. In some embodiments, the recombinant monomeric H-NOX protein comprises a His6 tag. In some embodiments, the recombinant monomeric H-NOX protein does not comprise a tag. In some embodiments, the tag (e.g. a His6 tag) is covalently linked to the polymerization domain using an amino acid spacer sequence. In some embodiments, the amino acid linker sequence is one, two, three, four, five, six, seven, eight, nine, ten or more than ten amino acids in length. Exemplary amino acid linker sequences include but are not limited to a Gly-Ser-Gly sequence and an Arg-Gly-Ser sequence. In some embodiments, the polymeric H-NOX protein is a trimeric H-NOX protein comprising three H-NOX domains, three trimerization sequences, and three His6 tags, wherein the H-NOX domain is covalently linked to the trimerization domain via an amino acid linker sequence and the trimerization domain is covalently linked to the His6 tag via an amino acid linker sequence. In some embodiments, the monomeric H-NOX protein comprises the following from the N-terminus to the C-terminus: an L144F T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, a foldon domain, an Arg-Gly-Ser linker sequence, and a His6 tag. In some embodiments, the monomeric H-NOX protein comprises the following from the N-terminus to the C-terminus: a W9F/L144F T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, a foldon domain, an Arg-Gly-Ser linker sequence, and a His6 tag. In some embodiments, the monomeric H-NOX protein comprises the following from the N-terminus to the C-terminus: a wild-type T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, a foldon domain, an Arg-Gly-Ser linker sequence, and a His6 tag.

In some embodiments the recombinant monomeric H-NOX protein comprises the amino acid sequence of SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10 or SEQ ID NO:12.

Characteristics of Wild-Type and Mutant H-NOX Proteins

As described herein, a large number of diverse H-NOX mutant proteins, including polymeric H-NOX proteins, providing ranges of NO and O2 dissociation constants, O2 koff, NO reactivity, and stability have been generated. To provide operative blood gas carriers, the H-NOX proteins may be used to functionally replace or supplement endogenous O2 carriers, such as hemoglobin. In some embodiments, H-NOX proteins such as polymeric H-NOX proteins, are used to deliver O2 to hypoxic tumor tissue (e.g. a glioblastoma) as an adjuvant to radiation therapy or chemotherapy. Accordingly, in some embodiments, an H-NOX protein has a similar or improved O2 association rate, O2 dissociation rate, dissociation constant for O2 binding, NO stability, NO reactivity, autoxidation rate, plasma retention time, or any combination of two or more of the foregoing compared to an endogenous O2 carrier, such as hemoglobin. In some embodiments, the H-NOX protein is a polymeric H-NOX protein. In some embodiments, the polymeric H-NOX protein is a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain. In some embodiments, the polymeric H-NOX protein is a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis W9F/L144F H-NOX domain and a foldon domain. In some embodiments, the polymeric H-NOX protein is a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain.

In various embodiments, the koff for O2 for an H-NOX protein, including a polymeric H-NOX protein, is between about 0.01 to about 200 s−1 at 20° C., such as about 0.1 to about 200 s−1, about 0.1 to 100 s−1, about 1.0 to about 16.0 s−1, about 1.35 to about 23.4 s−1, about 1.34 to about 18 s−1, about 1.35 to about 14.5 s−1, about 0.21 to about 23.4 s−1, about 1.35 to about 2.9 s−1, about 2 to about 3 s−1, about 5 to about 15 s−1, or about 0.1 to about 1 s−1. In some embodiments, the H-NOX protein has a koff for oxygen that is less than or equal to about 0.65 s−1 at 20° C. (such as between about 0.21 s−1 to about 0.65 s−1 at 20° C.).

In various embodiments, the kon for O2 for an H-NOX protein, including a polymeric H-NOX protein, is between about 0.14 to about 60 μM−1s−1 at 20° C., such as about 6 to about 60 μM−1s−1, about 6 to 12 μM−1s−1, about 15 to about 60 μM−1s−1, about 5 to about 18 μM−1s−1, or about 6 to about 15 μM−1s−1.

In various embodiments, the kinetic or calculated KD for O2 binding by an H-NOX protein, including a polymeric H-NOX protein, is between about 1 nM to 1 mM, about 1 μM to about 10 μM, or about 10 μM to about 50 μM. In some embodiments the calculated KD for O2 binding is any one of about 2 nM to about 2 μM, about 2 μM to about 1 mM, about 100 nM to about 1 μM, about 9 μM to about 50 μM, about 100 μM to about 1 mM, about 50 nM to about 10 μM, about 2 nM to about 50 μM, about 100 nM to about 1.9 μM, about 150 nM to about 1 μM, or about 100 nM to about 255 nM, about 20 nM to about 2 μM, 20 nM to about 75 nM, about 1 μM to about 2 μM, about 2 μM to about 10 μM, about 2 μM to about 9 μM, or about 100 nM to 500 nM at 20° C. In some embodiments, the kinetic or calculated KD for O2 binding is less than about any of 100 nM, 80 nM, 50 nM, 30 nM, 25 nM, 20 nM, or 10 nM at 20° C.

In various embodiments, the kinetic or calculated KD for O2 binding by an H-NOX protein, including a polymeric H-NOX protein, is within about 0.01 to about 100-fold of that of hemoglobin under the same conditions (such as at 20° C.), such as between about 0.1 to about 10-fold or between about 0.5 to about 2-fold of that of hemoglobin under the same conditions (such as at 20° C.). In various embodiments, the kinetic or calculated KD for NO binding by an H-NOX protein is within about 0.01 to about 100-fold of that of hemoglobin under the same conditions (such as at 20° C.), such as between about 0.1 to about 10-fold or between about 0.5 to about 2-fold of that of hemoglobin under the same conditions (such as at 20° C.).

In some embodiments, less than about any of 50, 40, 30, 10, or 5% of an H-NOX protein, including a polymeric H-NOX protein, is oxidized after incubation for about any of 1, 2, 4, 6, 8, 10, 15, or 20 hours at 20° C.

In various embodiments, the NO reactivity of an H-NOX protein, including a polymeric H-NOX protein, is less than about 700 s−1 at 20° C., such as less than about 600 s−1, 500 s−1, 400 s−1, 300 s−1, 200 s−1, 100 s−1, 75 s−1, 50 s−1, 25 s−1, 20 s−1, 10 s−1, 50 s−1, 3 s−1, 2 s−1, 1.8 s−1, 1.5 s−1, 1.2 s−1, 1.0 s−1, 0.8 s−1, 0.7 s−1, or 0.6 s−1 at 20° C. In various embodiments, the NO reactivity of an H-NOX protein is between about 0.1 to about 600 s−1 at 20° C., such as between about 0.5 to about 400 s−1, about 0.5 to about 100 s−1, about 0.5 to about 50 s−1, about 0.5 to about 10 s−1, about 1 to about 5 s−1, or about 0.5 to about 2.1 s−1 at 20° C. In various embodiments, the reactivity of an H-NOX protein is at least about 10, 100, 1,000, or 10,000 fold lower than that of hemoglobin under the same conditions, such as at 20° C.

In various embodiments, the rate of heme autoxidation of an H-NOX protein, including a polymeric H-NOX protein, is less than about 1.0 h−1 at 37° C., such as less than about any of 0.9 h−1, 0.8 h−1, 0.7 h−1, 0.6 h−1, 0.5 h−1, 0.4 h−1, 0.3 h−1, 0.2 h−1, 0.1 h−1, or 0.05 h−1 at 37 C. In various embodiments, the rate of heme autoxidation of an H-NOX protein is between about 0.006 to about 5.0 h−1 at 37° C., such as about 0.006 to about 1.0 h−1, 0.006 to about 0.9 h−1, or about 0.06 to about 0.5 h−1 at 37° C.

In various embodiments, a mutant H-NOX protein, including a polymeric H-NOX protein, has (a) an O2 or NO dissociation constant, association rate (kon for O2 or NO), or dissociation rate (koff for O2 or NO) within 2 orders of magnitude of that of hemoglobin, (b) has an NO affinity weaker (e.g., at least about 10-fold, 100-fold, or 1000-fold weaker) than that of sGC β1, respectively, (c) an NO reactivity with bound O2 at least 1000-fold less than hemoglobin, (d) an in vivo plasma retention time at least 2, 10, 100, or 1000-fold higher than that of hemoglobin, or (e) any combination of two or more of the foregoing.

Exemplary suitable O2 carriers provide dissociation constants within two orders of magnitude of that of hemoglobin, i.e. between about 0.01 and 100-fold, such as between about 0.1 and 10-fold, or between about 0.5 and 2-fold of that of hemoglobin. A variety of established techniques may be used to quantify dissociation constants, such as the techniques described herein (Boon, E. M. et al. (2005). Nature Chem. Biol. 1:53-59; Boon, E. M. et al. (October 2005). Curr. Opin. Chem. Biol. 9(5):441-446; Boon, E. M. et al. (2005). J. Inorg. Biochem. 99(4):892-902), Vandegriff, K. D. et al. (Aug. 15, 2004). Biochem J. 382(Pt 1):183-189, which are each hereby incorporated by reference in their entireties, particularly with respect to the measurement of dissociation constants), as well as those known to the skilled artisan. Exemplary O2 carriers provide low or minimized NO reactivity of the H-NOX protein with bound O2, such as an NO reactivity lower than that of hemoglobin. In some embodiments, the NO reactivity is much lower, such as at least about 10, 100, 1,000, or 10,000-fold lower than that of hemoglobin. A variety of established techniques may be used to quantify NO reactivity (Boon, E. M. et al. (2005). Nature Chem. Biol. 1:53-59; Boon, E. M. et al. (October 2005). Curr. Opin. Chem. Biol. 9(5):441-446; Boon, E. M. et al. (2005). J. Inorg. Biochem. 99(4):892-902), Vandegriff, K. D. et al. (Aug. 15, 2004). Biochem J. 382(Pt 1):183-189, which are each hereby incorporated by reference in their entireties, particularly with respect to the measurement of NO reactivity) as well as those known to the skilled artisan. Because wild-type T. tengcongensis H-NOX has such a low NO reactivity, other wild-type H-NOX proteins and mutant H-NOX proteins may have a similar low NO reactivity. For example, T. tengcongensis H-NOX Y140H has an NO reactivity similar to that of wild-type T. tengcongensis H-NOX.

In addition, suitable O2 carriers provide high or maximized stability, particularly in vivo stability. A variety of stability metrics may be used, such as oxidative stability (e.g., stability to autoxidation or oxidation by NO), temperature stability, and in vivo stability. A variety of established techniques may be used to quantify stability, such as the techniques described herein (Boon, E. M. et al. (2005). Nature Chem. Biol. 1:53-59; Boon, E. M. et al. (October 2005). Curr. Opin. Chem. Biol. 9(5):441-446; Boon, E. M. et al. (2005). J. Inorg. Biochem. 99(4):892-902), as well as those known to the skilled artisan. For in vivo stability in plasma, blood, or tissue, exemplary metrics of stability include retention time, rate of clearance, and half-life. H-NOX proteins from thermophilic organisms are expected to be stable at high temperatures. In various embodiments, the plasma retention times are at least about 2-, 10-, 100-, or 1000-fold greater than that of hemoglobin (e.g. Bobofchak, K. M. et al. (August 2003). Am. J. Physiol. Heart Circ. Physiol. 285(2):H549-H561). As will be appreciated by the skilled artisan, hemoglobin-based blood substitutes are limited by the rapid clearance of cell-free hemoglobin from plasma due the presence of receptors for hemoglobin that remove cell-free hemoglobin from plasma. Since there are no receptors for H-NOX proteins in plasma, wild-type and mutant H-NOX proteins are expected to have a longer plasma retention time than that of hemoglobin. If desired, the plasma retention time can be increased by PEGylating or crosslinking an H-NOX protein or fusing an H-NOX protein with another protein using standard methods (such as those described herein and those known to the skilled artisan).

