US20140106004A1 - Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence - Google Patents

Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence Download PDF

Info

Publication number
US20140106004A1
US20140106004A1 US13/713,031 US201213713031A US2014106004A1 US 20140106004 A1 US20140106004 A1 US 20140106004A1 US 201213713031 A US201213713031 A US 201213713031A US 2014106004 A1 US2014106004 A1 US 2014106004A1
Authority
US
United States
Prior art keywords
hemoglobin
cells
tumors
pharmaceutical composition
tumor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/713,031
Other languages
English (en)
Inventor
Bing Lou Wong
Norman Fung Man WAI
Sui Yi Kwok
Sze Hang LAU
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
VISION GLOBAL HOLDINGS Ltd
Original Assignee
VISION GLOBAL HOLDINGS Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by VISION GLOBAL HOLDINGS Ltd filed Critical VISION GLOBAL HOLDINGS Ltd
Priority to US13/713,031 priority Critical patent/US20140106004A1/en
Assigned to VISION GLOBAL HOLDINGS LTD. reassignment VISION GLOBAL HOLDINGS LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KWOK, SUI YI, LAU, SZE HANG, WAI, NORMAN FUNG MAN, WONG, BING LOU
Priority to MYPI2015701107A priority patent/MY186484A/en
Priority to BR112015007475A priority patent/BR112015007475A2/pt
Priority to MA37994A priority patent/MA37994A2/fr
Priority to MX2015004512A priority patent/MX367562B/es
Priority to EP13844670.3A priority patent/EP2906222A4/en
Priority to CN201380053346.1A priority patent/CN104717966B/zh
Priority to SG11201502133SA priority patent/SG11201502133SA/en
Priority to SG10201608747RA priority patent/SG10201608747RA/en
Priority to AP2015008315A priority patent/AP2015008315A0/xx
Priority to EA201500301A priority patent/EA201500301A1/ru
Priority to SG10201607846PA priority patent/SG10201607846PA/en
Priority to AU2013329121A priority patent/AU2013329121B2/en
Priority to CA2884521A priority patent/CA2884521C/en
Priority to JP2015536905A priority patent/JP6113850B2/ja
Priority to KR1020157012185A priority patent/KR20150065881A/ko
Priority to PCT/US2013/064418 priority patent/WO2014059199A1/en
Priority to UY35082A priority patent/UY35082A/es
Priority to TW102136951A priority patent/TW201414489A/zh
Priority to ARP130103741A priority patent/AR093023A1/es
Publication of US20140106004A1 publication Critical patent/US20140106004A1/en
Priority to US14/308,725 priority patent/US9056098B2/en
Priority to PH12015500562A priority patent/PH12015500562B1/en
Priority to IL237763A priority patent/IL237763A/en
Priority to ZA2015/01949A priority patent/ZA201501949B/en
Priority to CL2015000897A priority patent/CL2015000897A1/es
Priority to HK15107105.0A priority patent/HK1206281A1/zh
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/41Porphyrin- or corrin-ring-containing peptides
    • A61K38/42Haemoglobins; Myoglobins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/38Silver; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to a hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of tumor recurrence in humans and other animals.
  • the present invention relates to a composition including a hemoglobin-based oxygen carrier which is either administered alone or in combination with at least one chemotherapeutic agent for treating cancers, targeting cancerous cells/cancer stem cells/tissues containing any of these cells, and preventing the recurrence of tumors.
  • Hemoglobin plays an important role in most vertebrates for gaseous exchange between the vascular system and tissue. It is responsible for carrying oxygen from the respiratory system to the body cells via blood circulation and also carrying the metabolic waste product carbon dioxide away from body cells to the respiratory system, where the carbon dioxide is exhaled. Since hemoglobin has this oxygen transport feature, it can be used as a potent oxygen supplier if it can be stabilized ex vivo and used in vivo.
  • Naturally-occurring hemoglobin is a tetramer which is generally stable when present within red blood cells. However, when naturally-occurring hemoglobin is removed from red blood cells, it becomes unstable in plasma and splits into two ⁇ - ⁇ dimers. Each of these dimers is approximately 32 kDa in molecular weight. These dimers may cause substantial renal injury when filtered through the kidneys and excreted. The breakdown of the tetramer linkage also negatively impacts the sustainability of the functional hemoglobin in circulation.
  • Hypoxia is common in cancers. Hypoxia can lead to ionizing radiation and chemotherapy resistance by depriving tumor cells of the oxygen essential for the cytotoxic activities of these agents. Hypoxia may also reduce tumor sensitivity to radiation therapy and chemotherapy through one or more indirect mechanisms that include proteomic and genomic changes. Therefore, there is a need for improved cancer-treatment compositions, particularly, improved cancer-treatment compositions that enhance the efficacy of cytotoxic agents.
  • cancer stem cells play an important role in cancer and tumor development.
  • the stochastic model there is generally one class of tumor cells which are functionally homogeneous, and the genetic changes can lead to malignancy progression in all these tumor cells.
  • the cancer stem cell model proposes that a rare population of cells which have a distinct ability to consistently initiate tumor growth and are able to reproduce a hierarchy of functionally heterogeneous classes of cells may have different tumorigenic pathways compared with the majority of the cells in a tumor.
  • the tumor-initiating cells proposed in the cancer stem cell model can be progressively identified and purified from the rest of the cells. These cells are called cancer stem cells (CSCs). Like leukemia stem cells, other cancers such as breast cancer appear to be driven by the rare population of tumor-initiating cells. Two phenotypes of cells have been identified in breast cancer where one minority phenotype is able to form mammary tumors while another phenotype is not. In brain cancer, two types of cells are found: CD133 + cells possess differentiative, self-renewal, and tumor-initiating abilities in vivo whereas CD133 ⁇ cells cannot.
  • cancer stem cell niches have much lower oxygen tension.
  • a hypoxic niche is found to be located further away from vasculature of a tumor and contains cancer stem cells which differentially respond to hypoxia with distinct HIF (Hypoxia-inducible factors) induction patterns, in particular HIF-2 ⁇ . It becomes a new target in the signaling pathways that regulate cancer stem cell self-renewal, proliferation, and survival, and the inhibition of which will attenuate their tumor initiation potential.
  • HIF Hydrooxia-inducible factors
  • compositions that can provide high oxygen tension in cancer stem cells.
  • Such a composition could be used to produce oxidative stress or shocks which leads to DNA damage and subsequent DNA damage induced apoptosis in the cancer stem cells.
  • the present invention relates to a hemoglobin-based oxygen carrier-containing pharmaceutical composition for targeted treating and preventing recurrence of cancer in humans and other animals.
  • the first aspect of the present invention is to provide a hemoglobin-based oxygen carrier which is configured to target cancerous cells, cancer stem cells (CSCs) and/or cancerous progenitor cells, and/or tissues containing any of these cells in a human or animal body, triggering a receptor-mediated mechanism and leading a combined chemotherapeutic agent to localize together in the cytoplasm of the cancerous cells, CSCs, and/or tissues containing any of these cells, in order to increase the efficacy of both hemoglobin-based oxygen carrier and the chemotherapeutic agent.
  • CSCs cancer stem cells
  • chemotherapeutic agent to localize together in the cytoplasm of the cancerous cells, CSCs, and/or tissues containing any of these cells, in order to increase the efficacy of both hemoglobin-based oxygen carrier and the chemotherapeutic agent.
  • the localized hemoglobin-based oxygen carrier is also found to sensitize the cancerous cells and CSCs such that the cancerous cells and CSCs become more sensitive to the chemotherapeutic agent.
  • the second aspect of the present invention is to provide a method of using the hemoglobin-based oxygen carrier-containing pharmaceutical composition of the present invention for treating cancer and preventing recurrence of cancer by administering said composition to a subject in need thereof suffering from various tumors, cancers or diseases associated with tumors or cancers.
  • the hemoglobin-based oxygen carrier used in the present invention can be a heat stable cross-linked tetrameric, polymerized, pegylated or recombinant/modified hemoglobin which is used in combination with at least one chemotherapeutic agent for the treatment of various cancers such as leukemia, head and neck cancer, colorectal cancer, lung cancer, breast cancer, liver cancer, nasopharyngeal cancer, esophageal cancer and brain cancer.
  • the hemoglobin-based oxygen carrier itself is also found to have an ability to destroy cancer cells through improving the oxygenation of tumors in a hypoxic condition, thereby enhancing the sensitivity towards radiation and chemotherapeutic agents.
  • the hemoglobin-based oxygen carrier of the present invention can also be used alone for reducing cancerous tumor recurrence and minimizing tumor cell metastasis. Said hemoglobin is administered prior to ischemia for a tumor removal surgery and during re-establishment of blood supply (reperfusion) upon removal of tumor.
  • the hemoglobin-based oxygen carrier can also be used to increase oxygenation of cancerous tissues and with chemotherapeutic agents then subsequently reducing the size of a tumor.
  • the hemoglobin-based oxygen carrier-containing composition of the present invention can be administered alone or in combination with at least a chemotherapeutic agent for treating or preventing the recurrence of cancerous tumors.
  • the method of the present invention also includes using a combination of different chemotherapeutic drugs and/or radiotherapy with the hemoglobin-based oxygen carrier of the present invention to give a synergistic effect on cancer treatment and prevention of tumor recurrence.
  • the third aspect of the present invention relates to the composition of the present invention for providing oxidative stress or shock to the tumor in order to kill a rare population of self-renewing and tumor-initiating cells known as cancer stem cells.
  • the composition of the present invention for providing high oxygen tension to the tumor includes a hemoglobin-based oxygen carrier which includes tetrameric cross-linked hemoglobin or polymerized hemoglobin, where both of them are prepared to contain an undetectable amount of dimer and low percentage of met-hemoglobin.
  • the hemoglobin-based oxygen carrier in said composition is configured for penetration into the cancerous tissues of the tumor where the cancer stem cells are found to selectively proliferate within the tumor.
  • Said hemoglobin-based oxygen carrier can be used alone or in combination with at least one chemotherapeutic agent including Bortezomib, 5-fluorouracil, doxorubicin, cisplatin, or any combination thereof for oxygenating the tumor and providing oxidative stress or shock to said cancer stem cells in order to induce apoptosis or death of said cancer stem cells, which result in the effect in the treatment of and preventing from the recurrence of cancer or cancerous tumor.
  • the hemoglobin-based oxygen carrier of the present invention is also modified to avoid dissociation into dimer such that it becomes more stable and has a longer half life in the circulation.