In various embodiments, the H-NOX protein, including a polymeric H-NOX protein, has an O2 dissociation constant between about 1 nM to about 1 mM at 20° C. and a NO reactivity at least about 10-fold lower than that of hemoglobin under the same conditions, such as at 20° C. In some embodiments, the H-NOX protein has an O2 dissociation constant between about 1 nM to about 1 mM at 20° C. and a NO reactivity less than about 700 s−1 at 20° C. (e.g., less than about 600 s−1, 500 s−1, 100 s−1, 20 s−1, or 1.8 s−1 at 20° C.). In some embodiments, the H-NOX protein has an O2 dissociation constant within 2 orders of magnitude of that of hemoglobin and a NO reactivity at least about 10-fold lower than that of hemoglobin under the same conditions, such as at 20° C. In some embodiments, the H-NOX protein has a koff for oxygen between about 0.01 to about 200 s−1 at 20° C. and an NO reactivity at least about 10-fold lower than that of hemoglobin under the same conditions, such as at 20° C. In some embodiments, the H-NOX protein has a koff for oxygen that is less than about 0.65 s−1 at 20° C. (such as between about 0.21 s−1 to about 0.64 s−1 at 20° C.) and a NO reactivity at least about 10-fold lower than that of hemoglobin under the same conditions, such as at 20° C. In some embodiments of the invention, the O2 dissociation constant of the H-NOX protein is between about 1 nM to about 1 μM (1000 nM), about 1 μM to about 10 μM, or about 10 μM to about 50 μM. In particular embodiments, the O2 dissociation constant of the H-NOX protein is between about 2 nM to about 50 μM, about 50 nM to about 10 μM, about 100 nM to about 1.9 μM, about 150 nM to about 1 μM, or about 100 nM to about 255 nM at 20° C. In various embodiments, the O2 dissociation constant of the H-NOX protein is less than about 80 nM at 20° C., such as between about 20 nM to about 75 nM at 20° C. In some embodiments, the NO reactivity of the H-NOX protein is at least about 100-fold lower or about 1,000 fold lower than that of hemoglobin, under the same conditions, such as at 20° C. In some embodiments, the NO reactivity of the H-NOX protein is less than about 700 s−1 at 20° C., such as less than about 600 s−1, 500 s−1, 400 s−1, 300 s−1, 200 s−1, 100 s−1, 75 s−1, 50 s−1, 25 s−1, 20 s−1, 10 s−1, 50 s−1, 3 s−1, 2 s−1, 1.8 s−1, 1.5 s−1, 1.2 s−1, 1.0 s−1, 0.8 s−1, 0.7 s−1, or 0.6 s−1 at 20° C. In some embodiments, the koff for oxygen of the H-NOX protein is between 0.01 to 200 s−1 at 20° C., such as about 0.1 to about 200 s−1, about 0.1 to 100 s−1, about 1.35 to about 23.4 s−1, about 1.34 to about 18 s−1, about 1.35 to about 14.5 s−1, about 0.21 to about 23.4 s−1, about 2 to about 3 s−1, about 5 to about 15 s−1, or about 0.1 to about 1 s−1. In some embodiments, the O2 dissociation constant of the H-NOX protein is between about 100 nM to about 1.9 μM at 20° C., and the koff for oxygen of the H-NOX protein is between about 1.35 s−1 to about 14.5 s−1 at 20° C. In some embodiments, the rate of heme autoxidation of the H-NOX protein is less than about 1 h−1 at 37° C., such as less than about any of 0.9 h−1, 0.8 h−1, 0.7 h−1, 0.6 h−1, 0.5 h−1, 0.4 h−1, 0.3 h−1, 0.2 h−1, or 0.1 h−1. In some embodiments, the koff for oxygen of the H-NOX protein is between about 1.35 s−1 to about 14.5 s−1 at 20° C., and the rate of heme autoxidation of the H-NOX protein is less than about 1 h−1 at 37° C. In some embodiments, the koff for oxygen of the H-NOX protein is between about 1.35 s−1 to about 14.5 s−1 at 20° C., and the NO reactivity of the H-NOX protein is less than about 700 s−1 at 20° C. (e.g., less than about 600 s−1, 500 s−1, 100 s−1, 20 s−1, or 1.8 s−1 at 20° C.). In some embodiments, the rate of heme autoxidation of the H-NOX protein is less than about 1 h−1 at 37° C., and the NO reactivity of the H-NOX protein is less than about 700 s−1 at 20° C. (e.g., less than about 600 s−1, 500 s−1, 100 s−1, 20 s−1, or 1.8 s−1 at 20° C.).

In some embodiments, the viscosity of the H-NOX protein solution, including a polymeric H-NOX protein solution, is between 1 and 4 centipoise (cP). In some embodiments, the colloid oncotic pressure of the H-NOX protein solution is between 20 and 50 mm Hg.

Measurement of O2 and/or NO Binding

One skilled in the art can readily determine the oxygen and nitric oxide binding characteristics of any H-NOX protein including a polymeric H-NOX protein such as a trimeric H-NOX protein by methods known in the art and by the non-limiting exemplary methods described below.

Kinetic KD: Ratio of koff to kon

The kinetic KD value is determined for wild-type and mutant H-NOX proteins, including polymeric H-NOS proteins, essentially as described by Boon, E. M. et al. (2005). Nature Chemical Biology 1:53-59, which is hereby incorporated by reference in its entirety, particularly with respect to the measurement of O2 association rates, O2 dissociation rates, dissociation constants for O2 binding, autoxidation rates, and NO dissociation rates.

kon (O2 Association Rate)

O2 association to the heme is measured using flash photolysis at 20° C. It is not possible to flash off the FeII—O2 complex as a result of the very fast geminate recombination kinetics; thus, the Fe11—CO complex is subjected to flash photolysis with laser light at 560 nm (Hewlett-Packard, Palo Alto, Calif.), producing the 5-coordinate FeII intermediate, to which the binding of molecular O2 is followed at various wavelengths. Protein samples are made by anaerobic reduction with 10 mM dithionite, followed by desalting on a PD-10 column (Millipore, Inc., Billerica, Mass.). The samples are then diluted to 20 μM heme in 50 mM TEA, 50 mM NaCl, pH 7.5 buffer in a controlled-atmosphere quartz cuvette, with a size of 100 μL to 1 mL and a path-length of 1-cm. CO gas is flowed over the headspace of this cuvette for 10 minutes to form the FeII—CO complex, the formation of which is verified by UV-visible spectroscopy (Soret maximum 423 nm). This sample is then either used to measure CO-rebinding kinetics after flash photolysis while still under 1 atmosphere of CO gas, or it is opened and stirred in air for 30 minutes to fully oxygenate the buffer before flash photolysis to watch O2-rebinding events. O2 association to the heme is monitored at multiple wavelengths versus time. These traces are fit with a single exponential using Igor Pro software (Wavemetrics, Inc., Oswego, Oreg.; latest 2005 version). This rate is independent of observation wavelength but dependent on O2 concentration. UV-visible spectroscopy is used throughout to confirm all the complexes and intermediates (Cary 3K, Varian, Inc. Palo Alto, Calif.). Transient absorption data are collected using instruments described in Dmochowski, I. J. et al. (Aug. 31, 2000). J Inorg Biochem. 81(3):221-228, which is hereby incorporated by reference in its entirety, particularly with respect to instrumentation. The instrument has a response time of 20 ns, and the data are digitized at 200 megasamples s−1.

koff (O2 Dissociation Rate)

To measure the koff, FeII—O2 complexes of protein (5 μM heme), are diluted in anaerobic 50 mM TEA, 50 mM NaCl, pH 7.5 buffer, and are rapidly mixed with an equal volume of the same buffer (anaerobic) containing various concentrations of dithionite and/or saturating CO gas. Data are acquired on a HI-TECH Scientific SF-61 stopped-flow spectrophotometer equipped with a Neslab RTE-100 constant-temperature bath set to 20° C. (TGK Scientific LTD., Bradford on Avon, United Kingdom). The dissociation of O2 from the heme is monitored as an increase in the absorbance at 437 nm, a maximum in the FeII—FeII—O2 difference spectrum, or 425 nm, a maximum in the FeII—FeII—CO difference spectrum. The final traces are fit to a single exponential using the software that is part of the instrument. Each experiment is done a minimum of six times, and the resulting rates are averaged. The dissociation rates measured are independent of dithionite concentration and independent of saturating CO as a trap for the reduced species, both with and without 10 mM dithionite present.

Kinetic KD

The kinetic KD is determined by calculating the ratio of koff to kon using the measurements of koff and kon described above.

Calculated KD

To measure the calculated KD, the values for the koff and kinetic KD that are obtained as described above are graphed. A linear relationship between koff and kinetic KD is defined by the equation (y=mx+b). koff values were then interpolated along the line to derive the calculated KD using Excel: MAC 2004 (Microsoft, Redmond, Wash.). In the absence of a measured kon, this interpolation provides a way to relate koff to KD.

Rate of Autoxidation

To measure the rate of autoxidation, the protein samples are anaerobically reduced, then diluted to 5 μM heme in aerobic 50 mM TEA, 50 mM NaCl, pH 7.5 buffer. These samples are then incubated in a Cary 3E spectrophotometer equipped with a Neslab RTE-100 constant-temperature bath set to 37° C. and scanned periodically (Cary 3E, Varian, Inc., Palo Alto, Calif.). The rate of autoxidation is determined from the difference between the maximum and minimum in the FeIII—FeII difference spectrum plotted versus time and fit with a single exponential using Excel: MAC 2004 (Microsoft, Redmond, Wash.).

Rate of Reaction with NO

NO reactivity is measured using purified proteins (H-NOX, polymeric H-NOX, Homo sapiens hemoglobin (Hs Hb) etc.) prepared at 2 μM in buffer A and NO prepared at 200 μM in Buffer A (Buffer A: 50 mM Hepes, pH 7.5, 50 mM NaCl). Data are acquired on a HI-TECH Scientific SF-61 stopped-flow spectrophotometer equipped with a Neslab RTE-100 constant-temperature bath set to 20° C. (TGK Scientific LTD., Bradford on Avon, United Kingdom). The protein is rapidly mixed with NO in a 1:1 ratio with an integration time of 0.00125 sec. The wavelengths of maximum change are fit to a single exponential using the software that is part of the spectrometer, essentially measuring the rate-limiting step of oxidation by NO. The end products of the reaction are ferric-NO for the HNOX proteins and ferric-aquo for Hs Hb.

p50 Measurements

If desired, the p50 value for mutant or wild-type H-NOX proteins can be measured as described by Guarnone, R. et al. (September/October 1995). Haematologica 80(5):426-430, which is hereby incorporated by reference in its entirety, particularly with respect to the measurement of p50 values. The p50 value is determined using a HemOx analyzer. The measurement chamber starts at 0% oxygen and slowly is raised, incrementally, towards 100% oxygen. An oxygen probe in the chamber measures the oxygen saturation %. A second probe (UV-Vis light) measures two wavelengths of absorption, tuned to the alpha and beta peaks of the hemoprotein's (e.g., a protein such as H-NOX complexed with heme) UV-Vis spectra. These absorption peaks increase linearly as hemoprotein binds oxygen. The percent change from unbound to 100% bound is then plotted against the % oxygen values to generate a curve. The p50 is the point on the curve where 50% of the hemoprotein is bound to oxygen.

Specifically, the Hemox-Analyzer (TCS Scientific Corporation, New Hope, Pa.) determines the oxyhemoprotein dissociation curve (ODC) by exposing 50 μL of blood or hemoprotein to an increasing partial pressure of oxygen and deoxygenating it with nitrogen gas. A Clark oxygen electrode detects the change in oxygen tension, which is recorded on the x-axis of an x-y recorder. The resulting increase in oxyhemoprotein fraction is simultaneously monitored by dual-wavelength spectrophotometry at 560 nm and 576 nm and displayed on the y-axis. Blood samples are taken from the antemedial vein, anticoagulated with heparin, and kept at 4° C. on wet ice until the assay. Fifty μL of whole blood are diluted in 5 μL of Hemox-solution, a manufacturer-provided buffer that keeps the pH of the solution at a value of 7.4±0.01. The sample-buffer is drawn into a cuvette that is part of the Hemox-Analyzer and the temperature of the mixture is equilibrated and brought to 37° C.; the sample is then oxygenated to 100% with air. After adjustment of the pO2 value the sample is deoxygenated with nitrogen; during the deoxygenation process the curve is recorded on graph paper. The P50 value is extrapolated on the x-axis as the point at which O2 saturation is 50% using the software that is part of the Hemox-Analyzer. The time required for a complete recording is approximately 30 minutes.

H-NOX Nucleic Acids

The invention also features nucleic acids encoding any of the mutant H-NOX proteins, polymeric H-NOX, or recombinant monomer H-NOX protein subunits as described herein.

In particular embodiments, the nucleic acid includes a segment of or the entire nucleic acid sequence of any of nucleic acids encoding an H-NOX protein or an H-NOX domain. In some embodiments, the nucleic acid includes at least about 50, 100, 150, 200, 300, 400, 500, 600, 700, 800, or more contiguous nucleotides from a H-NOX nucleic acid and contains one or more mutations (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 mutations) compared to the H-NOX nucleic acid from which it was derived. In various embodiments, a mutant H-NOX nucleic acid contains less than about 20, 15, 12, 10, 9, 8, 7, 6, 5, 4, 3, or 2 mutations compared to the H-NOX nucleic acid from which it was derived. The invention also features degenerate variants of any nucleic acid encoding a mutant H-NOX protein.

In some embodiments, the nucleic acid includes nucleic acids encoding two or more H-NOX domains. In some embodiments, the nucleic acids including two or more H-NOX domains are linked such that a polymeric H-NOX protein is expressed from the nucleic acid. In further embodiments, the nucleic acid includes nucleic acids encoding one or more polymerization domains. In some embodiments, the nucleic acids including the two or more H-NOX domains and the one or more polymerization domains are linked such that a polymeric H-NOX protein is expressed from the nucleic acid.

In some embodiments, the nucleic acid includes a segment or the entire nucleic acid sequence of any nucleic acid encoding a polymerization domain. In some embodiments the nucleic acid comprises a nucleic acid encoding an H-NOX domain and a polymerization domain. In some embodiments, the nucleic acid encoding an H-NOX domain and the nucleic acid encoding a polymerization domain a linked such that the produced polypeptide is a fusion protein comprising an H-NOX domain and a polymerization domain.

In some embodiments, the nucleic acid comprises nucleic acid encoding one or more His6 tags. In some embodiments the nucleic acid further comprised nucleic acids encoding linker sequences positioned between nucleic acids encoding the H-NOX domain, the polymerization domain and/or a His6 tag.

In some embodiments, the invention provides a nucleic acid encoding an H-NOX domain and a foldon domain. In some embodiments, the H-NOX domain is a T. thermoanaerobacter H-NOX domain. In some embodiments, the H-NOX domain is a wild-type T. thermoanaerobacter H-NOX domain. In some embodiments, the H-NOX domain is a T. thermoanaerobacter L144F H-NOX domain. In some embodiments, the H-NOX domain is a T. thermoanaerobacter W9F/L144F H-NOX domain.

In some embodiments, the invention provides nucleic acids encoding the following 5′ to 3′: a L144F T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, and a foldon domain. In some embodiments, the invention provides nucleic acids encoding the following 5′ to 3′: a W9F/L144F T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, and a foldon domain. In some embodiments, the invention provides nucleic acids encoding the following 5′ to 3′: a wild-type T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, and a foldon domain.

In some embodiments, the invention provides nucleic acids encoding the following 5′ to 3′: a L144F T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, a foldon domain, an Arg-Gly-Ser linker sequence, and a His6 tag. In some embodiments, the invention provides nucleic acids encoding the following 5′ to 3′: a W9F/L144F T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, a foldon domain, an Arg-Gly-Ser linker sequence, and a His6 tag. In some embodiments, the invention provides nucleic acids encoding the following 5′ to 3′: a wild-type T. tengcongensis H-NOX domain, a Gly-Ser-Gly amino acid linker sequence, a foldon domain, an Arg-Gly-Ser linker sequence, and a His6 tag.

In some embodiments, the nucleic acid comprises the nucleic acid sequence set forth in SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9 or SEQ ID NO:11.

The invention also includes a cell or population of cells containing at least one nucleic acid encoding a mutant H-NOX protein described herein. Exemplary cells include insect, plant, yeast, bacterial, and mammalian cells. These cells are useful for the production of mutant H-NOX proteins using standard methods, such as those described herein.

In some embodiments, the invention provides a cell comprising a nucleic acid encoding an H-NOX domain and a foldon domain. In some embodiments, the H-NOX domain is a T. thermoanaerobacter H-NOX domain. In some embodiments, the H-NOX domain is a wild-type T. thermoanaerobacter H-NOX domain. In some embodiments, the H-NOX domain is a T. thermoanaerobacter L144F H-NOX domain. In some embodiments, the H-NOX domain is a T. thermoanaerobacter W9F/L144F H-NOX domain. In some embodiments, the invention provides a cell comprising a nucleic acid comprising the nucleic acid sequence set forth in SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, or SEQ ID NO:11.

Formulations of H-NOX Proteins

Any wild-type or mutant H-NOX protein, including polymeric H-NOX proteins, described herein may be used for the formulation of pharmaceutical or non-pharmaceutical compositions. In some embodiments, the formulations comprise a monomeric H-NOX protein comprising an H-NOX domain and a polymerization domain such that the monomeric H-NOX proteins associate in vitro or in vivo to produce a polymeric H-NOX protein. As discussed further below, these formulations are useful in a variety of therapeutic and industrial applications.

In some embodiments, the pharmaceutical composition includes one or more wild-type or mutant H-NOX proteins described herein including polymeric H-NOX proteins and a pharmaceutically acceptable carrier or excipient. Examples of pharmaceutically acceptable carriers or excipients include, but are not limited to, any of the standard pharmaceutical carriers or excipients such as phosphate buffered saline solutions, water, emulsions such as oil/water emulsion, and various types of wetting agents. Exemplary diluents for aerosol or parenteral administration are phosphate buffered saline or normal (0.9%) saline. Compositions comprising such carriers are formulated by well-known conventional methods (see, for example, Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990; and Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing, 2000, which are each hereby incorporated by reference in their entireties, particularly with respect to formulations). In some embodiments, the formulations are sterile. In some embodiments, the formulations are essentially free of endotoxin.

While any suitable carrier known to those of ordinary skill in the art may be employed in the pharmaceutical compositions of this invention, the type of carrier will vary depending on the mode of administration. Compositions can be formulated for any appropriate manner of administration, including, for example, intravenous, intra-arterial, intravesicular, inhalation, intraperitoneal, intrapulmonary, intramuscular, subcutaneous, intra-tracheal, transmucosal, intraocular, intrathecal, or transdermal administration. For parenteral administration, such as subcutaneous injection, the carrier may include, e.g., water, saline, alcohol, a fat, a wax, or a buffer. For oral administration, any of the above carriers or a solid carrier, such as mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, glucose, sucrose, or magnesium carbonate, may be employed. Biodegradable microspheres (e.g., polylactate polyglycolate) may also be used as carriers.

In some embodiments, the pharmaceutical or non-pharmaceutical compositions include a buffer (e.g., neutral buffered saline, phosphate buffered saline, etc), a carbohydrate (e.g., glucose, mannose, sucrose, dextran, etc.), an antioxidant, a chelating agent (e.g., EDTA, glutathione, etc.), a preservative, another compound useful for binding and/or transporting oxygen, an inactive ingredient (e.g., a stabilizer, filler, etc.), or combinations of two or more of the foregoing. In some embodiments, the composition is formulated as a lyophilizate. H-NOX proteins may also be encapsulated within liposomes or nanoparticles using well known technology. Other exemplary formulations that can be used for H-NOX proteins are described by, e.g., U.S. Pat. Nos. 6,974,795, and 6,432,918, which are each hereby incorporated by reference in their entireties, particularly with respect to formulations of proteins.

The compositions described herein may be administered as part of a sustained release formulation (e.g., a formulation such as a capsule or sponge that produces a slow release of compound following administration). Such formulations may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site. Sustained-release formulations may contain an H-NOX protein dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane. Carriers for use within such formulations are biocompatible, and may also be biodegradable. In some embodiments, the formulation provides a relatively constant level of H-NOX protein release. The amount of H-NOX protein contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release, and the nature of the condition to be treated or prevented.

In some embodiments, the pharmaceutical composition contains an effective amount of a wild-type or mutant H-NOX protein. In some embodiments, the pharmaceutical composition contains an effective amount of a polymeric H-NOX protein comprising two or more wild-type or mutant H-NOX domains. In some embodiments, the pharmaceutical composition contains an effective amount of a recombinant monomeric H-NOX protein comprising a wild-type or mutant H-NOX domain and a polymerization domain as described herein. In some embodiments, the formulation comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain. In some embodiments, the formulation comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis W9F/L144F H-NOX domain and a foldon domain. In some embodiments, the formulation comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain.

An exemplary dose of hemoglobin as a blood substitute is from about 10 mg to about 5 grams or more of extracellular hemoglobin per kilogram of patient body weight. Thus, in some embodiments, an effective amount of an H-NOX protein for administration to a human is between a few grams to over about 350 grams. Other exemplary doses of an H-NOX protein include about any of 4.4, 5, 10, or 13 G/DL (where G/DL is the concentration of the H-NOX protein solution prior to infusion into the circulation) at an appropriate infusion rate, such as about 0.5 ml/min (see, for example, Winslow, R. Chapter 12 in Blood Substitutes). It will be appreciated that the unit content of active ingredients contained in an individual dose of each dosage form need not in itself constitute an effective amount since the necessary effective amount could be reached by the combined effect of a plurality of administrations. The selection of the amount of an H-NOX protein to include in a pharmaceutical composition depends upon the dosage form utilized, the condition being treated, and the particular purpose to be achieved according to the determination of the ordinarily skilled artisan in the field.

Exemplary compositions include genetically engineered, recombinant H-NOX proteins, which may be isolated or purified, comprising one or more mutations that collectively impart altered O2 or NO ligand-binding relative to the corresponding wild-type H-NOX protein, and operative as a physiologically compatible mammalian blood gas carrier. For example, mutant H-NOX proteins as described herein. In some embodiments, the H-NOX protein is a polymeric H-NOX protein. In some embodiments, the H-NOX protein is a recombinant monomeric H-NOX protein comprising a wild-type or mutant H-NOX domain and a polymerization domain as described herein. In some embodiments, the composition comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain. In some embodiments, the composition comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis W9F/L144F H-NOX domain and a foldon domain. In some embodiments, the composition comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain.

To reduce or prevent an immune response in human subjects who are administered a pharmaceutical composition, human H-NOX proteins or domains (either wild-type human proteins or human proteins into which one or more mutations have been introduced) or other non-antigenic H-NOX proteins or domains (e.g., mammalian H-NOX proteins) can be used. To reduce or eliminate the immunogenicity of H-NOX proteins derived from sources other than humans, amino acids in an H-NOX protein or H-NOX domain can be mutated to the corresponding amino acids in a human H-NOX. For example, one or more amino acids on the surface of the tertiary structure of a non-human H-NOX protein can be mutated to the corresponding amino acid in a human H-NOX protein.

Therapeutic Applications of H-NOX Proteins

Any of the wild-type or mutant H-NOX proteins, including polymeric H-NOX proteins, or pharmaceutical compositions described herein may be used in therapeutic applications.

Particular H-NOX proteins, including polymeric H-NOX proteins, can be selected for such applications based on the desired O2 association rate, O2 dissociation rate, dissociation constant for O2 binding, NO stability, NO reactivity, autoxidation rate, plasma retention time, or any combination of two or more of the foregoing for the particular indication being treated. H-NOX proteins can be used to treat cardiovascular disease, neurological disease, tumor hypoxia, loss of blood, or wounds. For example, an O2-binding H-NOX protein can be used in most situations where red blood cells or plasma expanders are currently utilized.

H-NOX proteins, including polymeric H-NOX proteins, can be used as an adjunct with radiation or chemotherapy for the treatment of cancer. In some embodiments, an H-NOX protein is used as a radiation therapy adjuvant in solid tumors (e.g., individuals with poor pre-metastatic prognoses) or as a PDT therapy adjuvant in surface tumors (e.g., colon, lung, or skin cancer, or cancer in another accessible surface or location). H-NOX proteins can be used to treat anemia by providing additional oxygen-carrying capacity in a patient who is suffering from anemia. Exemplary neurological indications include ischemic stroke, traumatic brain injury, and spinal cord injury. The methods and compositions are applicable to both acute (providing rapid oxygen to tissues or a specific site, e.g. acute myocardial infarction, acute local or systemic tissue oxygenation, or blood transfusion), and chronic situations (e.g. post-acute recovery from cardiac infarction).

In a particular aspect, the invention provides methods of using H-NOX proteins to deliver O2 to brain tumors (e.g. a glioblastoma). In some embodiments, the administration of H-NOX is used as an adjunct to radiation therapy or chemotherapy. In some embodiments, the invention provides methods to treat a brain cancer (e.g. a glioblastoma) in an individual by administering an effective amount of an H-NOX protein and administering an effective amount of radiation to the individual. In some embodiments, the invention provides methods to reduce brain tumor growth (e.g. glioblastoma growth) in an individual by administering an effective amount of an H-NOX protein and administering an effective amount of radiation to the individual. In some embodiments, the H-NOX protein is a polymeric H-NOX protein (e.g. a trimeric H-NOX protein). In some embodiments, the polymeric H-NOX protein comprises one or more H-NOX domains comprising a mutation at a position corresponding to L144 of T. tengcongensis H-NOX. In some embodiments, the polymeric H-NOX protein comprises one or more H-NOX domains comprising a mutation corresponding to a L144F mutation of T. tengcongensis H-NOX. In some embodiments, the polymeric H-NOX protein comprises one or more H-NOX domains comprising a mutation at positions corresponding to W9 and L144 of T. tengcongensis H-NOX. In some embodiments, the polymeric H-NOX protein comprises one or more H-NOX domains comprising mutations corresponding to a W9F/L144F mutation of T. tengcongensis H-NOX. In some embodiments, the H-NOX domain is a human H-NOX domain. In some embodiments, the H-NOX domain is a canine H-NOX domain. In some embodiments, the polymeric H-NOX protein comprises a L144F T. tengcongensis H-NOX domain. In some embodiments, the polymeric H-NOX protein comprises a W9F/L144F T. tengcongensis H-NOX domain and a foldon domain.

In various embodiments, the invention features a method of delivering O2 to an individual (e.g., a mammal, such as a primate (e.g., a human, a monkey, a gorilla, an ape, a lemur, etc.), a bovine, an equine, a porcine, a canine, or a feline) by administering to an individual in need thereof a wild-type or mutant H-NOX protein, including a polymeric H-NOX protein in an amount sufficient to deliver O2 to the individual. In some embodiments, the invention provides methods of carrying or delivering blood gas to an individual such as a mammal, comprising the step of delivering (e.g., transfusing, etc.) to the blood of the individual (e.g., a mammal) one or more of H-NOX compositions. Methods for delivering O2 carriers to blood or tissues (e.g., mammalian blood or tissues) are known in the art. In various embodiments, the H-NOX protein is an apoprotein that is capable of binding heme or is a holoprotein with heme bound. The H-NOX protein may or may not have heme bound prior to the administration of the H-NOX protein to the individual. In some embodiments, O2 is bound to the H-NOX protein before it is delivered to the individual. In other embodiments, O2 is not bound to the H-NOX protein prior to the administration of the protein to the individual, and the H-NOX protein transports O2 from one location in the individual to another location in the individual.