  • the hemoglobin-based oxygen carrier in the composition of the present invention also sensitize the cancer stem cells to chemotherapeutic agent or radiotherapy.
  • the composition of the present invention is an effective adjunctive therapy which can be administered prior to or in combination with chemotherapy and/or radiotherapy.
  • the hemoglobin-based oxygen carrier can be administered to a subject in needs thereof at a concentration of 9.5 g/dL-10.5 g/dL for the purpose(s) of targeting the cells in the cancerous tissues or tumors, triggering the receptor-mediated mechanism, penetrating and being localized into the cancerous tissue or tumor cells, inducing apoptosis of the cancerous tissue or tumor cells, sensitizing the cells to the chemotherapeutic agent or radiotherapy which is administered concurrently or subsequently, either before, during or after a surgical removal of the cancerous tissue or tumor.
  • FIG. 1 is a set of microscopic images in the same magnification showing the uptake of (A) fluorescent-labeled heat stable hemoglobin-based oxygen carrier and (B) fluorescent-labeled polymerized hemoglobin into liver cancer cells.
  • FIG. 2 is two sets of microscopic images in the same magnification showing the uptake of fluorescent-labeled heat stable hemoglobin-based oxygen carrier into liver cancer cells via the Clathrin mediated pathway (upper panel) but not via Caveolin-1 mediated pathway (lower panel).
  • FIG. 3 shows the expression of different proteins in liver cancer cells after treating with the heat stable hemoglobin-based oxygen carrier in different concentrations.
  • FIG. 4 shows the expression of hypoxia-inducible factor 1 (HIF1 ⁇ ) gene in liver cancer cells (HepG2 and Huh7) after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • HIF1 ⁇ hypoxia-inducible factor 1
  • FIG. 5 shows the expression of Vascular Endothelial Growth Factor (VEGF) gene in liver cancer cells (HepG2 and Huh7) after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • VEGF Vascular Endothelial Growth Factor
  • FIG. 6 shows the expression of endothelin-1 (ET1) gene in liver cancer cells (HepG2 and Huh7) after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • FIG. 7 shows the expression of inducible nitric oxide synthase (iNOS) gene in liver cancer cells (HepG2 and Huh7) after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • iNOS inducible nitric oxide synthase
  • FIG. 8 shows the expression of von Hippel-Lindau (VHL) gene in liver cancer cells (HepG2 and Huh7) after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • VHL von Hippel-Lindau
  • FIG. 9 shows the expression of a heat shock protein 90 (HSP90) gene in liver cancer cells after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • HSP90 heat shock protein 90
  • FIG. 10 is a schematic diagram illustrating the proposed mechanism and signaling cascade involved in the inhibitory effect of the heat stable hemoglobin-based oxygen carrier on tumor recurrence.
  • FIG. 11 shows the expression of the heat shock protein 7C (HSP7C) gene in liver cancer cells after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • HSP7C heat shock protein 7C
  • FIG. 12 shows the expression of high-mobility group box 3 (HMGB3) gene in liver cancer cells after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • HMGB3 high-mobility group box 3
  • FIG. 13 shows the expression of replication factor 1C (RFC1) gene in liver cancer cells after treating with different concentrations of heat stable hemoglobin-based oxygen carrier and under normoxic vs hypoxic conditions.
  • FIG. 14 shows an improvement of oxygenation in normal tissue. Injection of 0.2 g/kg heat stable tetrameric hemoglobin solution results in a significant increase in (A) plasma hemoglobin concentration and (B) oxygen delivery to muscle.
  • FIG. 15 shows an improvement of oxygenation in hypoxic tumor tissue. Injection of 0.2 g/kg heat stable tetrameric hemoglobin solution results in a significant increase in oxygen delivery to the head and neck squamous cell carcinoma (HNSCC) xenograft.
  • HNSCC head and neck squamous cell carcinoma
  • FIG. 16 shows partial tumor shrinkage in rodent models of (A) nasopharyngeal carcinoma (NPC) and (B) liver tumor.
  • FIG. 17 shows a schematic drawing summarizing the surgical and hemoglobin product administration procedures during liver resection.
  • FIG. 18 shows representative examples of intra-hepatic liver cancer recurrence and metastasis and distant lung metastasis induced in the rats of the IR injury group after hepatectomy and ischemia/reperfusion procedures and its protection using the inventive heat stable tetrameric hemoglobin.
  • FIG. 19 shows the histological examination in experimental and control groups at four weeks after liver resection and IR injury procedures.
  • FIG. 20A shows the volume (cm 3 ) of recurred liver tumor found in rats of the IR injury group (Control group) after hepatectomy and IR procedures and rats having treated with the inventive heat stable tetrameric hemoglobin (Hb Treatment group).
  • FIG. 20B shows the liver recurrence rate (left) and the average recurred tumor size (right) of the IR injury rats after hepatectomy and IR procedures (Control group) and rats having treated with the inventive heat stable tetrameric hemoglobin (Hb group).
  • FIG. 21 shows representative examples of intra-hepatic liver cancer recurrence and metastasis and distant lung metastasis induced in the rats of the IR injury group after hepatectomy and ischemia/reperfusion procedures (control group: C10 & C13) and rats treated with the inventive heat stable tetrameric hemoglobin (Hb treatment group: Y9, Y10 & Y11).
  • FIG. 22 shows the representative examples of liver oxygen partial pressure (mmHg) from the first administration of the subject inventive hemoglobin product or RA buffer (control) throughout the hepatic surgery and reperfusion.
  • FIG. 23 shows a comparison between levels of circulating endothelial progenitor cells (EPC) in peripheral blood of rats with or without treatment of the subject hemoglobin product 28 days post-hepatic surgery.
  • EPC circulating endothelial progenitor cells
  • FIG. 24 shows the temporal localization of the heat-stable hemoglobin-based oxygen carrier within nasopharyngeal carcinoma Xenograft.
  • FIG. 25 shows the tumor growth inhibitory effect of the hemoglobin-based oxygen carrier alone or combined with radiation in a Hep-2 laryngeal cancer model; lower panel shows the representative image of tumor xenografts obtained from different treatment groups. *p ⁇ 0.05, **p ⁇ 0.01 versus control.
  • FIG. 26 shows the tumor growth inhibitory effect of the hemoglobin-based oxygen carrier combined with radiation in a C666-1 nasopharyngeal cancer model; lower panel shows representative image of tumor xenografts obtained from different treatment groups. **p ⁇ 0.01 versus control, #p ⁇ 0.05 versus radiation treatment only.
  • FIG. 27 shows the hemoglobin-based oxygen carrier enhances temozolomide (TMZ)-induced cytotoxicity in brain cancer cells.
  • TMZ temozolomide
  • FIG. 28 are microscopic images showing the morphological change of mammospheres formation by cancer stem cells: (A) Day 0, (B) Day 3, (C) Day 6, (D) Day 9-20, (E) Control (hollow mammospheres from mammary epithelial cells).
  • FIG. 29 are western blots showing the expression level of different markers Oct-4 and Sox-2 in unsorted mammospheres and sorted MCF7 CD44 + /CD24 ⁇ cells collected from different passages.
  • FIG. 30 are dot plots of different passages of MCF7 cells in terms of the aldehyde dehydrogenase (ALDH) activity: (A) Control (sorted MCF7 cells incubated with diethylaminobenzaldehyde (DEAB)); (B) sorted MCF7 cells at passage 0; (C) sorted MCF7 cells at passage 3; (D) sorted MCF7 cells at passage 5.
  • A Control (sorted MCF7 cells incubated with diethylaminobenzaldehyde (DEAB));
  • B sorted MCF7 cells at passage 0;
  • C sorted MCF7 cells at passage 3;
  • D sorted MCF7 cells at passage 5.
  • FIG. 31 is dot plots of MCF7 cells under hypoxic conditions and labeled with CD24 (PE-A) and CD44 (APC-A) antibodies in a flow cytometry analysis.
  • Quadrant 1 (Q1) are cells which are CD44 high and CD24 low .
  • FIG. 32 are microscopic images showing the morphological change of unsorted and CD24/CD44-sorted MCF7 cells after incubated with DMSO (Control) and 90 nM Taxol treatment for 16 hours and 4 days.
  • cancer stem cell refers to the biologically distinct cell within the neoplastic clone that is capable of initiating and sustaining tumor growth in vivo (i.e. the cancer-initiating cell).
  • Ha used herein refers to cross-linked tetrameric hemoglobin which is heat stable with undetectable amount of dimers and low percentage of met-hemoglobin.
  • the heat stable cross-linked tetrameric hemoglobin has a molecular weight of 60-70 kDa which is heat treated and added with 0.05%-0.4% of N-acetyl cysteine during the synthesis.
  • the resulting heat stable cross-linked tetrameric hemoglobin has undetectable amount of dimers and less than 5% of met-hemoglobin.
  • the heat stable cross-linked tetrameric hemoglobin is also free of vasoconstricting impurities and protein impurities, non-pyrogenic, endotoxin-free, phospholipid-free, and stroma-free.
  • the cross-linking within the tetrameric hemoglobin molecule can be between alpha/alpha subunits, alpha/beta subunits or alpha-beta subunits.
  • Modified hemoglobin or “Recombinant hemoglobin” defined herein refers to any natural hemoglobin or purified hemoglobin which is either chemically conjugated with or surface modified with at least one compound. Said compound may include poly(ethylene) glycol (PEG).
  • PEG poly(ethylene) glycol
  • One of the examples of the modified hemoglobin used in the present invention is pegylated hemoglobin.
  • Hemoglobin is an iron-containing oxygen-transport protein in red blood cells of the blood of mammals and other animals. Hemoglobin exhibits characteristics of both the tertiary and quaternary structures of proteins. Most of the amino acids in hemoglobin form alpha helices connected by short non-helical segments. Hydrogen bonds stabilize the helical sections inside the hemoglobin causing attractions within the molecule thereto folding each polypeptide chain into a specific shape.
  • a hemoglobin molecule is assembled from four globular protein subunits. Each subunit is composed of a polypeptide chain arranged into a set of ⁇ -helix structural segments connected in a “myoglobin fold” arrangement with an embedded heme group.
  • the heme group consists of an iron atom held in a heterocyclic ring, known as a porphyrin.
  • the iron atom binds equally to all four nitrogen atoms in the center of the ring which lie in one plane. Oxygen is then able to bind to the iron center perpendicular to the plane of the porphyrin ring.
  • a single hemoglobin molecule has the capacity to combine with four molecules of oxygen.
  • hemoglobin A In adult humans, the most common type of hemoglobin is a tetramer called hemoglobin A consisting of two ⁇ and two ⁇ non-covalently bound subunits designated as ⁇ 2 ⁇ 2, each made of 141 and 146 amino acid residues respectively.
  • the size and structure of ⁇ and ⁇ subunits are very similar to each other. Each subunit has a molecular weight of about 16 kDa for a total molecular weight of the tetramer of about 65 kDa.
  • the four polypeptide chains are bound to each other by salt bridges, hydrogen bonds and hydrophobic interaction.
  • the structure of bovine hemoglobin is similar to human hemoglobin (90.14% identity in ⁇ chain; 84.35% identity in ⁇ chain).
  • the difference is the two sulfhydryl groups in the bovine hemoglobin positioned at ⁇ Cys 93, while the sulfhydryls in human hemoglobin are at positioned at ⁇ Cys 104, ⁇ Cys 93 and ⁇ Cys 112 respectively.