Wild-type and mutant H-NOX proteins, including polymeric H-NOX proteins, with a relatively low KD for O2 (such as less than about 80 nM or less than about 50 nM) are expected to be particularly useful to treat tissues with low oxygen tension (such as tumors, some wounds, or other areas where the oxygen tension is very low, such as a p50 below 1 mm Hg). The high affinity of such H-NOX proteins for O2 may increase the length of time the O2 remains bound to the H-NOX protein, thereby reducing the amount of O2 that is released before the H-NOX protein reaches the tissue to be treated.

In some embodiments for the direct delivery of an H-NOX protein with bound O2 to a particular site in the body (such as a glioblastoma), the koff for O2 is more important than the KD value because O2 is already bound to the protein (making the kon less important) and oxygen needs to be released at or near a particular site in the body (at a rate influenced by the koff). In some embodiments, the koff may also be important when H-NOX proteins are in the presence of red cells in the circulation, where they facilitate diffusion of O2 from red cells, and perhaps prolonging the ability of diluted red cells to transport O2 to further points in the vasculature.

In some embodiments for the delivery of an H-NOX protein that circulates in the bloodstream of an individual, the H-NOX protein binds O2 in the lungs and releases O2 at one or more other sites in the body. For some of these applications, the KD value is more important than the koff since O2 binding is at or near equilibrium. In some embodiments for extreme hemodilution, the KD more important than the koff when the H-NOX protein is the primary O2 carrier because the H-NOX protein will bind and release O2 continually as it travels through the circulation. Since hemoglobin has a p50 of 14 mm Hg, red cells (which act like capacitors) have a p50 of ˜30 mm Hg, and HBOCs have been developed with ranges between 5 mm Hg and 90 mm Hg, the optimal KD range for H-NOX proteins may therefore be between ˜2 mm Hg to ˜100 mm Hg for some applications.

Polymeric H-NOX proteins can also be used for imaging. In particular, light imaging (e.g., optical coherence tomography; see, for example, Villard, J. W. (2002). Circulation 105:1843-1849, which is incorporated by reference in its entirety particularly with respect to optical coherence tomography) is obfuscated by erythrocytes. Perfusion with an H-NOX solution allows for clearer images of the circulation and vessel walls because the H-NOX protein is much smaller than erythrocytes.

H-NOX proteins, including polymeric H-NOX proteins, and pharmaceutical compositions of the invention can be administered to an individual by any conventional means such as by oral, topical, intraocular, intrathecal, intrapulmonary, intra-tracheal, or aerosol administration; by transdermal or mucus membrane adsorption; or by injection (e.g., subcutaneous, intravenous, intra-arterial, intravesicular, or intramuscular injection). H-NOX proteins may also be included in large volume parenteral solutions for use as blood substitutes. In exemplary embodiments, the H-NOX protein is administered to the blood (e.g., administration to a blood vessel such as a vein, artery, or capillary), a wound, a tumor, a hypoxic tissue, or a hypoxic organ of the individual.

In some embodiments, a sustained continuous release formulation of the composition is used. Administration of an H-NOX protein can occur, e.g., for a period of seconds to hours depending on the purpose of the administration. For example, as a blood delivery vehicle, an exemplary time course of administration is as rapid as possible. Other exemplary time courses include about any of 10, 20, 30, 40, 60, 90, or 120 minutes. Exemplary infusion rates for H-NOX solutions as blood replacements are from about 30 mL/hour to about 13,260 mL/hour, such as about 100 mL/hour to about 3,000 mL/hour. An exemplary total dose of H-NOX protein is about 900 mg/kg administered over 20 minutes at 13,260 mL/hour. An exemplary total dose of H-NOX protein for a swine is about 18.9 grams.

Exemplary dosing frequencies include, but are not limited to, at least 1, 2, 3, 4, 5, 6, or 7 times (i.e., daily) a week. In some embodiments, an H-NOX protein is administered at least 2, 3, 4, or 6 times a day. The H-NOX protein can be administered, e.g., over a period of a few days or weeks. In some embodiments, the H-NOX protein is administrated for a longer period, such as a few months or years. The dosing frequency of the composition may be adjusted over the course of the treatment based on the judgment of the administering physician.

In some embodiments of the invention, the H-NOX protein (e.g. a polymeric H-NOX protein) is used as an adjunct to radiation therapy or chemotherapy. For example, for the treatment of glioblastoma. In some embodiments, the H-NOX is administered to the individual any of at least 1, 2, 3, 4, 5 or 6 hours before administration of the radiation or chemotherapy. In some embodiments, the radiation is X irradiation. In some embodiments, the dose of X irradiation is any of about 0.5 gy to about 75 gy. In some embodiments, the cycle of H-NOX administration and radiation administration is repeated any one of one, two, three, four, five or six times. In some embodiments, the cycle of H-NOX administration and radiation administration is repeated after any one of about one week, two weeks, three weeks, four weeks, five weeks or six weeks. In some embodiments, the admiration of H-NOX and radiation therapy is used in conjunction with another therapy; for example, a chemotherapy.

As noted above, the selection of dosage amounts for H-NOX proteins depends upon the dosage form utilized, the frequency and number of administrations, the condition being treated, and the particular purpose to be achieved according to the determination of the ordinarily skilled artisan in the field. In some embodiments, an effective amount of an H-NOX protein for administration to human is between a few grams to over 350 grams.

In some embodiments, two or more different H-NOX proteins are administered simultaneously, sequentially, or concurrently. In some embodiments, another compound or therapy useful for the delivery of O2 is administered simultaneously, sequentially, or concurrently with the administration of one or more H-NOX proteins.

Other exemplary therapeutic applications for which H-NOX proteins can be used are described by, e.g., U.S. Pat. Nos. 6,974,795, and 6,432,918, which are each hereby incorporated by reference in their entireties, particularly with respect to therapeutic applications for O2 carriers.

Biomarkers to Monitor H-NOX Mediated Oxygenation

In some aspects, the invention provides methods for monitoring oxygenation of hypoxic tumors by H-NOX proteins. In some embodiments, the invention provides methods of treating a hypoxic brain tumor (e.g., a glioblastoma) in an individual comprising administering an effective amount of an H-NOX protein, determining the level of hypoxia in the brain tumor, and administering an effective amount of radiation to the individual where the level of tumor hypoxia is reduced following administration of the H-NOX protein compared to the level of tumor hypoxia measured in the tumor prior to H-NOX administration. In some embodiments, the level of hypoxia in the brain tumor is determined prior to administration of H-NOX. In some embodiments, radiation is administered to the individual when the hypoxia of the tumor is reduced by at least about any of 5%, 10%, 15%, 20%, 25%, 50%, 75% or 100%.

In some aspects, the invention provides methods for optimizing therapeutic efficacy for treatment of hypoxic brain tumor (e.g., a glioblastoma). An effective amount of an H-NOX protein is administered to the individual and the level of hypoxia in the brain tumor is determined one or more times following administration of the H-NOX protein. An effective amount of radiation is administered to the individual when the level of tumor hypoxia is reduced following administration of the H-NOX protein compared to the level of tumor hypoxia measured in the tumor prior to H-NOX administration. In some embodiments, the level of hypoxia in the brain tumor is determined prior to administration of H-NOX. In some embodiments, radiation is administered to the individual when the hypoxia of the tumor is reduced by at least about any of 5%, 10%, 15%, 20%, 25%, 50%, 75% or 100%. In some embodiments, radiation is administered to the individual when the level of tumor hypoxia is at or near a minimum following administration of H-NOX. In some embodiments, the level of hypoxia of the brain tumor is measured at one or more of about one hour, two hours, three hours, four hours, five hours, six hours, seven hours, eight hours, nine hours, ten hours, eleven hours, twelve hours, fourteen hours, sixteen hours, eighteen hours, 24 hours, 36 hours, 48 hours, 60 hours or 72 hours after administration of H-NOX.

In some aspects, the invention provides methods to monitor the efficacy of delivery of O2 to a hypoxic brain tumor (e.g., a glioblastoma) by an H-NOX protein. An H-NOX protein is administered to the individual and the level of tumor hypoxia is measured at one or more time points after administration of the H-NOX protein. A reduction in tumor hypoxia following administration of the H-NOX protein compared to the level of tumor hypoxia measured in the tumor prior to H-NOX administration indicates effective delivery of O2 to the brain tumor. In some embodiments, the level of hypoxia in the brain tumor is determined prior to administration of H-NOX. In some embodiments, the level of hypoxia of the brain tumor is measured at one or more of about one hour, two hours, three hours, four hours, five hours, six hours, seven hours, eight hours, nine hours, ten hours, eleven hours, twelve hours, fourteen hours, sixteen hours, eighteen hours, 24 hours, 36 hours, 48 hours, 60 hours or 72 hours after administration of H-NOX. In some embodiments, a reduction in tumor hypoxia by at least about any of 5%, 10%, 15%, 20%, 25%, 50% 75% or 100% indicates that the individual is suitable for administration of anticancer therapy. In some embodiments, a reduction in tumor hypoxia by at least about any of 5%, 10%, 15%, 20%, 25%, 50% 75% or 100% indicates that the individual is suitable for administration of radiation therapy.

In some aspects, the invention provides methods to monitor responsiveness or lack of responsiveness to treatment with an H-NOX in an individual suffering from a brain tumor (e.g., a glioblastoma). The hypoxic state of the tumor is measured following H-NOX administration. Responsiveness is indicated by a reduction in tumor hypoxia or oxygenation of the tumor. In some embodiments, the level of tumor hypoxia is measured at one or more time points after administration of the H-NOX protein. In some embodiments, the level of hypoxia in the brain tumor is determined prior to administration of H-NOX. In some embodiments, the level of hypoxia of the brain tumor is measured at one or more of about one hour, two hours, three hours, four hours, five hours, six hours, seven hours, eight hours, nine hours, ten hours, eleven hours, twelve hours, fourteen hours, sixteen hours, eighteen hours, 24 hours, 36 hours, 48 hours, 60 hours or 72 hours after administration of H-NOX. In some embodiments, a reduction in tumor hypoxia by at least about any of 5%, 10%, 15%, 20%, 25%, 50% 75% or 100% indicates responsiveness to treatment with an H-NOX.

In some aspects, the invention provides methods of identifying an individual with a brain tumor (e.g., a glioblastoma) who is more likely to exhibit benefit from a therapy comprising an H-NOX protein. The hypoxia level of the tumor is determined An effective amount of an H-NOX protein is administered to the individual and the level of hypoxia in the brain tumor is determined one or more times following administration of the H-NOX protein. A decrease in tumor hypoxia by about 5% indicates that the individual is more likely to exhibit benefit from H-NOX treatment combined with radiation treatment. In some embodiments, the individual is more likely to exhibit benefit from H-NOX treatment combined with anticancer treatment when the hypoxia of the tumor is reduced by at least about any of 5%, 10%, 15%, 20%, 25%, 50%, 75% or 100%. In some embodiments, the individual is more likely to exhibit benefit from H-NOX treatment combined with radiation treatment when the hypoxia of the tumor is reduced by at least about any of 5%, 10%, 15%, 20%, 25%, 50%, 75% or 100%. In some embodiments, the level of hypoxia of the brain tumor is measured at one or more of about one hour, two hours, three hours, four hours, five hours, six hours, seven hours, eight hours, nine hours, ten hours, eleven hours, twelve hours, fourteen hours, sixteen hours, eighteen hours, 24 hours, 36 hours, 48 hours, 60 hours or 72 hours after administration of H-NOX.

In some embodiments of the invention, the determination of tumor hypoxia is repeated after about one or more of one week, two weeks, three weeks, four weeks, five weeks, six weeks, two months, three months, four months, five months, six months, of one year. In some embodiments, administration of H-NOX is repeated if the tumor is hypoxic. In some embodiments, the administration of H-NOX is repeated if the tumor has increased hypoxia compared to the tumor after an initial administration of H-NOX. In some embodiments, the increased hypoxia is an increase by any of about 5%, 10%, 15%, 20%, 25%, 50%, 75%, 100% compared to the tumor after an initial administration of H-NOX. In some embodiments, radiation is administered to the individual one or more times after a repeat administration of H-NOX. In some embodiments, radiation is administered to the individual one or more times after a repeat administration of H-NOX where a decrease in tumor hypoxia is seen with one or more repeat administrations of H-NOX.

Methods to determine the level of tumor hypoxia are known in the art. Examples include but are not limited to measurement of any one of 18F-fluoromisonidazole (FMISO) tumor uptake, pimidazole uptake, 18F-fluoroazomycin arabinoside (FAZA) uptake, 18F-fluorodeoxyglucose (FDG) uptake, a nitroimidazole uptake, Copper(II)-diacetyl-bis(N4-methylthiosemicarbazone (Cu-ATSM) uptake, hexafluorobenzene (C6F6) uptake by 19F magnetic resonance imaging, hexamethyldisiloxane uptake by 1H MRI, tumor HIF-1α expression, tumor HIF-2α expression, tumor HIF-3α expression, tumor Glut-1 expression, tumor LDHA expression, tumor carbonic anhydrase IX (CA-9) expression, or lactate and/or pyruvate levels. In some embodiments of the methods of monitoring, treating, and optimization of therapy described above, tumor hypoxia is measured by 18F-FMISO uptake. In some embodiments, 18F-FMISO uptake is measured by Positron emission tomography (PET) scan, computed tomography (CT) scan or computed axial tomography (CAT) scan. Methods to detect expression of genes such as HIF-1α are known in the art; for example, by immunoassay, by immunohistochemistry, by quantitative PCR, by hybridization (for example, on a gene chip), and the like.

In some embodiments of the methods of monitoring, treating and/or optimizing therapeutic efficacy described above, the H-NOX protein is any of the H-NOX protein described herein. In some embodiments, the H-NOX protein comprises one or more distal pocket mutations (e.g. one distal pocket mutation, two distal pocket mutations, three distal pocket mutations, four distal pocket mutations, five distal pocket mutations). In some embodiments, the H-NOX protein is a polymeric H-NOX protein (e.g. a trimeric H-NOX protein). In some embodiments, the polymeric H-NOX protein comprises one or more H-NOX domains comprising a mutation at a position corresponding to L144 of T. tengcongensis H-NOX. In some embodiments, the polymeric H-NOX protein comprises one or more H-NOX domains comprising a mutation corresponding to a L144F mutation of T. tengcongensis H-NOX. In some embodiments, the polymeric H-NOX protein comprises one or more H-NOX domains comprising a mutation at positions corresponding to W9 and L144 of T. tengcongensis H-NOX. In some embodiments, the polymeric H-NOX protein comprises one or more H-NOX domains comprising mutations corresponding to a W9F/L144F mutation of T. tengcongensis H-NOX. In some embodiments, the H-NOX domain is a human H-NOX domain. In some embodiments, the H-NOX domain is a canine H-NOX domain. In some embodiments, the polymeric H-NOX protein comprises a L144F T. tengcongensis H-NOX domain. In some embodiments, the polymeric H-NOX protein comprises a W9F/L144F T. tengcongensis H-NOX domain and a foldon domain. In some embodiments, the H-NOX protein is a trimeric H-NOX protein. In some embodiments, the trimeric H-NOX protein comprises at least one T. tengcongensis H-NOX domain. In some embodiments, the trimeric H-NOX protein comprises at least one L144F T. tengcongensis H-NOX domain. In some embodiments, the trimeric H-NOX protein comprises at least one L144F T. tengcongensis H-NOX domain and at least one foldon domain. In some embodiments, the trimeric H-NOX protein comprises three L144F T. tengcongensis H-NOX domains, each fused to a foldon domain. In some embodiments, the H-NOX protein does not comprise a guanylyl cyclase domain.

In some embodiments of the methods of monitoring, treating and/or optimizing therapeutic efficacy described above, the H-NOX protein is modified with polyethylene glycol (e.g., pegylated). In some embodiments, the H-NOX protein is fused to another polypeptide; for example but not limited to albumin or an Fc region of an immunoglobulin.

In some embodiments of the methods of monitoring, treating and/or optimizing therapeutic efficacy described above, radiation is administered to the individual following H-NOX administration. In some embodiments, the radiation is X-radiation or gamma radiation. In some embodiments, the dose of X irradiation is any of about 0.5 gy to about 75 gy. In some embodiments, the dose of X irradiation is at least about any of 0.5 gy, 1 gy, 2 gy, 3 gy, 4 gy, 5 gy, 6 gy, 7 gy, 8 gy, 9 gy, 10 gy, 15 gy, 20 gy, 25 gy, 50 gy, or 75 gy. In some embodiments, the cycle of H-NOX administration and radiation administration is repeated any one of one, two, three, four, five or six times. In some embodiments, the level of tumor hypoxia is determined after H-NOX administration and before administration of radiation (e.g., X-radiation). In some embodiments, the cycle of H-NOX administration and radiation administration is repeated after any one of about one week, two weeks, three weeks, four weeks, five weeks or six weeks. In some embodiments, the radiation therapy is external beam radiation therapy. In some embodiments, the radiation therapy is 3-dimensional conformal radiation therapy (3D-CRT), intensity-modulated radiation therapy (IMRT), image-guided radiation therapy (IGRT), tomotherapy or stereotactic radiosurgery.

As noted above, the selection of dosage amounts for H-NOX proteins depends upon the dosage form utilized, the frequency and number of administrations, the condition being treated, and the particular purpose to be achieved according to the determination of the ordinarily skilled artisan in the field. In some embodiments, an effective amount of an H-NOX protein for administration to human is between a few grams to over 350 grams. In some embodiments, the dosage of H-NOX is determined by the methods of monitoring tumor oxygenation described above; for example, the amount of H-NOX administered to the individual may be increased if the level of tumor hypoxia does not decrease significantly. In some embodiments, the amount of H-NOX administered to the individual may be decreased if the level of tumor hypoxia decreases significantly in response to H-NOX administration.

In some embodiments of the methods of monitoring, treating and/or optimizing therapeutic efficacy described above, the individual is mammal, such as a primate (e.g., a human, a monkey, a gorilla, an ape, a lemur, etc.), a bovine, an equine, a porcine, a canine, or a feline. In some embodiments, the individual is pet, a laboratory research animal or a farm animal.

In some embodiments of the methods of monitoring, treating and/or optimizing therapeutic efficacy described above, the methods comprise the step of delivering (e.g., transfusing, etc.) to the blood of the individual (e.g., a mammal) one or more of H-NOX compositions. Methods for delivering O2 carriers to blood or tissues (e.g., mammalian blood or tissues) are known in the art. In various embodiments, the H-NOX protein is an apoprotein that is capable of binding heme or is a holoprotein with heme bound. The H-NOX protein may or may not have heme bound prior to the administration of the H-NOX protein to the individual. In some embodiments, O2 is bound to the H-NOX protein before it is delivered to the individual. In other embodiments, O2 is not bound to the H-NOX protein prior to the administration of the protein to the individual, and the H-NOX protein transports O2 from one location in the individual to another location in the individual.

In some embodiments of the methods of monitoring, treating and/or optimizing therapeutic efficacy described above, the brain tumor is a glioblastoma, an astrocytoma, a meningioma, an ependymoma, a medulloblatomoa, a pineocytoma, a pineoblastoma, or a craniopharyngioma.

Kits with H-NOX Proteins

Also provided are articles of manufacture and kits that include any of the H-NOX proteins described herein including polymeric H-NOX proteins, and suitable packaging. In some embodiments, the invention includes a kit with (i) an H-NOX protein (such as a wild-type or mutant H-NOX protein described herein or formulations thereof as described herein) and (ii) instructions for using the kit to deliver O2 to an individual. In various embodiments, the invention features a kit with (i) an H-NOX protein (such as a wild-type or mutant H-NOX protein described herein or formulations thereof as described herein) and (ii) instructions for using the kit for any of the industrial uses described herein (e.g., use of an H-NOX protein as a reference standard for analytical instrumentation needing such a reference standard, enhancement of cell growth in cell culture by maintaining or increasing O2 levels in vitro, addition of O2 to a solution, or removal of O2 from a solution).

In some embodiments, kits are provided for use in the treatment of brain cancer (e.g. glioblastoma). In some embodiments, the kit comprises a polymeric H-NOX protein. In some embodiments, the kit comprises an effective amount of a polymeric H-NOX protein comprising two or more wild-type or mutant H-NOX domains. In some embodiments, the kit comprises an effective amount of a recombinant monomeric H-NOX protein comprising a wild-type or mutant H-NOX domain and a polymerization domain as described herein. In some embodiments, the kit comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a mutation corresponding to a T. tengcongensis L144F H-NOX mutation and a trimerization domain. In some embodiments, the kit comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a mutation corresponding to a T. tengcongensis W9F/L144F H-NOX mutation and a trimerization domain. In some embodiments, the trimeric H-NOX protein comprises human H-NOX domains. In some embodiments, the trimeric H-NOX protein comprises canine H-NOX domains. In some embodiments, the kit comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain. In some embodiments, the kit comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis W9F/L144F H-NOX domain and a foldon domain. In some embodiments, the kit comprises a trimeric H-NOX protein comprising three monomers, each monomer comprising a T. tengcongensis L144F H-NOX domain and a foldon domain.

Suitable packaging for compositions described herein are known in the art, and include, for example, vials (e.g., sealed vials), vessels, ampules, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. These articles of manufacture may further be sterilized and/or sealed. Also provided are unit dosage forms comprising the compositions described herein. These unit dosage forms can be stored in a suitable packaging in single or multiple unit dosages and may also be further sterilized and sealed. Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable. The instructions relating to the use of H-NOX proteins generally include information as to dosage, dosing schedule, and route of administration for the intended treatment or industrial use. The kit may further comprise a description of selecting an individual suitable or treatment.

The containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses. For example, kits may also be provided that contain sufficient dosages of H-NOX proteins disclosed herein to provide effective treatment for an individual for an extended period, such as about any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, or more. Kits may also include multiple unit doses of H-NOX proteins and instructions for use and packaged in quantities sufficient for storage and use in pharmacies, for example, hospital pharmacies and compounding pharmacies. In some embodiments, the kit includes a dry (e.g., lyophilized) composition that can be reconstituted, resuspended, or rehydrated to form generally a stable aqueous suspension of H-NOX protein.

Exemplary Methods for Production of H-NOX Proteins

The present invention also provides methods for the production of any of the polymeric H-NOX proteins described herein. In some embodiments, the method involves culturing a cell that has a nucleic acid encoding a polymeric H-NOX protein under conditions suitable for production of the polymeric H-NOX protein. In various embodiments, the polymeric H-NOX is also purified (such as purification of the H-NOX protein from the cells or the culture medium). In some embodiments, the method involves culturing a cell that has a nucleic acid encoding a monomer H-NOX protein comprising an H-NOX domain and a polymerization domain. The monomers then associate in vivo or in vitro to form a polymeric H-NOX protein. A polymeric H-NOX protein comprising heterologous H-NOX domains may be generated by co-introducing two or more nucleic acids encoding monomeric H-NOX proteins with the desired H-NOX domains and where in the two or more monomeric H-NOX proteins comprise the same polymerization domain.

In some embodiments, a polymeric H-NOX protein comprising heterologous H-NOX domains is prepared by separately preparing polymeric H-NOX proteins comprising homologous monomeric H-NOX subunits comprising the desired H-NOX domains and a common polymerization domain. The different homologous H-NOX proteins are mixed at a desired ratio of heterologous H-NOX subunits, the homologous polymeric H-NOX proteins are dissociated (e.g. by heat, denaturant, high salt, etc.), then allowed to associate to form heterologous polymeric H-NOX proteins. The mixture of heterologous polymeric H-NOX proteins may be further purified by selecting for the presence of the desired subunits at the desired ratio. For example, each different H-NOX monomer may have a distinct tag to assist in purifying heterologous polymeric H-NOX proteins and identifying and quantifying the heterologous subunits.

As noted above, the sequences of several wild-type H-NOX proteins and nucleic acids are known and can be used to generate mutant H-NOX domains and nucleic acids of the present invention. Techniques for the mutation, expression, and purification of recombinant H-NOX proteins have been described by, e.g., Boon, E. M. et al. (2005). Nature Chemical Biology 1:53-59 and Karow, D. S. et al. (Aug. 10, 2004). Biochemistry 43(31):10203-10211, which is hereby incorporated by reference in its entirety, particularly with respect to the mutation, expression, and purification of recombinant H-NOX proteins. These techniques or other standard techniques can be used to generate any mutant H-NOX protein.

In particular, mutant H-NOX proteins described herein can be generated a number of methods that are known in the art. Mutation can occur at either the amino acid level by chemical modification of an amino acid or at the codon level by alteration of the nucleotide sequence that codes for a given amino acid. Substitution of an amino acid at any given position in a protein can be achieved by altering the codon that codes for that amino acid. This can be accomplished by site-directed mutagenesis using, for example: (i) the Amersham technique (Amersham mutagenesis kit, Amersham, Inc., Cleveland, Ohio) based on the methods of Taylor, J. W. et al. (Dec. 20, 1985). Nucleic Acids Res. 13(24):8749-8764; Taylor, J. W. et al. (Dec. 20, 1985). Nucleic Acids Res. 13(24):8765-8785; Nakamaye, K. L. et al. (Dec. 22, 1986). Nucleic Acids Res. 14(24):9679-9698; and Dente et al. (1985). in DNA Cloning, Glover, Ed., IRL Press, pages 791-802, (ii) the Promega kit (Promega Inc., Madison, Wis.), or (iii) the Biorad kit (Biorad Inc., Richmond, Calif.), based on the methods of Kunkel, T. A. (January 1985). Proc. Natl. Acad. Sci. USA 82(2):488-492; Kunkel, T. A. (1987). Methods Enzymol. 154:367-382; Kunkel, U.S. Pat. No. 4,873,192, which are each hereby incorporated by reference in their entireties, particularly with respect to the mutagenesis of proteins. Mutagenesis can also be accomplished by other commercially available or non-commercial means, such as those that utilize site-directed mutagenesis with mutant oligonucleotides.