  • hemoglobin In naturally-occurring hemoglobin inside the red blood cells, the association of an ⁇ chain with its corresponding ⁇ chain is very strong and does not disassociate under physiological conditions. However, the association of one ⁇ dimer with another ⁇ dimer is fairly weak outside red blood cells. The bond has a tendency to split into two ⁇ dimers each approximately 32 kDa. These undesired dimers are small enough to be filtered by the kidneys and be excreted, with the result being potential renal injury and substantially decreased intravascular retention time. Therefore, stabilized cross-linked tetrameric, polymeric and/or recombinant/modified hemoglobin are the important molecule in a pharmaceutical composition for oxygen delivery.
  • the source of hemoglobin can be from, but not limited to, human, bovine, porcine, equine, and canine whole blood.
  • the pharmaceutical composition of the present invention contains a heat stable hemoglobin-based oxygen carrier which is configured to attach to receptors on tumor cells to facilitate selective targeting of hypoxic tumor cells over normal, non-hypoxic healthy tissue and that can be used in cancer treatment as it can be taken up preferentially into cancer cells.
  • a heat stable hemoglobin-based oxygen carrier which is configured to attach to receptors on tumor cells to facilitate selective targeting of hypoxic tumor cells over normal, non-hypoxic healthy tissue and that can be used in cancer treatment as it can be taken up preferentially into cancer cells.
  • FIG. 1A live cell imaging is used to show how the heat-stable tetrameric hemoglobin (Hb) has efficacy against liver cancer.
  • a fluorescently-conjugated Hb is prepared by allowing conjugation between Hb and fluorescein isothiocyanate (FITC) (buffered with NaHCO 3 at pH9.3) for 1 hour at room temperature in an enclosed system purged with N 2 .
  • FITC fluorescein iso
  • Hb-FITC protein purification columns
  • the freshly conjugated Hb-FITC probe is immediately employed for live cell uptake studies.
  • Liver cancer cells, HepG2, and the metastatic liver cancer cells, Huh7, are exposed to 0.0125 g/dL for 15 min prior to live cell acquisition.
  • the uptake of Hb-FITC into both types of liver cancer cells after 15 min of exposure is observed ( FIG. 1A ).
  • the uptake of Hb-FITC peaks after 1 hour of exposure ( FIG. 1A ).
  • the monolayer liver cancer cells are observed to curl-up into a three-dimensional structure, and Hb-FITC is detected to be more preferentially taken up by these cancer cells than normal cells.
  • the uptake of polymerized hemoglobin into liver cancer cell is shown in FIG. 1B .
  • Hb-FITC colocalizes with RFP-Clathrin, but not mCherry-Caveolin1, suggesting that hemoglobin molecule enters into liver cancer cells through Clathrin-mediated endocytosis.
  • the IC 50 of Cisplatin, Doxorubicin, Bortezomib, and 5-fluorouracil (5FU) in HepG2 cells are 130 uM, 10 uM, 0.5 uM, and 4 mM respectively, and the IC 50 of Cisplatin, Doxorubicin, Bortezomib, and 5FU in Huh7 cells are 70 uM, 5 uM, 55 uM, and 3.5 mM respectively.
  • the IC 50 of Cisplatin, Doxorubicin, Bortezomib, and 5FU in HepG2 cells are 170 uM, 30 uM, 0.7 uM, and 4 mM respectively, and the IC 50 of Cisplatin, Doxorubicin, Bortezomib, and 5FU in Huh7 cells are 100 uM, 6 uM, 60 uM, and 4 mM respectively.
  • the 3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay result suggests that under normoxic condition, Huh7 cells are more sensitive to Cisplatin and Doxorubicin, but are 110-fold more resistance to Bortezomib as compared to HepG2 cells under normoxic condition (a target drug against the proteasomal subunits PSMB1, 5 and 6). Under hypoxic condition, Huh7 cells become more sensitive to Cisplatin and Doxorubicin, and are also highly resistant to Bortezomib (86-fold) as compared to HepG2 cells under hypoxic condition. The results reveal that metastatic liver cancer cells (Huh7) are generally more resistant to Bortezomib than non-metastatic liver cancer cells (HepG2) notwithstanding under normoxic or hypoxic condition.
  • 5FU is a pyrimidine analog that inhibits thymidylate synthase.
  • Bortezomib is the first therapeutic proteasome inhibitor used initially for treating myeloma patients. It is reported to cause apoptosis in liver cancer cells (Koschny et AL., Hepatology, 2007). Taken together, hemoglobin molecule is observed to have significant synergistic effects with 5FU and Bortezomib on both non-metastatic and metastatic cancer.
  • hypoxia is a common physiological feature of tumors. Intratumoural hypoxia is also common in liver cancer. The condition of hypoxia is known to activate a signaling cascade that results in the stabilization of the hypoxia-inducible factor 1 (HIF1 ⁇ ) transcription factor and activation of HIF1 ⁇ effector genes (over 60 genes) that possess a hypoxia response element (HRE).
  • HIF1 ⁇ downstream effectors are involved in cell survival, adaptation, anaerobic metabolism, immune reaction, cytokine production, vascularization and general tissue homeostasis.
  • Hb is demonstrated to affect HIF1 ⁇ protein expression in the HepG2 and the metastatic Huh7 liver cancer models.
  • Hb downregulates HIF1 ⁇ in both normoxia and hypoxia, suggesting that the depletion of HIF1 ⁇ by Hb alone (40% compared with untreated control) affects the binding of HIF1 ⁇ to its downstream effectors and results in transcriptional repression of these effector genes.
  • Similar downregulation patterns can be detected in the upstream regulators of HIF1 ⁇ ( FIG. 4 ), heat shock protein 90 (HSP90) ( FIG. 9 ) and von Hippel-Lindau (VHL) ( FIG. 8 ), after treatment with Hb.
  • FIG. 10 The proposed mechanism involved in the inhibitory effect of Hb on tumor recurrence and its signaling cascade is illustrated in FIG. 10 .
  • PDH prolyl hydroxylase domain-containing protein
  • EPC endothelial progenitor cell
  • a pharmaceutical composition including a hemoglobin-based oxygen carrier configured to target DNA-damage-sensing cell regulation apparatus is also found to go through novel regulatory pathways.
  • two of the proteins which are the intrinsic parts of the DNA-damage-sensing apparatus RRC1 ( FIG. 13 ) and the HSP7C (heat shock protein 7C) ( FIG. 11 )
  • RFC1 replication factor 1C
  • HSP7C heat shock protein 7C
  • Hb is a potential Reactive Oxygen Species (ROS) inducer, and it is clearly important for the metastatic liver cancer cells, Huh7, to sense and respond to the ROS-mediated DNA damage.
  • ROS Reactive Oxygen Species
  • the oxygen carrier-containing pharmaceutical composition of the present invention serves as a tissue oxygenation agent to improve the oxygenation in tumor tissues, thereby enhancing chemosensitivity and radiation sensitivity.
  • HNSCC head and neck squamous cell carcinoma
  • FaDu a representative oxygen profile of a human head and neck squamous cell carcinoma (HNSCC) xenograft (FaDu) is shown in FIG. 15 .
  • HNSCC head and neck squamous cell carcinoma
  • FIG. 15 After intravenous injection of 0.2 g/kg of the heat stable tetrameric hemoglobin, a significant increase in the mean pO 2 of more than 6.5-fold and 5-fold is observed at 3 and 6 hours, respectively ( FIG. 15 ).
  • the oxygen carrier-containing pharmaceutical composition of the present invention serves as a tissue oxygenation agent to improve the oxygenation in tumor tissues, thereby enhancing chemo- and radiation sensitivity.
  • tumor growth is delayed.
  • the representative curves show significant tumor shrinkage in rodent models of nasopharyngeal carcinoma. Nude mice bearing CNE2 xenografts are treated with X-ray alone (2Gy) or in combination with the heat stable tetrameric hemoglobin (2Gy+Hb).
  • 1.2 g/kg of the heat stable tetrameric hemoglobin is injected intravenously into the mouse approximately 3 to 6 hours before X-ray irradiation and results in a partial shrinkage of nasopharyngeal carcinoma xenograft.
  • significant liver tumor shrinkage is observed after injecting the composition, in conjunction with a chemotherapeutic agent.
  • FIG. 16B the representative chart shows significant tumor shrinkage in a rat orthotopic liver cancer model. Buffalo rats bearing a liver tumor orthograft (CRL1601 cell line) are treated with 3 mg/kg cisplatin alone, or in combination with 0.4 g/kg of the heat stable tetrameric hemoglobin (Cisplatin+Hb). Administration of the heat stable tetrameric hemoglobin before cisplatin injection results in a partial shrinkage of the liver tumor.
  • HepG2 and Huh7 cell lines are used. These cells are cultured in DMEM (Invitrogen) with 10% Fetal bovine serum (FBS), 100 U/ml penicillin and 100 ⁇ g/ml streptomycin at 37° C. For normoxic condition, cells are incubated with ambient O 2 concentration and 5% CO 2 , for hypoxic condition, cells are incubated with 0.1-0.5% O 2 (Quorum FC-7 automatic CO 2 /O 2 /N 2 gas mixer) and 5% CO 2 .
  • FBS Fetal bovine serum
  • FBS Fetal bovine serum
  • hypoxic condition cells are incubated with 0.1-0.5% O 2 (Quorum FC-7 automatic CO 2 /O 2 /N 2 gas mixer) and 5% CO 2 .
  • HepG2 or Huh7 cells are seeded onto glass bottom microwell dishes (MatTek Corporation). Live cells at defined zooms (63 ⁇ , 20 ⁇ ) are acquired using Zeiss Observer.Z1 widefield microscope, equipped with atmospheric/temperature-controlled chamber and motorized stage for multi-positional acquisition. The incubation is performed in an enclosed live cell imaging system purged with 0.1% O 2 and 5% CO 2 (premixed). Cells transfected with pcDNA3, pRFP-Caveolin1, or pRFP-Clathrin are exposed to HB-FITC for 15 min prior to the acquisition of images every 3 min for a period of 2 hours. Images are deconvolved and compacted into time-lapse movies using the MetaMorph software (Molecular Device).
  • Cell viability is measured using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) proliferation assay. Briefly, HepG2 or Huh7 cells are seeded in a 96-well flat-bottomed microplate (6000 cells/well) and cultured in 100 ⁇ L growth medium at 37° C. and 5% CO 2 for 24 h. Cell culture medium in each well is then replaced by 100 ⁇ L cell growth medium, containing either no drug, Hb alone or Hb with another chemotherapeutics at their IC 50 concentrations.
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • RNA is isolated using the Trizol reagent (Invitrogen) and 5 ⁇ g of the total RNA is reverse transcribed with an oligo-dT primer and Superscript II reverse transcriptase (Invitrogen).
  • One-tenth of the first strand cDNA is used for quantitative measurements of HIF1alpha, VHL, HSP90, VEGF, iNOS, ET1, HSP7c, RFC1, HMGB3, and GAPDH transcript levels by the SYBR Green PCR Master Mix kit (Applied Biosystems) with specific primers (shown below).
  • the fluorescence signals are measured in real time during the extension step by the 7900HT Fast Real Time PCR System (Applied Biosystems).
  • the threshold cycle (Ct) is defined as the fractional cycle number at which the fluorescence signal reached 10-fold standard deviation of the baseline (from cycles 2 to 10).
  • the ratio change in the target gene relative to the GAPDH control gene is determined by the 2 ⁇ ct method.
  • HIF1 ⁇ SEQ NO. 1: Forward Primer: 5-GGCGCGAACGACAAGAAAAAG-3 (420-440)
  • SEQ NO. 2 Reverse Primer: 5-CCTTATCAAGATGCGAACTCACA-3 (21-44)
  • SEQ NO. 3 Forward Primer: CAGAGCAGGAAAAGGAGTCA (2414-2433)
  • SEQ NO. 4 Reverse Primer: AGTAGCTGCATGATCGTCTG (2645-2625)
  • SEQ NO. 5 Forward Primer: 5′-AATGGAATGGAGCAAAAGACAATT-3′ (2694-2720)
  • SEQ NO. 6 Reverse Primer: 5′-ATTGATTGCCCCAGCAGTCTAC-3′ (2764-2743)
  • VEGF SEQ NO.
  • Proteins are harvested and protein concentrations are determined. Protein (30 ⁇ g) is resolved on 10% SDS-PAGE, transferred onto a nitrocellulose membrane (PVDF, BioRad). Actin is used as loading control. Relative protein expression levels are quantified by gel documentation system (Ultra-Violet Product Ltd).