Site-directed mutagenesis can also be accomplished using PCR-based mutagenesis such as that described in Zhengbin et al. (1992). pages 205-207 in PCR Methods and Applications, Cold Spring Harbor Laboratory Press, New York; Jones, D. H. et al. (February 1990). Biotechniques 8(2):178-183; Jones, D. H. et al. (January 1991). Biotechniques 10(1):62-66, which are each hereby incorporated by reference in their entireties, particularly with respect to the mutagenesis of proteins. Site-directed mutagenesis can also be accomplished using cassette mutagenesis with techniques that are known to those of skill in the art.

A mutant H-NOX nucleic acid and/or polymerization domain can be incorporated into a vector, such as an expression vector, using standard techniques. For example, restriction enzymes can be used to cleave the mutant H-NOX nucleic acid and the vector. Then, the compatible ends of the cleaved mutant H-NOX nucleic acid and the cleaved vector can be ligated. The resulting vector can be inserted into a cell (e.g., an insect cell, a plant cell, a yeast cell, or a bacterial cell) using standard techniques (e.g., electroporation) for expression of the encoded H-NOX protein.

In particular, heterologous proteins have been expressed in a number of biological expression systems, such as insect cells, plant cells, yeast cells, and bacterial cells. Thus, any suitable biological protein expression system can be utilized to produce large quantities of recombinant H-NOX protein. In some embodiments, the H-NOX protein (e.g., a mutant or wild-type H-NOX protein) is an isolated protein.

If desired, H-NOX proteins can be purified using standard techniques. In some embodiments, the protein is at least about 60%, by weight, free from other components that are present when the protein is produced. In various embodiments, the protein is at least about 75%, 90%, or 99%, by weight, pure. A purified protein can be obtained, for example, by purification (e.g., extraction) from a natural source, a recombinant expression system, or a reaction mixture for chemical synthesis. Exemplary methods of purification include immunoprecipitation, column chromatography such as immunoaffinity chromatography, magnetic bead immunoaffinity purification, and panning with a plate-bound antibody, as well as other techniques known to the skilled artisan. Purity can be assayed by any appropriate method, e.g., by column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis. In some embodiments, the purified protein is incorporated into a pharmaceutical composition of the invention or used in a method of the invention. The pharmaceutical composition of the invention may have additives, carriers, or other components in addition to the purified protein.

In some embodiments, the polymeric H-NOX protein comprises one or more His6 tags. An H-NOX protein comprising at least one His6 tag may be purified using chromatography; for example, using Ni2+-affinity chromatography. Following purification, the His6 tag may be removed; for example, by using an exopeptidase. In some embodiments, the invention provides a purified polymeric H-NOX protein, wherein the polymeric H-NOX protein was purified through the use of a His6 tag. In some embodiments, the purified H-NOX protein is treated with an exopeptidase to remove the His6 tags.

EXAMPLES

The examples, which are intended to be purely exemplary of the invention and should therefore not be considered to limit the invention in any way, also describe and detail aspects and embodiments of the invention discussed above. The examples are not intended to represent that the experiments below are all or the only experiments performed. Unless indicated otherwise, temperature is in degrees Centigrade and pressure is at or near atmospheric.

Example 1 H-NOX Proteins Demonstrated Tumor Penetration and Oxygenation in an In Vivo Mouse Model of Cancer

Oxygen is a critical factor that enhances radiation-induced DNA damage and tumor killing. Low oxygen levels or hypoxia within solid tumors can blunt the therapeutic effects of tumor therapy. For example in hypoxic regions of the tumor, radiation therapy has been found to be three times less effective as compared to tumor regions with normal oxygen levels. As a result, many patients with tumors containing regions of hypoxia often show incomplete responses to conventional tumor therapy and have poor prognosis for survival. The correlation of hypoxia with poor patient outcomes has been observed in a wide range of tumors arising from, among others, prostate, sarcoma, pancreatic, head-and-neck, cervical, and brain cancers. See Moeller, B J et al. (2007) Cancer Metastasis Rev 26:241-248; Vaupel, P, (2004) Semin Radiat Oncol, 14:198-206; Varlotto, J, et al. (2005) Int J Radiat Oncol Biol Phys, 63:25-36; Rockwell, S, et al. (2009) Curr Mol Med. 9:442-458; which are all incorporated herein in their entirety by reference.

To determine the ability of H-NOX proteins to penetrate tumors, groups of 6 mice bearing subcutaneous HCT116 colon-derived tumors were injected via the tail vein with 750 mg/kg of a H-NOX monomer, 750 mg/kg of a T. tengcongensis L144F H-NOX trimer or saline control. The mice were subsequently sacrificed at 30 minutes or 60 minutes post-injection. The tumors were resected, sectioned, stained with an anti-H-NOX antibody, and imaged for H-NOX staining intensity (FIG. 1A). Quantification of the stained HCT-116 tumor sections demonstrated that the 23 kDa T. tengcongensis L144F H-NOX monomer accumulated in tumors by 30 minutes and exhibited partial clearance by 60 minutes (FIG. 1B). In comparison, the 80 kDa L144F H-NOX trimer accumulated in tumors by 30 minutes and continued to persist in the tumors at 60 minutes post-injection with accumulation peaking at 4 hours post-injection (FIG. 1B).

To determine if the H-NOX proteins reduced hypoxia in the tumors, groups of 6 mice bearing subcutaneous HCT116 colon-derived tumors were injected via the tail vein with 750 mg/kg of a L144F H-NOX monomer, 750 mg/kg of a L144F H-NOX trimer or saline control. Prior to euthanasia, mice were given hypoxia marker pimonidazole via intraperitoneal injection and active vasculature marker DiOC73 via intravenous injection. Tumors were harvested at either 30 minutes or 60 minutes after H-NOX protein injection, and assayed by immunohistochemistry for pimonidazole with Hydroxyprobe-1 monoclonal antibody and total vasculature with anti-CD31 antibody (FIG. 2A). Quantification of the stained HCT-116 tumor sections demonstrated that in contrast to control-treated mice the 23 kDa H-NOX monomer decreased hypoxia 30 minutes post-injection but that there was no recovery in hypoxia 60 minutes post-injection (FIG. 2B). In comparison, the 80 kDa L144F H-NOX trimer did not appear to reduce hypoxia at 30 minutes post-injection, but substantially reduced hypoxia at 60 minutes post-injection (FIG. 2B). Further experiments confirmed that in mice bearing subcutaneous HCT116 colon-derived tumors the H-NOX monomer distributed throughout the tumor tissue (FIG. 3A, bottom panel) and relieved tumor hypoxia at distances far from the vasculature as detected by anti-pimonidazole antibody (FIG. 3B, bottom panel). The Hypoxyprobe-1 (anti-pimonidazole antibody) stain was quantified in tumor tissue isolated from six mice by amount of staining as a function of distance from the vasculature. It was found that the average Hypoxyprobe-1 staining was reduced from about 13 μM in saline treated mice to 5 μM in H-NOX monomer treated mice at a distance of about 150 μm from the nearest blood vessel (FIG. 3C). These results were further confirmed in mice bearing murine RIF-1 sarcoma xenografts.

Mice bearing RIF-1 sarcoma tumors were injected via the tail vein with 750 mg/kg of T. tengcongensis L144F H-NOX trimer or saline control. Prior to euthanasia, mice were given hypoxia marker pimonidazole via intraperitoneal injection. Tumors were harvested at 120 minutes after L144F H-NOX trimer injection, and assayed by immunofluorescence imaging for H-NOX trimer distribution (FIG. 4), or by western blot for pimonidazole with Hydroxyprobe-1 monoclonal antibody, hypoxia-inducible factor 1 (HIF-1α) with anti-HIF-1α antibody, H-NOX protein with an anti-H-NOX antibody, and total protein with anti-actin antibody (FIG. 5). Immunofluorescence staining demonstrated distribution of L144F H-NOX trimer in tumor sections prepared from large isolated tumors approximately 400 mm3 and 800 mm3 in size (FIG. 4). Western blot analysis of cell lysates from harvested tumors of treated mice demonstrated that the L144F H-NOX trimer localized to tumor tissue and that these tumors had decreased pimonidazole protein adducts as compared to untreated mice (FIG. 5A). Quantification of the western blots further confirmed low levels of pimonidazole protein adducts as well as low levels of HIF-1α protein in the tumors of treated mice as compared to saline treated mice (FIGS. 5B and 5C).

Example 2 11-NOX Proteins Demonstrated Tumor Penetration and Oxygenation in an In Vivo Mouse Model of Glioblastoma

To further characterize the ability of H-NOX proteins to penetrate into tumor tissue, three mouse models of glioblastoma were used to assess the distribution of T. tengcongensis L144F H-NOX monomer and T. tengcongensis L144F H-NOX trimer in brain tumors. BT-12 cells, a childhood atypical teratoid/rhabdoid infant brain tumor line that is highly invasive into the spinal column, were used to generate a mouse model of child glioblastoma, GBM-43 cells were used for generating a radioresistant model of adult glioblastoma, and U251 cells were used to generate a hypoxic model of adult glioblastoma. The glioblastoma mouse models were generated as previous described. See Ozawa, T, et al., (2010) J Vis Exp, July 13; (41) which is incorporated in its entirety herein by reference. Briefly, BT-12 cells, U251 cells, or GBM-42 cells were harvested for intracranial injection and resuspended in Dulbecco's Modified Eagle Medium (DMEM) at a concentration of about 1×108 cells per mL. Mice were anesthetized by intraperitoneal (IP) injection of ketamine (100 mg/kg) and xylazine (10 mg/kg). The anesthetic depth was monitored prior to the first incision as well as at regular intervals through the procedure, using the pedal withdrawal reflex by pinching the foot pad on both feet. A 1 cm sagittal incision was made along the scalp, and the skull suture lines were exposed. A small hole was created by puncture with a 25 g needle, at 3 mm lateral and 0.5 mm anterior of the bregma. Using a sterile Hamilton syringe (Stoelting), 3×105 cells in 3 μl was injected at a depth of 3 mm over a 60 second period. After injection, the syringe was held in place for 1 minute and then slowly removed. The skull was cleaned with 3% hydrogen peroxide and then sealed with bone wax before closing the scalp using 7 mm surgical staples (Stoelting). Mice received a subcutaneous injection of 0.1 mg/kg buprenorphine, were placed on a heating pad and monitored until they regained mobility for use in these studies.

To determine if L144F H-NOX trimers could penetrate brain tissue, mice bearing U251 orthotopic brain tumors were injected via tail vein with either 750 mg/kg T. tengcongensis L144F H-NOX monomer or 750 mg/kg T. tengcongensis L144F H-NOX trimer. Prior to euthanasia, mice were given the hypoxia marker pimonidazole by intraperitoneal injection. For immunohistochemistry analysis, brains were isolated, sectioned and stained for pimonidazole with Hydroxyprobe-1 monoclonal antibody, hypoxia-inducible factor 1 (HIF-1α) with anti-HIF-1α antibody, H-NOX protein with an anti-H-NOX antibody, and HLA-ABC protein with an anti-HLA-ABC antibody (NvusBiological rat monoclonal antibody clone #YTH862.2) about two hours after H-NOX protein administration. A set of brain tissue samples was further stained with secondary antibodies conjugated to anti-rabbit antibody conjugated with FITC (green channel) manufactured by Jackson ImmunoResearch and DAPI for immunofluorescence imaging. Mice treated with H-NOX trimer demonstrated increased staining for H-NOX as compared to control treated mice indicating that the H-NOX trimer penetrated brain tissue (FIG. 6A). In addition, decreased staining for pimonidazole with Hydroxyprobe-1 monoclonal antibody showed that H-NOX trimer administration substantially reduced hypoxia at 60 minutes post-injection (FIG. 6B). Decreased staining for pimonidazole and HIF-1α protein was further observed in immunofluorescence images (FIGS. 7A and 7C). Quantification of the immunofluorescence images demonstrated low levels of pimonidazole staining as well as low levels of HIF-1α protein staining in the tumors of L144F H-NOX trimer treated mice as compared to saline treated mice (FIGS. 7B and 7D).

FIG. 8 shows the biodistribution of H-NOX trimer in U251 orthotopic brain tumor and healthy brain. Fluorescent imaging of H-NOX trimer at high magnification shows weak diffusion outside vessels in healthy brain.

To compare penetration and retention times between T. tengcongensis L144F H-NOX monomer and T. tengcongensis L144F H-NOX trimer, mice bearing orthotopic brain tumors were injected via tail vein with either 750 mg/kg Alexa-647 labeled H-NOX monomer or 750 mg/kg Alexa-647 labeled H-NOX trimer and subjected to bioluminescence imaging at various time points. Alexa-647 labeled H-NOX proteins were generated to confirm fluorescence excitation and emission spectra of fluorescently labeled H-NOX proteins as follows.

Purified protein, H-NOX monomer protein, H-NOX trimer, or BSA (Sigma, used as a control), was thawed on ice and buffer exchanged into endotoxin-free Labeling Buffer (50 mM HEPES, 50 mM NaCl, pH 8.0) using endotoxin-free dialysis cassettes (Pierce Slide-A-Lyzer, 7 kDa MWCO). Protein concentration after dialysis into Labeling Buffer was determined by UV-vis spectroscopy. Alexa 647 dye (Alexa Fluor® 647 carboxylic acid, succinimidyl ester, Invitrogen # A-20006) was prepared immediately before addition to the labeling reactions. Dye was warmed to room temperature and then dissolved in DMSO at a final concentration of 10 mg/mL. The mixture was vortexed for 10 seconds and then dye was added to each labeling reaction. Labeling reactions used a range of protein:dye ratios to control the extent of Alexa labeling. Reactions consisted of protein (in Labeling Buffer) and dye for a final DMSO concentration of 5-10%. Reactions were incubated for 1 hour at room temperature (protected from light) with moderate shaking. After the reaction, free Alexa dye was removed by extensive dialysis into endotoxin-free formulation buffer (30 mM Triethanolamine, 50 mM NaCl, pH 7.4) using endotoxin-free dialysis cassettes (Pierce Slide-A-Lyzer, 7 kDa MWCO cutoff).

After dialysis into formulation buffer, the protein concentration and extent of labeling was determined by UV-vis spectroscopy using the intrinsic absorbance of H-NOX (at 280 and 415 nm) and Alexa dye (653 nm) to determine the molar ratio of dye to protein after labeling. Fluorescence of the labeled protein was analyzed by excitation at 647 nm to collect an emission spectrum. The emission spectrum of the labeled protein was consistent with published data and Invitrogen data. Labeled protein was further analyzed by size exclusion chromatography to ensure that labeling did not affect the oligomerization state of the protein. Final endotoxin contamination in the labeled protein was determined using the Charles River LAL Gel Clot assay (0.03 EU/mL sensitivity).

For bioluminescence imaging, mice were anesthetized by IP injection of ketamine (100 mg/kg) and xylazine (10 mg/kg), and then injected by IP with 33.3 mg of D-luciferin (potassium salt, Gold Biotechnology, St. Louis, Mo., USA) dissolved in sterile saline. Tumor bioluminescence was determined 10 minutes after luciferin injection, using the IVIS Lumina System (Caliper Life Sciences, Alameda, Calif., USA) and LivingImage software, as the sum of photon counts per second in regions of interest defined by a lower threshold value of 25% of peak pixel intensity. Imaging acquisition was non-invasive, and animal body temperature was maintained using a heated imaging platform. For BT-12 mice treated with H-NOX monomer or H-NOX trimer, imaging was performed at 0, 0.5, 1, 2, and 4 hrs post injection. For GBM-41 mice treated with H-NOX monomers or H-NOX trimers, imaging was performed at 0, 0.5, 1, 2, 4, and 6 hrs post injection. For U251 mice treated with H-NOX monomers or H-NOX trimers, imaging was performed at 0, 0.5, 1, 2, 4, 6, and 72 hrs post injection. Tumor bioluminescence has previously been shown to be directly proportional to tumor volume in mice bearing orthotopic GB xenografts. See Moeller, B J et al., (2007) Cancer Metastasis Rev, 26:241-248, which is incorporated herein in its entirety by reference. Comparison of H-NOX monomer and trimer biodistribution demonstrated that in the BT-12 mouse model of glioblastoma, both L144F H-NOX monomer (FIGS. 9A and 23A) and L144F H-NOX trimers (FIGS. 9B and 23A) penetrated brain tumors. H-NOX trimer had a significantly longer retention time in tumors as compared to H-NOX monomers. Whereas HNOX monomer was largely eliminated from tumors by 2 hours (FIGS. 9A and 23A), H-NOX trimer continued to accumulate in tumors for several hours (FIGS. 9B and 23A). H-NOX intracranial localization was confirmed by ex vivo imaging of brain tissue isolated from mice 30 and 60 minutes post injection with the H-NOX monomer (FIGS. 10A, 21C and 7B) and 60 minutes post injection with the H-NOX trimer (FIGS. 10C and 21D). Further visualization by bioluminescence at 30, 60, 120, and 240 minutes post injection demonstrated that the H-NOX monomer (FIG. 11) and H-NOX trimer (FIG. 12) also localized to metastatic colonies in the spinal column Whereas H-NOX monomer substantially accumulated in the spinal column at 30 minutes (FIG. 11A) as compared to H-NOX trimer (FIG. 12A), it was largely eliminated by 2 hours (FIG. 11B-D) while the H-NOX trimer continued to accumulate in the spinal column for several hours (FIG. 12B-D). By using a smaller amount of labeled protein and increasing the signal intensity, it was revealed that H-NOX monomer accumulated in the kidneys over time suggesting a route of elimination (FIG. 13).

The accumulation of L144F H-NOX trimers in the brain and spinal column was confirmed in the GBM-43 (FIG. 14) and U251 mouse models (FIGS. 15 and 23B). Localization of L144F H-NOX trimers was further investigated in U251 mice that were injected with a higher H-NOX trimer dose of 295 mg/kg and a lower dose of 30 mg/kg. Bioluminescence imaging at 0, 0.5, 1, 2, 4, and 6 hr post-injection demonstrated that the L144F H-NOX trimer accumulated in brain tumors of the mice at both the high and low concentrations of H-NOX trimer administration (FIGS. 16 and 17, respectively). In comparison, localization of an H-NOX trimer assembled from a H-NOX monomer L144F variant did not accumulate in small brain tumors as evidenced by bioluminescence images 0, 0.5, 1, 2, 4, and 6 hr post-injection with 30 mg/kg (FIG. 18). Ex vivo bioluminescence imaging of isolated brain from mice treated with 30 mg/kg L144F H-NOX trimer (FIG. 19A) or 750 mg/kg L144F trimer (FIG. 19B) showed that the amount of H-NOX protein in a single dose had little effect on H-NOX localization to intracranial tumors. Furthermore, real-time bioluminescence imaging of mice bearing large (FIG. 19C) or small tumors (FIG. 19D) showed that after administration of 295 mg/kg of L144F H-NOX trimer, the trimer distributed to intracranial tumors regardless of tumor size (FIG. 19B). Real-time and ex vivo bioluminescence imaging of three mouse models of glioblastoma, GBM, U251, and BT-12, demonstrated that L144F H-NOX trimer distributed to intracranial tumors and spinal tumors in all three models (FIGS. 20, 21 and 23A and B) Immunofluorescence imaging of a tumor section stained with antibodies to H-NOX protein and the vasculature showed that L144F H-NOX trimer left the vasculature and diffused throughout the brain tumor (FIG. 22). Overall, these data identified H-NOX proteins with clinically relevant tumor biodistribution profiles.

Mice were sacrificed and the whole extracted brain analyzed by imaging ex vivo, confirming localization of the protein to the tumor bearing part of the brain. More detailed examination by immunohistochemical analysis revealed different pattern of protein localization between normal and tumor brain tissue (FIG. 23C). While protein was detectable in blood vessels throughout the brain, it extravasated and penetrated deep into tissue only in tumor areas diffusing far from the blood vessels identified with CD31 marker (FIG. 23D). Similar results have been obtained in other brain orthotopic models including human patient-derived tumors GBM 43, GBM 39, GBM 6 and immunocompetent mouse glioblastoma GL261, as well as in subcutaneous xenograft HCT116 and immunocompetent RIF-1 models (data not shown).

To verify the partition of H-NOX trimers between plasma and brain, L144F trimer was tested using a group of three female FVB mice (FIG. 24). Candidate H-NOX trimers were injected at time 0 at a dose of 200 and 750 mg/kg by intravenous bolus injection into the tail vein. At 30 min, 1 hr, 1.5 hr and 2 hr post injection of the candidate H-NOX trimer or buffer control, mice were sacrificed. About one ml of blood was collected by intracardiac puncture and brain were harvested. Collected blood was processed for plasma and brain samples were lyzed to extract proteins. Plasma and brain were subsequently analyzed for the presence of H-NOX trimer using an ELISA assay with a polyclonal antibody against the H-NOX protein.

Example 3 11-NOX Trimers Enhanced Effects of Radiation in In Vivo Mouse Models of Glioblastoma

To determine if oxygenation of hypoxic tumors due to H-NOX penetration could enhance radiation-induced tumor killing, studies were conducted in groups of 10 athymic U251 mice bearing intracranial glioblastoma tumors to evaluate the effects of radiation therapy (RT) in the presence of H-NOX trimer. Mice were treated with three fractions of radiation therapy at 2 Gy per fraction on days 15, 17, and 20 post-tumor implantation either with or without administration of 750 mg/kg Alexa-647 labeled T. tengcongensis L144F H-NOX trimer delivered by intravenous injection. Mice were monitored up to day 29 and subjected to bioluminescence imaging at days 15, 17, 20, 22, 24, and 29. Mean bioluminescence imaging (BLI) scores determined for each treatment group demonstrated that multiple doses of L144F H-NOX trimer resulted in statistically significant delays in tumor growth (FIGS. 25A and 25B) and despite the aggressive and mildly hypoxic nature of the treated U251 orthotopic tumors, animal survival was also significantly enhanced in L144F H-NOX treated groups (FIG. 25C). Tumors were also harvested for immunohistochemistry staining and analysis.

The effect of T. tengcongensis L144F H-NOX trimer on radiation therapy of human glioblastoma was further investigated in two mouse models bearing intracranial glioblastoma tumors, U251 and GBM43. In one study, groups of 10 female athymic U251 mice bearing intracranial glioblastoma tumors were treated with either 1) treatment buffer alone, 2) treatment buffer in combination with a single dose of 2 Gy radiation (irradiator set up=0.81; dose rate of Cesium irradiator was 247 CGy/min), 3) 750 mg/kg L144F H-NOX trimer by IV alone, or 4) L144F H-NOX trimer in combination with a single dose of 2 Gy radiation (irradiator set up=0.81; dose rate of Cesium irradiator was 247 CGy/min) Mice receiving the combination treatment were irradiated 2 hours post L144F H-NOX trimer delivery at the supratentorial portion of the brain. Treatment for all mice began 14 days after intracranial injection of mice with 3.0×105 U251 cells. It was found that animal survival increased in cohorts receiving the combination treatment of L144F H-NOX trimer and 2 Gy radiation (FIG. 26A). In another study, groups of 10 GBM43 mice bearing intracranial glioblastoma tumors were treated with either 1) 2 Gy radiation therapy; 2) 4 Gy radiation therapy; 3) 8 Gy radiation therapy; 4) 2 cycles of 4 Gy radiation therapy; 5) 4 Gy radiation therapy in combination with L144F H-NOX trimer; or 6) treatment buffer. Mice receiving the combination treatment were irradiated 1 to 1.5 hours post H-NOX trimer delivery and mice receiving multiple doses of RT had administration of RT separated by 4 days. Radiation treatment was administered at the supratentorial portion of the brain for all RT groups. Treatment for all mice began 7 days post-tumor implantation. It was found that animal survival in cohorts receiving the combination treatment of L144F H-NOX trimer and 4 Gy radiation was similar to animal survival in cohorts receiving 4 Gy treatment alone (FIG. 26B).

Example 4 Single Dose of Trimeric H-NOX Reduces Tumor Hypoxia Both in the Tumor Core and at the Invasive Edges

Mice bearing orthotopic glioblastoma U251 tumors were treated with either trimeric Tt L144F H-NOX (OMX-4.80) or buffer alone (FIGS. 27A and 27D, top panels) via tail vein bolus injection. Prior to euthanasia, mice were intraperitoneally injected with the hypoxia marker pimonidazole. Tumors were harvested 2 hr-30 hr after H-NOX administration, and assayed by immunohistochemistry for pimonidazole (Hypoxyprobe-1 mAb) and total cell nuclear (DAPI) staining in FIG. 27A and FIG. 27B, or for HIF1α and tumor cell marker (HLA) in FIG. 27C and FIG. 27D. Representative tumor sections from mice treated with buffer or H-NOX are shown in FIG. 27a. Hypoxia staining (pimonidazole) is shown in green and total cell nuclear staining (DAPI) is shown in dark blue. Only 2 h and 24 h time points are shown. Quantification of pimonidazole staining intensity in tumor sections is shown in FIG. 27B. Tumors treated with OMX-4.80 show hypoxia reduction at 4-24 hr. Quantification of HIF1α staining intensity in tumor sections is shown in FIG. 27C. Tumors treated with H-NOX exhibit hypoxia reduction at 16-24 hr. This suggests an oxygenation window between 2 and 30 hours post H-NOX injection. Representative tumor sections from mice treated with buffer or H-NOX are shown in FIG. 27D. Hypoxia staining (HIF1α) is shown in green and staining of human tumor cells in red (HLA). Only 2 h and 24 h time points are shown.

To investigate reduction of hypoxia at the invasive edge of the tumor, mice bearing orthotopic glioblastoma U251 tumors were treated with either buffer control (FIGS. 28A, and 28C) or trimeric Tt L144F H-NOX, OMX-4.80 (FIGS. 28B, 28D, 28E, and 28F), via tail vein injection and received an injection of hypoxia marker, pimonidazole, one hour prior to sacrifice. Brains containing tumors were extracted and subjected to immunohistochemical analysis using anti-pimonidazole (FIGS. 28A and 28B), HIF1α (FIGS. 28C and 28D), or OMX-4.80 antibodies (FIGS. 28E and 28F). Slides were counterstained with DNA marker (DAPI, blue labeled nuclei) and images merged (FIGS. 28A-28D, and 28F). Invasive edges (white arrows) of the non-treated tumors tend to exhibit higher level of hypoxia, as determined by both external (pimo) and cellular (HIF1α) hypoxia markers. Upon trimeric Tt L144F H-NOX treatment, hypoxia is significantly reduced throughout tumor tissue (see FIG. 28), including the invasive edges (FIGS. 28B and 28D) where OMX-4.80 can be readily detected (FIGS. 28E and 28F).