  • FIG. 14 Some studies for the normal tissue oxygenation by heat stable tetrameric hemoglobin are carried out (shown in FIG. 14 ).
  • a comparative pharmacokinetic and pharmacodynamic study is conducted in buffalo rats. Male inbred buffalo rats are individually administered with 0.2 g/kg heat stable tetrameric hemoglobin solution or ringer's acetate buffer (control group).
  • the concentration-time profile of plasma hemoglobin is determined by HemocueTM photometer at 1, 6, 24, 48 hours and compared with the baseline reading. The methods are based on photometric measurement of hemoglobin where the concentration of hemoglobin is directly read out as g/dL.
  • Oxygen partial pressure (pO 2 ) is directly measured by the OxylabTM tissue oxygenation and temperature monitor (Oxford Optronix Limited) in hind leg muscle of buffalo rats. Rats are anesthetized by intra-peritoneal injection of 30-50 mg/kg pentobarbitone solution followed by insertion of oxygen sensor into the muscle. All pO 2 readings are recorded by Datatrax2 data acquisition system (World Precision Instrument) in a real-time manner Results demonstrate that after an intravenous injection of 0.2 g/kg of the heat stable tetrameric hemoglobin, the mean pO 2 value rises from baseline to about two-fold of the relative mean oxygen partial pressure within 15 minutes and extends to 6 hours. Further, the oxygen level on average is still maintained at 25% to 30% above the baseline value 24 to 48 hours post injection ( FIG. 14B ).
  • HNSCC head and neck squamous cell carcinoma
  • FaDu cell line A hypopharyngeal squamous cell carcinoma (FaDu cell line) is obtained from the American Type Culture Collection. Approximately 1 ⁇ 10 6 cancer cells are injected subcutaneously into four to six week-old inbred BALB/c AnN-nu (nude) mice. When the tumor xenograft reaches a diameter of 8-10 mm, oxygen partial pressure (pO 2 ) within the tumor mass is directly monitored by the OxylabTM tissue oxygenation and temperature monitor (Oxford Optronix Limited).
  • FIG. 16A A significant tumor shrinkage is observed after administration of heat stable tetrameric hemoglobin solution in combination with X-ray irradiation ( FIG. 16A ).
  • a human nasopharyngeal carcinoma xenograft model is employed. Approximately 1 ⁇ 10 6 cancer cells (CNE2 cell line) are injected subcutaneously into four to six week-old inbred BALB/c AnN-nu (nude) mice. When the tumor xenograft reaches a diameter of 8-10 mm, tumor-bearing mice are randomized into three groups as follows:
  • mice bearing CNE2 xenografts are irradiated with X-irradiation alone (Group 2) or in combination with heat stable tetrameric hemoglobin (Group 3).
  • mice are anesthetized by an intra-peritoneal injection of 50 mg/kg pentobarbitone solution. 2 Grays of X-ray is delivered to the xenograft of tumor-bearing mice by a linear accelerator system (Varian Medical Systems).
  • 1.2 g/kg heat stable tetrameric hemoglobin is injected intravenously through the tail vein into the mouse before X-ray treatment. Tumor dimensions and body weights are recorded every alternate day starting with the first day of treatment.
  • Tumor weights are calculated using the equation 1 ⁇ 2 ⁇ LW 2 , where L and W represent the length and width of the tumor mass, measured by a digital caliper (Mitutoyo Co, Tokyo, Japan) at each measurement.
  • Group 1 is the non-treatment control group.
  • Results show that significant shrinkage of the CNE2 xenograft is observed in mice treated with the heat stable tetrameric hemoglobin solution in conjunction with X-irradiation (Group 3, FIG. 16A ).
  • FIG. 16B A rat orthotopic liver cancer model is employed. Approximately 2 ⁇ 10 6 rat liver tumor cells labeled with luciferase gene (CRL1601-Luc) are injected into the left lobe of the liver in a buffalo rat. Tumor growth is monitored by a Xenogen in vivo imaging system. Two to three weeks after injection, the tumor tissue is harvested, dissected into small pieces and orthotopically implanted into the left liver lobe of a second group of rats. Rats bearing liver tumor are randomized into three groups as follows:
  • Rats implanted with liver tumor tissue are treated with 3 mg/kg of cisplatin alone (Group 2) or in conjunction with heat stable tetrameric hemoglobin (Group 3).
  • rats are anesthetized by an intra-peritoneal injection of 30-50 mg/kg pentobarbitone solution and cisplatin are administered via the left portal vein.
  • 0.4 g/kg heat stable tetrameric hemoglobin is injected intravenously before cisplatin treatment.
  • Group 1 is the non-treatment control group.
  • a significant shrinkage of liver tumor is observed 3 weeks after treatment ( FIG. 16B ).
  • Surgical resection of liver tumors is a frontline treatment of liver cancer.
  • post-operative recurrence and metastasis of cancer remains a major attribute of unfavorable prognosis in these patients.
  • previous studies reported that hepatic resection is associated with a 5-year survival rate of 50% but also a 70% recurrence rate.
  • follow-up studies on hepatocellular carcinoma (HCC) patients also reveal that extrahepatic metastases from primary HCC were detected in approximately 15% of HCC patients with the lungs being the most frequent site of extrahepatic metastases.
  • surgical stress, especially ischemia/reperfusion (IR) injury introduced during liver surgery is a major cause of tumor progression.
  • hepatic vascular control is commonly used by surgeons to prevent massive hemorrhage during hepatectomy.
  • inflow occlusion by clamping of the portal triad has been used to minimize blood loss and reduce the requirement of perioperative transfusions.
  • Pringle maneuver induces various degrees of ischemic injury in the remnant liver and is associated with cancer recurrence and metastasis.
  • IR injury and tumor progression are also supported by previous animal studies. Firstly, the effect of IR injury and hepatic resection on liver cancer recurrence and metastasis was demonstrated in a recent study with an orthotopic liver cancer model. Hepatic IR injury and hepatectomy resulted in prominent recurrence and metastasis of liver tumors. Similar results were obtained in a colorectal liver metastasis mouse model where introduction of IR injury accelerates the outgrowth of colorectal liver metastasis.
  • IP ischemic preconditioning
  • hypoxia inducible factor-1 HIF-1
  • VEGF vascular endothelial growth factor
  • CTCs circulating cancer cells
  • the cross-linked tetrameric hemoglobin of the present invention is used to prevent post-operative liver tumor recurrence and metastasis following hepatic resection.
  • a rat orthotopic liver cancer model is established.
  • Hepatocellular carcinoma cell line (McA-RH7777 cells) is used to establish the orthotopic liver cancer model in Buffalo rats (Male, 300-350 g).
  • FIG. 17 shows a schematic drawing summarizing the surgical and hemoglobin product administration procedures.
  • McA-RH7777 cells (approximately 1 ⁇ 10 6 cells/100 ⁇ L) are injected into the hepatic capsule of buffalo rat to induce solid tumor growth.
  • tumor tissue is collected and cut into 1-2 mm 3 cubes and implanted into the left liver lobes of a new group of buffalo rats.
  • the rats undergo liver resection (left lobe bearing liver tumor) and partial hepatic IR injury (30 minutes of ischemia on right lobe).
  • rats Two groups of rats with implanted tumor tissue are used for comparison of tumor recurrence and metastases.
  • rats are anesthetized with pentobarbital and administered intravenously with 0.2 g/kg at a concentration of 10 g/dL of the heat stable tetrameric hemoglobin of the present invention 1 hour before ischemia.
  • Ischemia is introduced in the right lobe of the liver by clamping of right branches of hepatic portal vein and hepatic artery with a bulldog clamp. Subsequently, ligation is performed in the left liver lobe followed by resection of the left liver lobe bearing the liver tumor.
  • liver and lungs of Buffalo rats are sampled at 4 weeks after Ischemia/reperfusion and hepatectomy procedures for morphological examination. Tissue is harvested, parafilm-embedded and sectioned followed by Hematoxylin and Eosin (H&E) staining. Local recurrence/metastasis (intrahepatic) and distant metastasis (lungs) are confirmed by histological examination.
  • Table 2 summarizes the comparison of tumor recurrence/metastasis at four weeks after liver resection and IR injury in a rat orthotopic liver cancer model.
  • FIG. 18 shows representative examples of intra-hepatic liver cancer recurrence and metastasis and distant lung metastasis induced in the rats of the IR injury group after hepatectomy and ischemia/reperfusion procedures and its protection using the inventive heat stable tetrameric hemoglobin.
  • FIG. 18A extensive intrahepatic liver cancer recurrence/metastasis is observed in the IR injury group. Distant lung metastasis is also occurred in the same rat (indicated by a solid arrow).
  • FIG. 18B intrahepatic liver cancer recurrence/metastasis is observed in another case in the IR injury group (indicated by a dotted arrow).
  • FIG. 18C shows a representative example of protection from intrahepatic liver cancer recurrence/metastasis and distant lung metastasis in the inventive heat stable tetrameric hemoglobin treated rat.
  • FIG. 19 shows the histological examination in both groups at four weeks after liver resection and IR injury procedures. Histological examination (H&E staining) of liver and lung tissues in both the IR injury and hemoglobin treatment groups is performed to confirm the identity of the tumor nodules. Representative fields showing intrahepatic recurrence (T1 and T2) and lung metastasis (M) in the IR injury group are shown (top). Histological examination showing a normal liver architecture in the treatment group (N1) and a tumor nodule detected in the liver after hemoglobin treatment (T3) are included for comparison (bottom). In addition, lung tissue without metastasis is shown in the treatment group (N2) for comparison.
  • H&E staining Histological examination showing intrahepatic recurrence (T1 and T2) and lung metastasis (M) in the IR injury group are shown (top).
  • recurrence rate of tumor and size of the recurred tumor post-ischemia/reperfusion and hepatectomy procedures are investigated.
  • rats with implanted tumor tissue prepared by injection of McA-RH7777 cells as described above are treated intravenously with either approximately 0.2-0.4 g/kg of the heat stable tetrameric hemoglobin of the present invention or Ringer's acetate (RA) buffer as a negative control prior to ischemia and at reperfusion upon hepatic resection procedure as described in FIG. 17 .
  • RA Ringer's acetate
  • a total of 26 rats are tested, where 13 rats are treated with the subject hemoglobin and 13 are negative control rats which are merely treated with RA buffers. All rats are sacrificed 4 weeks after the hepatectomy and IR procedures, livers and lungs of the test rats are examined for tumor recurrence/metastasis and the relative size of the recurred tumors are measured.
  • FIG. 20A shows liver tumor recurrence in test rats and the volume of individual recurred tumors. Liver tumor recurred/metastasis in 9 of the 13 non-treated control rats, whereas only 4 of the 13 treated rats experienced tumor recurrences/metastasis. It is also evident that where tumor recurrence is seen, the sizes of the recurred tumors of rats having treated with the subject hemoglobin are significantly smaller than those untreated. The results show that tumor recurrence rate is greatly reduced and recurred tumor size is significantly reduced with treatment of the subject invention, as summarized in FIG. 20B .