The ability of the trimeric Tt L144F H-NOX to oxygenate hypoxic tumor areas in orthotopic GBM models using two markers of hypoxia—the external hypoxia marker, pimonidazole, an analog of the clinically relevant PET hypoxia marker-18F-FMISO, (FIGS. 27A and 28A,B), and hypoxia inducible factor, HIF1α (FIGS. 27B and 28C,D). This data demonstrates that single dose of H-NOX administered i.v. induces a greater than 50% decrease in expression of both hypoxia markers and that this effect is maintained for over 20 hr (FIG. 27). Furthermore, H-NOX can reach and efficiently oxygenate invasive edges of the tumor (FIG. 28) which express hypoxia-related aggressive tumor phenotypes associated with therapy resistance and poor patient outcomes.

Example 5 Treatment with Trimeric H-NOX Enhances Efficacy of a Single Dose of Radiation

Trimeric H-NOX sensitizes human GBM intracranial xenografts and syngeneic RIF1 tumors to radiation therapy. Athymic mice bearing orthotopic U251 (GBM) xenografts were treated with 10 Gy RT either with or without pre-treatment with trimeric Tt L144F H-NOX (650 mg/kg, IV) or with 15 Gy alone (FIG. 29A-D). Treatment was administered on day 14 post-tumor implantation. Syngeneic RIF1 tumors, 250-300 mm3 size, were treated with 15 Gy RT either with or without pre-treatment with trimeric Tt L144F H-NOX (750 mg/kg, IV) or with 25 Gy alone (FIG. 29D). FIG. 29A shows mean bioluminescence imaging (BLI) scores±SEM from mice bearing human GBM intracranial xenografts and treated with trimeric Tt L144F H-NOX and 10 gray radiation or buffer and 10 gray radiation, as well as an untreated (buffer, no RT) control group. The BLI scores of the H-NOX+RT mice are significantly lower than those from mice treated with RT alone (p=0.015, Student's t-test). FIG. 29B shows that trimeric Tt L144F H-NOX significantly enhanced survival, as compared to mice that received only radiotherapy (p<0.05, logrank test). FIG. 29C shows tumor growth delay in the H-NOX+10 Gy group was not statistically different from 15 Gy alone group. Tumor volume measurements in RIF1 tumors showed 6.4 days growth delay at 4× volume in trimeric Tt L144F H-NOX+15 Gy group relative to 15 Gy alone, an equivalent to 25 Gy RT. Unlike, trimeric Tt L144F H-NOX, treatment with an inactive form of the H-NOX protein that does not release oxygen, did not cause any radiation enhancement as compared to radiation alone (FIG. 29D).

A single dose of trimeric H-NOX, administered 24 hr prior to radiation, delayed tumor growth 2.7 fold and increased survival 48% of mice bearing 14 d-old orthotopic GBM tumors (FIG. 29). This effect was equivalent to increasing the RT dose by 50% resulting in OER of 1.5 (FIG. 29C). Similar results were obtained in syngeneic subcutenous tumor model with OER of 1.7. Importantly, inactive form of the trimeric H-NOX protein that cannot release oxygen under hypoxic tumor conditions, did not affect the tumor growth (FIG. 29D).

Example 6 Targeting Hypoxia in Glioblastoma Multiforme with H-NOX

Brain tumors were collected from mice bearing intracranial orthotopic glioblastoma multiforme tumors (U251, GBM6, GBM39, GBM43 and GL261). Hypoxia was evaluated by immunohistochemistry of endogenous markers: the transcription factor HIF-1α (FIG. 31B) and by the exogenous clinically relevant hypoxia marker, pimonidazole (FIG. 31A). Prior to euthanasia, mice were intraperitoneally injected with the hypoxia marker pimonidazole. Tumors were harvested and assayed by immunohistochemistry for pimonidazole (Hypoxyprobe-1 polyclonal Ab). Pimonidazole staining reveals distinct patterns of hypoxia in each GBM models (FIG. 31A and Table 2). Other tumors were harvested and assayed by immunohistochemistry for HIF-1α. HIF-1α staining reveals distinct patterns of hypoxia in each GBM models. Table 2 shows a comparison of hypoxic levels between GBM models as measured by pimonidazole staining or HIF-1α staining (“+”, weak staining; “++”, moderate staining; “+++”, strong staining) is determined by visual scoring of the % of total tumor area exhibiting pimonidazole immunoreactivity.

TABLE 2 Tumor hypoxia in four models of glioblastoma GBM model Pimonidazole HIF-1α GL261 +++ + U251 ++ +++ GBM43 +/++ ++ GBM6 + +

As described in Example 2, all hypoxia markers exhibited high signal in poorly vascularized areas of U251 and GL261, as determined by costaining with CD31 endothelial marker. In contrast, GBM6, GBM43 and GBM39 tumors showed significant range in hypoxia marker signal intensity between samples, indicating heterogeneity in extent of hypoxia within and between GBM models. When H-NOX was administered to mice bearing intracranial U251 tumors, H-NOX significantly decreased HIF-1α stabilization and pimonidazole accumulation, demonstrating that H-NOX increases tumor oxygenation. In addition, H-NOX treated tumors exhibited fewer Ki67+ cells, suggesting that H-NOX-mediated acute re-oxygenation inhibits tumor cell proliferation.

FIG. 32A shows a schematic representation of quantitative oxygen dependencies for OMX-4.80, OMX-1.80, bioreductive activation of imaging agents (pimonidazole), and biological responses to hypoxia (HIF-1α). Three commonly used units for oxygen concentration are shown on the x axis.

Mice bearing orthotopic glioblastoma U251 tumors treated with either polymeric OMX-4.80 (750 mg/kg) or buffer alone via tail vein bolus injection in Example 4 were used to compare hypoxia measured by pimonidazole and measured by HIF-1α in response to H-NOX treatment. As described in Example 4, prior to euthanasia, mice were intraperitoneally injected with the hypoxia marker pimonidazole. Tumors were harvested 2 hours-30 hours after OMX-4.80 administration, and assayed by immunohistochemistry for pimonidazole (Hypoxyprobe-1 polyclonal Ab) and HIF-1α. Quantification of immunohistochemical signal for pimonidazole and HIF-1α was performed with ImageJ. As shown in FIG. 32B, HIF-1α levels correlate significantly with pimonidazole levels (Spearman's correlation coefficient; r=0.6468, p<0.0001). The solid line represents the linear regression curve of the best fit.

Claims

1. A method of treating a hypoxic brain tumor in an individual comprising

a) administering an effective amount of an H-NOX protein to the individual,
b) determining the level of hypoxia in the brain tumor following administration of the H-NOX protein, and
c) administering an effective amount of radiation to the individual wherein the tumor hypoxia measured in step b) is reduced compared to the level of hypoxia in the brain tumor prior to H-NOX administration.

2. A method of treating a hypoxic brain tumor in an individual comprising

a) determining the level of hypoxia in the brain tumor,
b) administering an effective amount of an H-NOX protein to the individual,
c) determining the level of hypoxia in the brain tumor following administration of the H-NOX protein, and
d) administering an effective amount of radiation to the individual wherein the tumor hypoxia measured in step c) is reduced compared to the level of hypoxia measured in step a).

3. A method of optimizing therapeutic efficacy for treatment of a hypoxic brain tumor in an individual, the method comprising

a) administering H-NOX to the individual,
b) measuring the level of hypoxia of the tumor one or more times after administration of the H-NOX protein,
c) administering radiation therapy when tumor hypoxia is reduced compared to the level of hypoxia prior to H-NOX administration.

4. The method of claim 3, wherein the hypoxia is reduced by at least about 5%, 10%, 15%, 20%, 25% or 50%.

5. A method of monitoring the efficacy of delivery of O2 to hypoxic brain tumor by an H-NOX protein in an individual, the method comprising

a) administering an effective amount of H-NOX protein to the individual,
b) measuring the level of hypoxia in the tumor at one or more time points after administration of the H-NOX protein, wherein a reduction of tumor hypoxia compared to the level of hypoxia in the tumor prior to administration of H-NOX indicates effective delivery of O2 to the brain tumor.

6. The method of claim 5, wherein the reduction in tumor hypoxia indicates that the individual is suitable for administration of radiation therapy.

7. The method of claim 3, wherein the level of hypoxia in the tumor is measured one or more of one hour, two hours, three hours, four hours, eight hours, twelve hours, 24 hours, 48 hours or 72 hours after administration of H-NOX.

8. A method of monitoring responsiveness or lack of responsiveness to treatment with a H-NOX in an individual suffering from a brain tumor comprising measuring the hypoxic state of the tumor following H-NOX administration, wherein responsiveness is indicated by a reduction in tumor hypoxia.

9. The method of claim 8, wherein the responsiveness indicates that the individual is suitable for administration of radiation therapy.

10. A method of identifying an individual with a brain tumor who is more likely to exhibit benefit from a therapy comprising an H-NOX protein, said method comprising

a) determining the hypoxia level of the tumor,
a) administering H-NOX to the individual,
b) measuring the level of hypoxia of the tumor,
c) wherein about a 5% decrease in hypoxia indicates the individual is more likely to exhibit benefit from radiation treatment in combination with H-NOX treatment.

11. The method of claim 10, wherein the decrease in hypoxia of step c) is a at least a 10%, a 15%, a 20%, a 25%, a 50%, a 75% or a 100% decrease in hypoxia.

12. The method of claim 1, wherein tumor hypoxia is measured by one or more of 18F-fluoromisonidazole (FMISO) tumor uptake, pimidazole uptake, 18F-fluoroazomycin arabinoside (FAZA) uptake, a nitroimidazole uptake, Copper(II)-diacetyl-bis(N4-methylthiosemicarbazone (Cu-ATSM) uptake, 19F magnetic resonance imaging of hexafluorobenzene (C6F6) uptake, 1H MRI of hexamethyldisiloxane uptake, tumor HIF-1α expression, tumor Glut-1 expression, tumor LDHA expression, tumor carbonic anhydrase IX (CA-9) expression, or lactate and/or pyruvate levels.

13. The method of claim 1, wherein the determination of the level of hypoxia in the tumor is repeated.

14-15. (canceled)

16. The method of claim 15, further comprising administration of radiation following administration of H-NOX.

17. (canceled)

18. The method of claim 1, wherein the radiation is X-radiation.

19. The method of claims 18, wherein the X-radiation is administered at about 0.5 gray to about 75 gray.

20. The method of claim 1, where the brain cancer is glioblastoma.

21. (canceled)

22. The method of claim 1, wherein the individual is a human.

23-24. (canceled)

25. The method of claim 1, wherein the H-NOX protein is a T. tengcongensis H-NOX, a L. pneumophilia 2 H-NOX, a H. sapiens β1, a R. norvegicus β1, a C. lupus H-NOX domain, a D. melangaster β1, a D. melangaster CG14885-PA, a C. elegans GCY-35, a N. punctiforme H-NOX, C. crescentus H-NOX, a S. oneidensis H-NOX, or C. acetobutylicum H-NOX.

26. The method of claim 1, wherein the H-NOX protein comprises a H-NOX domain corresponding to the H-NOX domain of T. tengcongensis set forth in SEQ ID NO:2.

27. The method of claim 1, wherein the H-NOX comprises one or more distal pocket mutations.

28. The method of claim 27, wherein the distal pocket mutation is an amino acid substitution at a site corresponding to L144 of T. tengcongensis H-NOX.

29. (canceled)

30. The method of claim 26, wherein the amino acid substitution at position 144 is an L144F substitution.

31-34. (canceled)

35. The method of claim 1, wherein the H-NOX protein is a polymeric H-NOX protein.

36. The method of claim 35, wherein the polymeric H-NOX protein comprises monomers, wherein the monomers comprise an H-NOX domain and a polymerization domain.

37. The method of claim 36, wherein the H-NOX domain is covalently linked to the polymerization domain.

38. The method of claim 35, wherein the polymeric H-NOX protein is a trimeric H-NOX protein.

39. The method of claim 38, wherein the trimeric H-NOX protein comprises one or more trimerization domains.

40. (canceled)

41. The method of claim 39, wherein the trimerization domain is a foldon domain.

42. The method of claim 41, wherein the foldon domain comprises the amino acid sequence of SEQ ID NO:4.

43. (canceled)

44. The method of claim 1, wherein the H-NOX protein is covalently bound to polyethylene glycol.

45-56. (canceled)

Patent History
Publication number: 20150273024
Type: Application
Filed: Oct 31, 2014
Publication Date: Oct 1, 2015
Inventors: STEPHEN P. L. CARY (SAN MATEO, CA), ANA KRTOLICA (SAN FRANCISCO, CA), NATACHA LE MOAN (SAN FRANCISCO, CA), LAURA SERWER (BRISBANE, CA), JENNIFER A. GETZ (SAN BRUNO, CA)
Application Number: 14/530,569
Classifications
International Classification: A61K 38/41 (20060101); A61N 5/10 (20060101); G01N 33/574 (20060101);