  • FIG. 21 illustrates representative examples of liver and lung tissues harvested 4 weeks post hepatectomy and IR procedures of rats having treated with the subject inventive heat stable tetrameric hemoglobin and the IR injury (negative control) group.
  • rats C10 and 13 extensive intrahepatic liver cancer recurrence/metastasis and distant lung metastasis are observed (circled).
  • intrahepatic liver cancer recurrence/metastasis and distant lung metastasis are prevented by the treatment of the subject inventive hemoglobin, as seen in rats Y9, Y10 and Y11.
  • FIG. 22 shows that increased oxygenation with the subject hemoglobin treatment is observed after introduction of ischemia.
  • the liver having treated with the subject hemoglobin has approximately 3-fold higher oxygen partial pressure than without treatment after reperfusion. It is confirmed that the treatment of the subject hemoglobin prior to ischemia and at reperfusion upon tumor resection significantly improves the oxygenation of the liver tissue as compared to non-treatment.
  • CSCs cancer stem cells
  • EPCs circulating endothelial progenitor cells
  • the level of circulating EPCs is evaluated by expression of surface molecules such as CD133, CD34 and VEGFR2.
  • the circulating endothelial progenitor cell levels post-hepatic resection surgery and IR procedure with or without the treatment of the subject hemoglobin product is investigated.
  • Two groups of rats with implanted hepatic tumor are subjected to treatment of the subject hemoglobin or RA buffer (control), respectively prior to ischemia and at reperfusion upon hepatic resection as shown in FIG. 17 .
  • Number of circulating EPC of the two group of rats are then measured at 0, 3, 7 14, 21 and 28 days after hepatic resection and IR procedures. Results ( FIG.
  • inventive hemoglobin is labeled with Alexa Fluor® 750 SAIVITM Antibody Labeling System according to manufacturer's instruction.
  • fluorescently labeled inventive hemoglobin fl-Hb
  • unlabeled counterpart in a ratio of approximately 1:80.
  • the mixture is injected intravenously into nude mice bearing nasopharyngeal carcinoma xenograft (C666-1).
  • the amount of fl-Hb is around 0.2 mg to ensure sufficient fluorescent signal to be captured by the Maestro2 imaging system.
  • Nude mice are anesthetized at different time points before exposure to the Maestro2 fluorescent imaging system for analysis.
  • FIG. 24 shows representative image of Hb concentrated within the tumor xenograft (indicated by an arrow).
  • the hemoglobin-based oxygen carrier of the present invention is administered once before radiation, and the result shows that tumor growth inhibitory effects in the Hep-2 laryngeal cancer model.
  • the tumor volume of high dose of Hb (2.2 g/kg) combined with radiation at the end of experiment is 90.0 mm 3 , which is significantly smaller than the control group (336.1 mm 3 ) (P ⁇ 0.01).
  • the tumor volume of radiation alone is 143.1 mm 3 , and the combination q value of administering a high dose of Hb is 1.17, indicating a synergistic effect of this combination (q>1.15, synergistic effect).
  • FIG. 25 shows the tumor growth inhibition effects of the hemoglobin-based oxygen carrier of the present invention followed by radiation.
  • the hemoglobin-based oxygen carrier of the present invention is administered once before radiation, and the result shows that tumor growth inhibitory effect in the C666-1 nasopharyngeal cancer model.
  • the tumor volume of high dose of Hb (2.2 g/kg) combined with radiation at the end of experiment is 110.3 mm 3 , which is significantly smaller compared with the control group (481.1 mm 3 ) (P ⁇ 0.01), and also significantly smaller compared with the radiation alone group (160 mm 3 ) (P ⁇ 0.05).
  • the combination q value of Hb high dose is 1.24, indicating a synergistic effect of this combination (q>1.15, synergistic effect).
  • FIG. 26 shows the tumor growth inhibition effects of the hemoglobin-based oxygen carrier of the present invention followed by radiation.
  • GBM Glioblastoma multiforme
  • TMZ alkylating agent temozolomide
  • GBM cells sensitive (D54-S) and resistant (D54-R) to temozolomide are treated with various concentration (0.015 to 0.03 g/dL) of Hb alone, TMZ alone or in combination under hypoxia (1% oxygen) for 72 hours followed by cell viability assays.
  • FIG. 27A shows representative 96-well plates of D54-S and D54-R cells after different treatment conditions.
  • FIG. 27B shows a dose-dependent enhancement of TMZ-induced cytotoxicity by Hb.
  • a breast cancer cell line, MCF7 cells is labelled with CD24 and CD44 antibodies and analyzed by flow cytometry using PE and APC isotypes which are excited by 488 nm (blue laser) and 633 nm (red laser), respectively, and the respective emissions are measured by 585 nm and 660 nm Band Pass filters.
  • the flow cytometry result shows that the percentage of the commercially available MCF7 cells which highly express CD44 but not CD24 is only about 0.5% in the total population.
  • MCF7 cells are cultured in suspension on non-coated petri dishes in MammoCultTM for at least 7-9 days before spheroids formation.
  • the culture medium contains both MammoCult Basal Medium and MammoCult Proliferation Supplement for human mammospheres.
  • the culture medium is also supplemented with 0.48 ⁇ g/mL freshly dissolved hydrocortisone and 4 ⁇ g/mL heparin before use.
  • the culture medium in the petri dishes is changed every 1-2 days and the frequency can be determined from the color of the medium.
  • the morphology of the cell is observed under microscope.
  • FIG. 28 shows the cell morphology observed in the phase contrast field under a light microscope.
  • solid mammospheres are observed at about 9 th to 20 th days of growth after pouring the flow-sorted MCF7 cells onto the petri dishes.
  • the self-renewal ability is further confirmed by passing the cancer stem cells for about 9 passages and each subsequent passage after passage 0 may take about 9-14 days to develop into solid mammospheres.
  • the solid mammospheres are separated into single cells by chemical (e.g. trypsinization) or mechanical means in a sterile environment (e.g. using cell scraper to detach the cell clump from the Petri dish followed by pipetting up and down).
  • FIG. 29 shows western blots of lysed cells collected in different passages.
  • sample 1 is for unsorted cells from mammospheres and sample 2 is for CD44+/CD24 ⁇ sorted cells from mammospheres at passage 1.
  • sample 1 is for unsorted cells from mammospheres and samples 2, 3 and 4 are for CD44+/CD22 ⁇ sorted cells from mammospheres at passage 1, 2 and 3, respectively.
  • stem cell marker Oct-4 39 kDa
  • Sox-2 40 kDa
  • the expression level of these markers between unsorted and sorted cells is different.
  • the CD44+/CD24 ⁇ sorted cells have higher expression level of Oct-4 than that of unsorted cells in the same passage.
  • the self-renewal ability of the cancer stem cells becomes higher in terms of the expression level of these stem cells markers from one passage to another because of the application of cell sorting in each passage to select CD44+/CD24 ⁇ cells.
  • FIG. 30A is the result of the analysis on a control (cells incubated with diethylaminobenzaldehyde (DEAB), an inhibitor of ALDH);
  • FIG. 30B is the result of cells collected at passage 0, where it shows 1% of the cell population having the ALDH activity;
  • FIG. 30 C is the result of the cells collected at passage 3, where it shows about 8.7% of the cell population having ALDH activity; cells collected at passage 5 have about 10-13% of the population having ALDH activity ( FIG.
  • the MCF7 cells are incubated under hypoxic condition (5% CO 2 and 1.1% O 2 ) for 9-20 days before passing to the cell sorter where two filters are used: PE-A for CD24 marker while APC-A for CD44 marker.
  • Quadrant 1 where cells are positive to CD44 and negative to CD24 ( FIG. 31 ) are sorted for further analysis.
  • chemotherapeutic agent and/or the hemoglobin-based oxygen carrier of the present invention are administered to MCF7 cells isolated from mammospheres which are obtained at later passages, e.g. passages 7 and 8.
  • the drug resistance of CD44+/CD24 ⁇ to chemotherapeutic agent is shown in FIG. 32 .
  • Unsorted MCF7 cells and CD44+/CD24 ⁇ sorted cells are incubated with DMSO (as control) and 90 nM of Taxol for 16 hours and 4 days. Phase contrast images ( FIG.
  • the high resistance of the CSCs to chemotherapeutic agent is further confirmed by the results of MTT assays on single cells from two passages (P7 and P8) after the mammospheres are treated with different combination of chemical(s) for at least 24 hours before trypsinization of mammospheres.
  • the mammospheres are grown under the hypoxic conditions (5% CO2, 1.1% O2) to mimic the physiological environment of a tumor.
  • Different combination of chemical(s) used in the MTT assays include the Hb alone (0.2 g/dL), Bortezomib (“Bort”, 0.5 ⁇ M) alone, 5-fluorouracil (“5FU”, 5 ⁇ M) alone, or any combination of the above.
  • the combinational drug i.e.
  • the trypsinized cells are incubated with 0.2 g/dL of Hb for 24 hours followed by the addition of the intended chemotherapeutic agent(s) and incubated for another 24 hours.
  • the absorbance is measured by the spectrometer and the normalized value of the absorbance is given in Table 3 below. In the normalized value, “1” represents 100% of survival rate; 0.75 represents 75% of survival rate, etc.
  • the survival rate of cells from two passages is about 61-65% survival rate.
  • the survival rate of cells from two passages In the set of administering 0.5 ⁇ M of Bortezomib alone, cells from two passages have about 78%-91% survival rate.
  • the survival rate of cells from two passages In the set of administering 5 ⁇ M of 5FU alone, cells from two passages have about 72%-87% survival rate.
  • the survival rate of cells from two passages is about 38%-49%.
  • the survival rate of cells from two passages is about 52%-72%.
  • the survival rate of cells from two passages is about 60%-64%.
  • the survival rate of cells from two passages is about 33%-39%.
  • the survival rate of the cells in the combination of Bortezomib and 5FU is about 60%-64%, it is still comparatively higher than that of the cells treated with the hemoglobin-based oxygen carrier and Bortezomib. It is interesting to note that hemoglobin-based oxygen carrier alone can kill the CSCs by almost the same percentage as that of using the combination of Bortezomib and 5FU. Finally, the most effective combination of killing the CSCs in this test is the hemoglobin-based oxygen carrier plus Bortezomib and 5FU because the survival rate is only about 33%-39% which is far lower than any of the other combination as described herein.
  • the chemotherapeutic agent administered in combination with the hemoglobin-based oxygen carrier of the present invention is not limited to Bortezomib or 5FU. Any other conventional chemotherapeutic agents which have been proven to be less effective in treating cancer/tumor or any other therapy such as radiotherapy can also be used in combination with the hemoglobin-based oxygen carrier of the present invention with an improved efficacy in killing CSCs.
  • the different functions discussed herein may be performed in a different order and/or concurrently with each other. Furthermore, if desired, one or more of the above-described functions may be optional or may be combined.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Inorganic Chemistry (AREA)
  • Oncology (AREA)
  • Pathology (AREA)
  • Radiology & Medical Imaging (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US13/713,031 2012-10-12 2012-12-13 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence Abandoned US20140106004A1 (en)

Priority Applications (26)

Application Number Priority Date Filing Date Title
US13/713,031 US20140106004A1 (en) 2012-10-12 2012-12-13 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
PCT/US2013/064418 WO2014059199A1 (en) 2012-10-12 2013-10-11 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
AU2013329121A AU2013329121B2 (en) 2012-10-12 2013-10-11 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
JP2015536905A JP6113850B2 (ja) 2012-10-12 2013-10-11 がん性組織の治療およびがん性腫瘍の再発および転移を予防するための医薬組成物の調製のためのヘモグロビン系酸素運搬体の使用方法
MA37994A MA37994A2 (fr) 2012-10-12 2013-10-11 Compositions pharmaceutiques contenant un transporteur d'oxygène à base d'hémoglobine pour traitement ciblant un cancer et prévention de récidive de cancer
MX2015004512A MX367562B (es) 2012-10-12 2013-10-11 Composicion farmaceutica que contiene portador de oxigeno a base de hemoglobina para tratamiento dirigido a cancer y prevencion de recurrencia de cancer.
EP13844670.3A EP2906222A4 (en) 2012-10-12 2013-10-11 PHARMACEUTICAL COMPOSITIONS CONTAINING HEMOGLOBIN-BASED OXYGEN CARRIER FOR TREATMENT TARGETING CANCER AND PREVENTION OF CANCER RECIPIENT
CN201380053346.1A CN104717966B (zh) 2012-10-12 2013-10-11 用于癌症靶向治疗和预防癌症复发的含有基于血红蛋白的氧载体的药物组合物
SG11201502133SA SG11201502133SA (en) 2012-10-12 2013-10-11 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
SG10201608747RA SG10201608747RA (en) 2012-10-12 2013-10-11 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
AP2015008315A AP2015008315A0 (en) 2012-10-12 2013-10-11 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatmentand prevention of cancer recurrence
EA201500301A EA201500301A1 (ru) 2012-10-12 2013-10-11 Содержащая носитель кислорода на основе гемоглобина фармацевтическая композиция для адресного лечения рака и предотвращения рецидивов рака
SG10201607846PA SG10201607846PA (en) 2012-10-12 2013-10-11 Hemoglobin-Based Oxygen Carrier-Containing Pharmaceutical Composition for Cancer Targeting Treatment and Prevention of Cancer Recurrence
MYPI2015701107A MY186484A (en) 2012-10-12 2013-10-11 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
CA2884521A CA2884521C (en) 2012-10-12 2013-10-11 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
BR112015007475A BR112015007475A2 (pt) 2012-10-12 2013-10-11 composição farmacêutica contendo carreador de oxigênio à base de hemoglobina para tratamento voltado para o câncer e prevenção de recidiva do câncer e utilização do carreador de oxigênio à base de hemoglobina
KR1020157012185A KR20150065881A (ko) 2012-10-12 2013-10-11 암 표적화 치료 및 암 재발 예방을 위한 헤모글로빈 기반 산소운반체 함유 약학적 조성물
TW102136951A TW201414489A (zh) 2012-10-12 2013-10-14 用於癌症標靶治療及預防癌症復發之含有基於血紅素的氧載體之醫藥組成物
UY35082A UY35082A (es) 2012-10-12 2013-10-14 Sustancia portadora de oxigeno basada en la hemoglobina que contiene una composicion farmaceutica para el tratamiento del cancer y la la prevencion de la recurrencia del cancer
ARP130103741A AR093023A1 (es) 2012-10-12 2013-10-15 Composicion farmaceutica que contiene un portador de oxigeno a base de hemoglobina para la focalizacion del tratamiento del cancer y prevencion de la repeticion del mismo, metodo
US14/308,725 US9056098B2 (en) 2012-10-12 2014-06-19 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
PH12015500562A PH12015500562B1 (en) 2012-10-12 2015-03-16 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
IL237763A IL237763A (en) 2012-10-12 2015-03-16 Hemoglobin-based Oxygen-Based Pharmaceutical for Cancer-Targeted Treatment and Cancer Prevention
ZA2015/01949A ZA201501949B (en) 2012-10-12 2015-03-20 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
CL2015000897A CL2015000897A1 (es) 2012-10-12 2015-04-09 Composición farmacéutica que contiene un portador de oxígeno basado en hemoglobina para el tratamiento de focalización de cáncer y prevención de recurrencia de cáncer.
HK15107105.0A HK1206281A1 (zh) 2012-10-12 2015-07-24 用於癌症靶向治療和預防癌症復發的含有基於血紅蛋白的氧載體的藥物組合物

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261712853P 2012-10-12 2012-10-12
US13/713,031 US20140106004A1 (en) 2012-10-12 2012-12-13 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/308,725 Continuation US9056098B2 (en) 2012-10-12 2014-06-19 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence

Publications (1)

Publication Number Publication Date
US20140106004A1 true US20140106004A1 (en) 2014-04-17

Family

ID=50475529

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/713,031 Abandoned US20140106004A1 (en) 2012-10-12 2012-12-13 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
US14/308,725 Active US9056098B2 (en) 2012-10-12 2014-06-19 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/308,725 Active US9056098B2 (en) 2012-10-12 2014-06-19 Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence

Country Status (22)

Country Link
US (2) US20140106004A1 (zh)
EP (1) EP2906222A4 (zh)
JP (1) JP6113850B2 (zh)
KR (1) KR20150065881A (zh)
CN (1) CN104717966B (zh)
AP (1) AP2015008315A0 (zh)
AR (1) AR093023A1 (zh)
AU (1) AU2013329121B2 (zh)
BR (1) BR112015007475A2 (zh)
CA (1) CA2884521C (zh)
CL (1) CL2015000897A1 (zh)
EA (1) EA201500301A1 (zh)
HK (1) HK1206281A1 (zh)
IL (1) IL237763A (zh)
MA (1) MA37994A2 (zh)
MX (1) MX367562B (zh)
PH (1) PH12015500562B1 (zh)
SG (3) SG11201502133SA (zh)
TW (1) TW201414489A (zh)
UY (1) UY35082A (zh)
WO (1) WO2014059199A1 (zh)
ZA (1) ZA201501949B (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017124037A1 (en) * 2016-01-15 2017-07-20 The Children's Medical Center Corporation Therapeutic use of mitochondria and combined mitochondrial agents
US9814759B2 (en) * 2014-07-02 2017-11-14 Cheer Global Ltd. Pharmaceutical composition comprising recombinant hemoglobin protein or subunit-based therapeutic agent for cancer targeting treatment
CN111558032A (zh) * 2020-05-19 2020-08-21 中国科学院宁波材料技术与工程研究所 一种蛋白纳米药物及其制备方法与应用
US11491480B2 (en) 2014-06-13 2022-11-08 Children's Medical Center Corporation Products and methods to isolate mitochondria
US11903975B2 (en) 2015-11-30 2024-02-20 Flagship Pioneering Innovations V, Inc. Methods and compositions relating to chondrisomes from blood products

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112015028340A2 (pt) * 2013-05-13 2017-09-19 Vision Global Holdings Ltd Composições farmacêuticas envolvendo agente terapêutico baseado em hemoglobina modificada para direcionamento e tratamento de câncer e diagnóstico por imagem e um método para o preparo do agente terapêutico
CA3028589A1 (en) * 2016-06-21 2017-12-28 Therapure Biopharma Inc. Hemoglobin-targeted drug delivery for the treatment of cancer
CA3060509A1 (en) * 2017-04-21 2018-10-25 Federica Sotgia Targeting hypoxic cancer stem cells (cscs) with doxycycline: implications for improving anti-angiogenic therapy
EP3612187B1 (en) 2017-04-21 2022-04-13 Lunella Biotech, Inc. Vitamin c and doxycycline: a synthetic lethal combination therapy for eradicating cancer stem cells (cscs)
MX2019013735A (es) 2017-05-19 2020-01-15 Lunella Biotech Inc Antimitoscinas: inhibidores especificos de la biogenesis mitocondrial para erradicar celulas madre cancerosas.
CU20190106A7 (es) 2017-06-26 2020-11-30 Lunella Biotech Inc Mitocetoscinas: agentes terapéuticos basados en mitocondrias que fijan como objetivo el metabolismo de cetonas en células cancerosas
WO2019124423A1 (ja) * 2017-12-19 2019-06-27 国立大学法人 岡山大学 がんの進行抑制、治療、予防及び/又は再発予防剤
CN114344263B (zh) * 2022-02-21 2023-08-01 杭州普略生物科技有限公司 一种用于靶向巨噬细胞增强肿瘤治疗效果的纳米蛋白胶束及其制备方法和应用
KR20230141306A (ko) 2022-03-31 2023-10-10 충남대학교산학협력단 저산소증 완화능을 갖는 페길화 헤모글로빈 나노클러스터를 포함하는 항암 화학-광역학 치료용 조성물

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5478806A (en) * 1989-11-22 1995-12-26 Enzon, Inc. Enhancement of antitumor therapy with hemoglobin-based conjugates
CA2236344A1 (en) * 1998-04-30 1999-10-30 Hemosol Inc. Hemoglobin-haptoglobin complexes
US20050164915A1 (en) * 2002-04-01 2005-07-28 Sangart, Inc. Compositions for oxygen transport comprising a high oxygen affinity modified hemoglobin
CN1741821B (zh) * 2002-10-29 2011-08-10 恩根尼公司 用于癌症治疗的组合物
WO2010085654A1 (en) * 2009-01-23 2010-07-29 Cancer Research Technology Limited Hedgehog pathway inhibitors
DK2440239T3 (en) * 2009-06-09 2017-10-16 Prolong Pharmaceuticals Llc HEMOGLOBIN FORMATIONS
EP2451459B1 (en) * 2009-07-07 2017-11-22 Normoxys, Inc. Method of reducing multi-drug resistance using inositol tripyrophosphate
US8808748B2 (en) * 2010-04-20 2014-08-19 Vindico NanoBio Technology Inc. Biodegradable nanoparticles as novel hemoglobin-based oxygen carriers and methods of using the same
US7989593B1 (en) * 2010-05-27 2011-08-02 Bing Lou Wong Method for the preparation of a high-temperature stable oxygen-carrier-containing pharmaceutical composition and the use thereof
US20110319332A1 (en) * 2010-06-23 2011-12-29 Bing Lou Wong Treatment methods using a heat stable oxygen carrier-containing pharmaceutical composition
US8048856B1 (en) * 2010-06-23 2011-11-01 Billion King, Ltd. Treatment methods using a heat stable oxygen carrier-containing pharmaceutical composition
US7932356B1 (en) * 2010-06-23 2011-04-26 Bing Lou Wong Method for the preparation of a heat stable oxygen carrier-containing pharmaceutical composition

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11491480B2 (en) 2014-06-13 2022-11-08 Children's Medical Center Corporation Products and methods to isolate mitochondria
US9814759B2 (en) * 2014-07-02 2017-11-14 Cheer Global Ltd. Pharmaceutical composition comprising recombinant hemoglobin protein or subunit-based therapeutic agent for cancer targeting treatment
TWI633118B (zh) * 2014-07-02 2018-08-21 環喜有限公司 用於癌症標靶治療之包含基於重組血紅素蛋白或次單元之治療劑的醫藥組成物
US11903975B2 (en) 2015-11-30 2024-02-20 Flagship Pioneering Innovations V, Inc. Methods and compositions relating to chondrisomes from blood products
US11903974B2 (en) 2015-11-30 2024-02-20 Flagship Pioneering Innovations V, Inc. Methods and compositions relating to chondrisomes from cultured cells
WO2017124037A1 (en) * 2016-01-15 2017-07-20 The Children's Medical Center Corporation Therapeutic use of mitochondria and combined mitochondrial agents
US10370458B2 (en) 2016-01-15 2019-08-06 Children's Medical Center Corporation and Beth Israel Therapeutic use of mitochondria and combined mitochondrial agent
CN111558032A (zh) * 2020-05-19 2020-08-21 中国科学院宁波材料技术与工程研究所 一种蛋白纳米药物及其制备方法与应用

Also Published As

Publication number Publication date
MX367562B (es) 2019-08-27
SG10201607846PA (en) 2016-11-29
MX2015004512A (es) 2015-12-01
MA37994A2 (fr) 2016-06-30
AP2015008315A0 (en) 2015-03-31
BR112015007475A2 (pt) 2017-08-08
JP2015534946A (ja) 2015-12-07
PH12015500562A1 (en) 2015-05-04
IL237763A (en) 2017-07-31
US20140303085A1 (en) 2014-10-09
EA201500301A1 (ru) 2015-08-31
AR093023A1 (es) 2015-05-13
WO2014059199A8 (en) 2014-05-30
CA2884521A1 (en) 2014-04-17
SG11201502133SA (en) 2015-04-29
PH12015500562B1 (en) 2015-05-04
WO2014059199A1 (en) 2014-04-17
UY35082A (es) 2014-07-31
KR20150065881A (ko) 2015-06-15
EP2906222A1 (en) 2015-08-19
AU2013329121B2 (en) 2017-05-25
CA2884521C (en) 2018-05-01
CN104717966B (zh) 2018-08-24
JP6113850B2 (ja) 2017-04-12
EP2906222A4 (en) 2016-06-22
TW201414489A (zh) 2014-04-16
ZA201501949B (en) 2016-01-27
AU2013329121A1 (en) 2015-04-09
US9056098B2 (en) 2015-06-16
SG10201608747RA (en) 2016-12-29
HK1206281A1 (zh) 2016-01-08
CN104717966A (zh) 2015-06-17
CL2015000897A1 (es) 2015-08-21

Similar Documents

Publication Publication Date Title
US9056098B2 (en) Hemoglobin-based oxygen carrier-containing pharmaceutical composition for cancer targeting treatment and prevention of cancer recurrence
Luo et al. Tumor-targeted hybrid protein oxygen carrier to simultaneously enhance hypoxia-dampened chemotherapy and photodynamic therapy at a single dose
Domanska et al. A review on CXCR4/CXCL12 axis in oncology: no place to hide
Alghamri et al. Systemic delivery of an adjuvant CXCR4–CXCL12 signaling inhibitor encapsulated in synthetic protein nanoparticles for glioma immunotherapy
AU2014265659B2 (en) Pharmaceutical composition comprising modified hemoglobin- based therapeutic agent for cancer targeting treatment and diagnostic imaging
Jin et al. Combination chemotherapy of doxorubicin and paclitaxel for hepatocellular carcinoma in vitro and in vivo
WO2018021200A1 (ja) Runx阻害剤
US20180252721A1 (en) Selection of patients for combination therapy
Lu et al. S-Nitrosocaptopril prevents cancer metastasis in vivo by creating the hostile bloodstream microenvironment against circulating tumor cells
Zhang et al. Polycysteine as a new type of radio-protector ameliorated tissue injury through inhibiting ferroptosis in mice
JP6527825B2 (ja) 癌を処置するための環形動物のヘモグロビンの使用
Luke et al. ASP9853, an inhibitor of inducible nitric oxide synthase dimerization, in combination with docetaxel: preclinical investigation and a Phase I study in advanced solid tumors
JP2018035112A (ja) 抗癌剤の抗腫瘍効果の増強剤、癌治療剤、及び癌治療用医薬組成物。
US20200155594A1 (en) Hornerin: a novel non-vegf mediated angiogenic protein expressed in both human and mouse angiogenic endothelial cells and human pancreatic cancer cells
Shenoy et al. A closer look at the role of iron in glioblastoma
CA2844032A1 (en) Method for treating cancer by combined use of drugs
US10550130B2 (en) Benzo-thiazolo-imidazole compounds and uses thereof
US20220409581A1 (en) Methods and compositions for treatment of solid tumors using f16 isoindole small molecules
US20220143028A1 (en) Inhibitors of atypical protein kinase c and their use in treating hedgehog pathway-dependent cancers
US20180230466A1 (en) Methods for treating tumors
AU2015346430B2 (en) Apoaequorin-containing compositions and methods of using same to treat neuronal inflammation
Yang et al. DTTZ suppresses ferroptosis and reverses mitochondrial dysfunction in normal tissues affected by chemotherapy
AU2019266231A1 (en) Selection of patients for combination therapy
KR20190123147A (ko) 세포에서의 자가포식작용 억제용 조성물, 및 이를 포함하는 종양성 질환의 예방 또는 치료용, 또는 항암제 내성 억제용 약학적 조성물
Kruizinga Role of VEGFA, CXCR4 and VHL mutation in tumour behaviour

Legal Events

Date Code Title Description
AS Assignment

Owner name: VISION GLOBAL HOLDINGS LTD., HONG KONG

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WONG, BING LOU;WAI, NORMAN FUNG MAN;KWOK, SUI YI;AND OTHERS;REEL/FRAME:029936/0850

Effective date: 20130306

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION